Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.617
Filter
Add more filters

Publication year range
1.
Cell ; 187(4): 897-913.e18, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38280374

ABSTRACT

Canonically, the complement system is known for its rapid response to remove microbes in the bloodstream. However, relatively little is known about a functioning complement system on intestinal mucosal surfaces. Herein, we report the local synthesis of complement component 3 (C3) in the gut, primarily by stromal cells. C3 is expressed upon commensal colonization and is regulated by the composition of the microbiota in healthy humans and mice, leading to an individual host's specific luminal C3 levels. The absence of membrane attack complex (MAC) components in the gut ensures that C3 deposition does not result in the lysis of commensals. Pathogen infection triggers the immune system to recruit neutrophils to the infection site for pathogen clearance. Basal C3 levels directly correlate with protection against enteric infection. Our study reveals the gut complement system as an innate immune mechanism acting as a vigilant sentinel that combats pathogens and spares commensals.


Subject(s)
Complement C3 , Intestinal Mucosa , Microbiota , Animals , Humans , Mice , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Neutrophils , Complement C3/metabolism , Stromal Cells/metabolism
2.
Cell ; 185(22): 4190-4205.e25, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36243004

ABSTRACT

Neuroepithelial crosstalk is critical for gut physiology. However, the mechanisms by which sensory neurons communicate with epithelial cells to mediate gut barrier protection at homeostasis and during inflammation are not well understood. Here, we find that Nav1.8+CGRP+ nociceptor neurons are juxtaposed with and signal to intestinal goblet cells to drive mucus secretion and gut protection. Nociceptor ablation led to decreased mucus thickness and dysbiosis, while chemogenetic nociceptor activation or capsaicin treatment induced mucus growth. Mouse and human goblet cells expressed Ramp1, receptor for the neuropeptide CGRP. Nociceptors signal via the CGRP-Ramp1 pathway to induce rapid goblet cell emptying and mucus secretion. Notably, commensal microbes activated nociceptors to control homeostatic CGRP release. In the absence of nociceptors or epithelial Ramp1, mice showed increased epithelial stress and susceptibility to colitis. Conversely, CGRP administration protected nociceptor-ablated mice against colitis. Our findings demonstrate a neuron-goblet cell axis that orchestrates gut mucosal barrier protection.


Subject(s)
Colitis , Goblet Cells , Mice , Humans , Animals , Goblet Cells/metabolism , Nociceptors/metabolism , Calcitonin Gene-Related Peptide/metabolism , Colitis/metabolism , Mucus/metabolism , Receptor Activity-Modifying Protein 1/metabolism
3.
Cell ; 181(6): 1276-1290.e13, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32402238

ABSTRACT

At the species level, immunity depends on the selection and transmission of protective components of the immune system. A microbe-induced population of RORγ-expressing regulatory T cells (Tregs) is essential in controlling gut inflammation. We uncovered a non-genetic, non-epigenetic, non-microbial mode of transmission of their homeostatic setpoint. RORγ+ Treg proportions varied between inbred mouse strains, a trait transmitted by the mother during a tight age window after birth but stable for life, resistant to many microbial or cellular perturbations, then further transferred by females for multiple generations. RORγ+ Treg proportions negatively correlated with IgA production and coating of gut commensals, traits also subject to maternal transmission, in an immunoglobulin- and RORγ+ Treg-dependent manner. We propose a model based on a double-negative feedback loop, vertically transmitted via the entero-mammary axis. This immunologic mode of multi-generational transmission may provide adaptability and modulate the genetic tuning of gut immune responses and inflammatory disease susceptibility.


Subject(s)
Digestive System/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Disease Susceptibility/immunology , Female , Gastrointestinal Microbiome/immunology , Homeostasis/immunology , Immunoglobulin A/immunology , Inflammation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred NOD , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
4.
Cell ; 180(4): 655-665.e18, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32004463

ABSTRACT

Human endocannabinoid systems modulate multiple physiological processes mainly through the activation of cannabinoid receptors CB1 and CB2. Their high sequence similarity, low agonist selectivity, and lack of activation and G protein-coupling knowledge have hindered the development of therapeutic applications. Importantly, missing structural information has significantly held back the development of promising CB2-selective agonist drugs for treating inflammatory and neuropathic pain without the psychoactivity of CB1. Here, we report the cryoelectron microscopy structures of synthetic cannabinoid-bound CB2 and CB1 in complex with Gi, as well as agonist-bound CB2 crystal structure. Of important scientific and therapeutic benefit, our results reveal a diverse activation and signaling mechanism, the structural basis of CB2-selective agonists design, and the unexpected interaction of cholesterol with CB1, suggestive of its endogenous allosteric modulating role.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Receptor, Cannabinoid, CB1/chemistry , Receptor, Cannabinoid, CB2/chemistry , Signal Transduction , Allosteric Regulation , Allosteric Site , Animals , CHO Cells , Cannabinoid Receptor Agonists/chemistry , Cannabinoids/chemistry , Cannabinoids/pharmacology , Cell Line, Tumor , Cholesterol/chemistry , Cholesterol/pharmacology , Cricetinae , Cricetulus , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Molecular Dynamics Simulation , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Sf9 Cells , Spodoptera
5.
Cell ; 176(3): 459-467.e13, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30639103

ABSTRACT

The cannabinoid receptor CB2 is predominately expressed in the immune system, and selective modulation of CB2 without the psychoactivity of CB1 has therapeutic potential in inflammatory, fibrotic, and neurodegenerative diseases. Here, we report the crystal structure of human CB2 in complex with a rationally designed antagonist, AM10257, at 2.8 Å resolution. The CB2-AM10257 structure reveals a distinctly different binding pose compared with CB1. However, the extracellular portion of the antagonist-bound CB2 shares a high degree of conformational similarity with the agonist-bound CB1, which led to the discovery of AM10257's unexpected opposing functional profile of CB2 antagonism versus CB1 agonism. Further structural analysis using mutagenesis studies and molecular docking revealed the molecular basis of their function and selectivity for CB2 and CB1. Additional analyses of our designed antagonist and agonist pairs provide important insight into the activation mechanism of CB2. The present findings should facilitate rational drug design toward precise modulation of the endocannabinoid system.


Subject(s)
Receptor, Cannabinoid, CB2/metabolism , Receptor, Cannabinoid, CB2/ultrastructure , Animals , Cannabinoid Receptor Antagonists/pharmacology , Cannabinoids/pharmacology , Drug Design , Endocannabinoids , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/chemistry , Receptors, Cannabinoid/chemistry , Receptors, Cannabinoid/metabolism , Receptors, Cannabinoid/ultrastructure , Receptors, G-Protein-Coupled/metabolism , Sf9 Cells , Structure-Activity Relationship
6.
Cell ; 170(6): 1247-1257.e12, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28844695

ABSTRACT

The respiratory megacomplex represents the highest-order assembly of respiratory chain complexes, and it allows mitochondria to respond to energy-requiring conditions. To understand its architecture, we examined the human respiratory chain megacomplex-I2III2IV2 (MCI2III2IV2) with 140 subunits and a subset of associated cofactors using cryo-electron microscopy. The MCI2III2IV2 forms a circular structure with the dimeric CIII located in the center, where it is surrounded by two copies each of CI and CIV. Two cytochrome c (Cyt.c) molecules are positioned to accept electrons on the surface of the c1 state CIII dimer. Analyses indicate that CII could insert into the gaps between CI and CIV to form a closed ring, which we termed the electron transport chain supercomplex. The structure not only reveals the precise assignment of individual subunits of human CI and CIII, but also enables future in-depth analysis of the electron transport chain as a whole.


Subject(s)
Electron Transport Chain Complex Proteins/chemistry , Multienzyme Complexes/chemistry , Cryoelectron Microscopy , Electron Transport Chain Complex Proteins/isolation & purification , Electron Transport Chain Complex Proteins/metabolism , Electron Transport Complex I/chemistry , Electron Transport Complex I/isolation & purification , Electron Transport Complex I/metabolism , Electron Transport Complex II/chemistry , Electron Transport Complex II/isolation & purification , Electron Transport Complex II/metabolism , Humans , Mitochondria/chemistry , Mitochondria/metabolism , Models, Molecular , Multienzyme Complexes/isolation & purification , Multienzyme Complexes/metabolism
7.
Cell ; 167(6): 1598-1609.e10, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27912063

ABSTRACT

The mammalian respiratory chain complexes assemble into supercomplexes (SCs) and reside in the inner mitochondrial membrane to transfer electrons and establish the proton gradient for complex V to synthesize ATP. The precise arrangement of SCs is largely unknown. Here, we report a 4.0-Å cryo-electron microscopy (cryo-EM) structure of the major SC in porcine heart, the 1.7-MDa SCI1III2IV1. The complex III (CIII) dimer and complex IV (CIV) bind at the same side of the L-shaped complex I (CI). Several accessory or supernumerary subunits of CI, such as NDUFA11, NDUFB4, NDUFB8, and NDUFB9, directly contribute to the oligomerization of CI, CIII, and CIV. COX7C and COX7A of CIV attach CIV to the concave surface formed by CIII and the distal end of membrane arm of CI. The structure suggests a possible mechanism by which electrons are transferred from NADH to cytochrome c and provides a platform for future functional dissection of respiration.


Subject(s)
Electron Transport , Mitochondria, Heart/chemistry , Mitochondrial Membranes/chemistry , Animals , Cryoelectron Microscopy , Models, Molecular , Multienzyme Complexes/chemistry , Proton Pumps/chemistry , Sus scrofa
8.
Cell ; 161(6): 1252-65, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26046436

ABSTRACT

Small-molecule probes can illuminate biological processes and aid in the assessment of emerging therapeutic targets by perturbing biological systems in a manner distinct from other experimental approaches. Despite the tremendous promise of chemical tools for investigating biology and disease, small-molecule probes were unavailable for most targets and pathways as recently as a decade ago. In 2005, the NIH launched the decade-long Molecular Libraries Program with the intent of innovating in and broadening access to small-molecule science. This Perspective describes how novel small-molecule probes identified through the program are enabling the exploration of biological pathways and therapeutic hypotheses not otherwise testable. These experiences illustrate how small-molecule probes can help bridge the chasm between biological research and the development of medicines but also highlight the need to innovate the science of therapeutic discovery.


Subject(s)
Drug Discovery , Small Molecule Libraries , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , National Institutes of Health (U.S.) , United States
9.
Nature ; 619(7971): 837-843, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37380774

ABSTRACT

The human gut microbiome constantly converts natural products derived from the host and diet into numerous bioactive metabolites1-3. Dietary fats are essential micronutrients that undergo lipolysis to release free fatty acids (FAs) for absorption in the small intestine4. Gut commensal bacteria modify some unsaturated FAs-for example, linoleic acid (LA)-into various intestinal FA isomers that regulate host metabolism and have anticarcinogenic properties5. However, little is known about how this diet-microorganism FA isomerization network affects the mucosal immune system of the host. Here we report that both dietary factors and microbial factors influence the level of gut LA isomers (conjugated LAs (CLAs)) and that CLAs in turn modulate a distinct population of CD4+ intraepithelial lymphocytes (IELs) that express CD8αα in the small intestine. Genetic abolition of FA isomerization pathways in individual gut symbionts significantly decreases the number of CD4+CD8αα+ IELs in gnotobiotic mice. Restoration of CLAs increases CD4+CD8αα+ IEL levels in the presence of the transcription factor hepatocyte nuclear factor 4γ (HNF4γ). Mechanistically, HNF4γ facilitates CD4+CD8αα+ IEL development by modulating interleukin-18 signalling. In mice, specific deletion of HNF4γ in T cells leads to early mortality from infection by intestinal pathogens. Our data reveal a new role for bacterial FA metabolic pathways in the control of host intraepithelial immunological homeostasis by modulating the relative number of CD4+ T cells that were CD4+CD8αα+.


Subject(s)
Fatty Acids , Gastrointestinal Microbiome , Intraepithelial Lymphocytes , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Isomerism , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Lipolysis , Linoleic Acid/metabolism , Immunity, Mucosal
10.
Nature ; 617(7960): 377-385, 2023 05.
Article in English | MEDLINE | ID: mdl-37138075

ABSTRACT

The gut microbiota is a crucial regulator of anti-tumour immunity during immune checkpoint inhibitor therapy. Several bacteria that promote an anti-tumour response to immune checkpoint inhibitors have been identified in mice1-6. Moreover, transplantation of faecal specimens from responders can improve the efficacy of anti-PD-1 therapy in patients with melanoma7,8. However, the increased efficacy from faecal transplants is variable and how gut bacteria promote anti-tumour immunity remains unclear. Here we show that the gut microbiome downregulates PD-L2 expression and its binding partner repulsive guidance molecule b (RGMb) to promote anti-tumour immunity and identify bacterial species that mediate this effect. PD-L1 and PD-L2 share PD-1 as a binding partner, but PD-L2 can also bind RGMb. We demonstrate that blockade of PD-L2-RGMb interactions can overcome microbiome-dependent resistance to PD-1 pathway inhibitors. Antibody-mediated blockade of the PD-L2-RGMb pathway or conditional deletion of RGMb in T cells combined with an anti-PD-1 or anti-PD-L1 antibody promotes anti-tumour responses in multiple mouse tumour models that do not respond to anti-PD-1 or anti-PD-L1 alone (germ-free mice, antibiotic-treated mice and even mice colonized with stool samples from a patient who did not respond to treatment). These studies identify downregulation of the PD-L2-RGMb pathway as a specific mechanism by which the gut microbiota can promote responses to PD-1 checkpoint blockade. The results also define a potentially effective immunological strategy for treating patients who do not respond to PD-1 cancer immunotherapy.


Subject(s)
Drug Resistance, Neoplasm , Immunotherapy , Melanoma , Microbiota , Animals , Humans , Mice , Cell Adhesion Molecules, Neuronal , Disease Models, Animal , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Fecal Microbiota Transplantation , Germ-Free Life , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/immunology , Melanoma/microbiology , Melanoma/therapy , Protein Binding/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
11.
Nature ; 622(7984): 850-862, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37794185

ABSTRACT

Immune checkpoint blockade is effective for some patients with cancer, but most are refractory to current immunotherapies and new approaches are needed to overcome resistance1,2. The protein tyrosine phosphatases PTPN2 and PTPN1 are central regulators of inflammation, and their genetic deletion in either tumour cells or immune cells promotes anti-tumour immunity3-6. However, phosphatases are challenging drug targets; in particular, the active site has been considered undruggable. Here we present the discovery and characterization of ABBV-CLS-484 (AC484), a first-in-class, orally bioavailable, potent PTPN2 and PTPN1 active-site inhibitor. AC484 treatment in vitro amplifies the response to interferon and promotes the activation and function of several immune cell subsets. In mouse models of cancer resistant to PD-1 blockade, AC484 monotherapy generates potent anti-tumour immunity. We show that AC484 inflames the tumour microenvironment and promotes natural killer cell and CD8+ T cell function by enhancing JAK-STAT signalling and reducing T cell dysfunction. Inhibitors of PTPN2 and PTPN1 offer a promising new strategy for cancer immunotherapy and are currently being evaluated in patients with advanced solid tumours (ClinicalTrials.gov identifier NCT04777994 ). More broadly, our study shows that small-molecule inhibitors of key intracellular immune regulators can achieve efficacy comparable to or exceeding that of antibody-based immune checkpoint blockade in preclinical models. Finally, to our knowledge, AC484 represents the first active-site phosphatase inhibitor to enter clinical evaluation for cancer immunotherapy and may pave the way for additional therapeutics that target this important class of enzymes.


Subject(s)
Immunotherapy , Neoplasms , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Animals , Humans , Mice , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Drug Resistance, Neoplasm , Immune Checkpoint Inhibitors , Immunotherapy/methods , Interferons/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 2/antagonists & inhibitors , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
12.
Immunity ; 50(3): 692-706.e7, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30824326

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a severe form of lung fibrosis with a high mortality rate. However, the etiology of IPF remains unknown. Here, we report that alterations in lung microbiota critically promote pulmonary fibrosis pathogenesis. We found that lung microbiota was dysregulated, and the dysregulated microbiota in turn induced production of interleukin-17B (IL-17B) during bleomycin-induced mouse lung fibrosis. Either lung-microbiota depletion or IL-17B deficiency ameliorated the disease progression. IL-17B cooperated with tumor necrosis factor-α to induce expression of neutrophil-recruiting genes and T helper 17 (Th17)-cell-promoting genes. Three pulmonary commensal microbes, which belong to the genera Bacteroides and Prevotella, were identified to promote fibrotic pathogenesis through IL-17R signaling. We further defined that the outer membrane vesicles (OMVs) that were derived from the identified commensal microbes induced IL-17B production through Toll-like receptor-Myd88 adaptor signaling. Together our data demonstrate that specific pulmonary symbiotic commensals can promote lung fibrosis by regulating a profibrotic inflammatory cytokine network.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/microbiology , Interleukin-17/metabolism , Lung/metabolism , Lung/microbiology , Microbiota/physiology , Animals , Bacteroides/metabolism , Cytokines/metabolism , Disease Models, Animal , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Neutrophils/metabolism , Prevotella/metabolism , Signal Transduction/physiology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/metabolism
13.
Proc Natl Acad Sci U S A ; 121(6): e2309243121, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38289950

ABSTRACT

Staphylococcus aureus skin colonization and eosinophil infiltration are associated with many inflammatory skin disorders, including atopic dermatitis, bullous pemphigoid, Netherton's syndrome, and prurigo nodularis. However, whether there is a relationship between S. aureus and eosinophils and how this interaction influences skin inflammation is largely undefined. We show in a preclinical mouse model that S. aureus epicutaneous exposure induced eosinophil-recruiting chemokines and eosinophil infiltration into the skin. Remarkably, we found that eosinophils had a comparable contribution to the skin inflammation as T cells, in a manner dependent on eosinophil-derived IL-17A and IL-17F production. Importantly, IL-36R signaling induced CCL7-mediated eosinophil recruitment to the inflamed skin. Last, S. aureus proteases induced IL-36α expression in keratinocytes, which promoted infiltration of IL-17-producing eosinophils. Collectively, we uncovered a mechanism for S. aureus proteases to trigger eosinophil-mediated skin inflammation, which has implications in the pathogenesis of inflammatory skin diseases.


Subject(s)
Dermatitis, Atopic , Eosinophilia , Staphylococcal Infections , Animals , Mice , Eosinophils/metabolism , Staphylococcus aureus/metabolism , Peptide Hydrolases/metabolism , Skin/metabolism , Dermatitis, Atopic/metabolism , Staphylococcal Infections/metabolism , Cellulitis/metabolism , Cellulitis/pathology , Inflammation/metabolism
14.
EMBO Rep ; 25(7): 2914-2949, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38783164

ABSTRACT

Neutrophil extracellular traps (NETs) are a key antimicrobial feature of cellular innate immunity mediated by polymorphonuclear neutrophils (PMNs). NETs counteract microbes but are also linked to inflammation in atherosclerosis, arthritis, or psoriasis by unknown mechanisms. Here, we report that NET-associated RNA (naRNA) stimulates further NET formation in naive PMNs via a unique TLR8-NLRP3 inflammasome-dependent pathway. Keratinocytes respond to naRNA with expression of psoriasis-related genes (e.g., IL17, IL36) via atypical NOD2-RIPK signaling. In vivo, naRNA drives temporary skin inflammation, which is drastically ameliorated by genetic ablation of RNA sensing. Unexpectedly, the naRNA-LL37 'composite damage-associated molecular pattern (DAMP)' is pre-stored in resting neutrophil granules, defining sterile NETs as inflammatory webs that amplify neutrophil activation. However, the activity of the naRNA-LL37 DAMP is transient and hence supposedly self-limiting under physiological conditions. Collectively, upon dysregulated NET release like in psoriasis, naRNA sensing may represent both a potential cause of disease and a new intervention target.


Subject(s)
Alarmins , Cathelicidins , Extracellular Traps , Inflammation , Neutrophils , Extracellular Traps/metabolism , Neutrophils/metabolism , Neutrophils/immunology , Inflammation/metabolism , Inflammation/genetics , Animals , Humans , Mice , Alarmins/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Keratinocytes/metabolism , RNA/genetics , RNA/metabolism , Psoriasis/genetics , Psoriasis/metabolism , Psoriasis/pathology , Signal Transduction , Neutrophil Activation/genetics , Immunity, Innate/genetics
15.
Nature ; 585(7823): 135-140, 2020 09.
Article in English | MEDLINE | ID: mdl-32610344

ABSTRACT

Chemokines and their receptors mediate cell migration, which influences multiple fundamental biological processes and disease conditions such as inflammation and cancer1. Although ample effort has been invested into the structural investigation of the chemokine receptors and receptor-chemokine recognition2-4, less is known about endogenous chemokine-induced receptor activation and G-protein coupling. Here we present the cryo-electron microscopy structures of interleukin-8 (IL-8, also known as CXCL8)-activated human CXC chemokine receptor 2 (CXCR2) in complex with Gi protein, along with a crystal structure of CXCR2 bound to a designed allosteric antagonist. Our results reveal a unique shallow mode of binding between CXCL8 and CXCR2, and also show the interactions between CXCR2 and Gi protein. Further structural analysis of the inactive and active states of CXCR2 reveals a distinct activation process and the competitive small-molecule antagonism of chemokine receptors. In addition, our results provide insights into how a G-protein-coupled receptor is activated by an endogenous protein molecule, which will assist in the rational development of therapeutics that target the chemokine system for better pharmacological profiles.


Subject(s)
Models, Molecular , Receptors, Interleukin-8B/chemistry , Receptors, Interleukin-8B/metabolism , Signal Transduction , Allosteric Regulation , Allosteric Site , Chemokines/classification , Chemokines/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Interleukin-8/metabolism , Protein Binding , Structure-Activity Relationship , Substrate Specificity
16.
Nature ; 577(7791): 543-548, 2020 01.
Article in English | MEDLINE | ID: mdl-31915378

ABSTRACT

Although maternal antibodies protect newborn babies from infection1,2, little is known about how protective antibodies are induced without prior pathogen exposure. Here we show that neonatal mice that lack the capacity to produce IgG are protected from infection with the enteric pathogen enterotoxigenic Escherichia coli by maternal natural IgG antibodies against the maternal microbiota when antibodies are delivered either across the placenta or through breast milk. By challenging pups that were fostered by either maternal antibody-sufficient or antibody-deficient dams, we found that IgG derived from breast milk was crucial for protection against mucosal disease induced by enterotoxigenic E. coli. IgG also provides protection against systemic infection by E. coli. Pups used the neonatal Fc receptor to transfer IgG from milk into serum. The maternal commensal microbiota can induce antibodies that recognize antigens expressed by enterotoxigenic E. coli and other Enterobacteriaceae species. Induction of maternal antibodies against a commensal Pantoea species confers protection against enterotoxigenic E. coli in pups. This role of the microbiota in eliciting protective antibodies to a specific neonatal pathogen represents an important host defence mechanism against infection in neonates.


Subject(s)
Antibodies/immunology , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Infections/immunology , Escherichia coli Infections/prevention & control , Immunity, Maternally-Acquired/immunology , Infant, Newborn/immunology , Microbiota/immunology , Milk, Human/immunology , Animals , Antibodies/blood , Antibodies/metabolism , Breast Feeding , Cross Reactions/immunology , Escherichia coli Infections/microbiology , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Male , Mice , Mothers , Pantoea/immunology , Receptors, Fc/immunology , Receptors, Fc/metabolism , Symbiosis/immunology
17.
Nature ; 577(7790): 410-415, 2020 01.
Article in English | MEDLINE | ID: mdl-31875848

ABSTRACT

The metabolic pathways encoded by the human gut microbiome constantly interact with host gene products through numerous bioactive molecules1. Primary bile acids (BAs) are synthesized within hepatocytes and released into the duodenum to facilitate absorption of lipids or fat-soluble vitamins2. Some BAs (approximately 5%) escape into the colon, where gut commensal bacteria convert them into various intestinal BAs2 that are important hormones that regulate host cholesterol metabolism and energy balance via several nuclear receptors and/or G-protein-coupled receptors3,4. These receptors have pivotal roles in shaping host innate immune responses1,5. However, the effect of this host-microorganism biliary network on the adaptive immune system remains poorly characterized. Here we report that both dietary and microbial factors influence the composition of the gut BA pool and modulate an important population of colonic FOXP3+ regulatory T (Treg) cells expressing the transcription factor RORγ. Genetic abolition of BA metabolic pathways in individual gut symbionts significantly decreases this Treg cell population. Restoration of the intestinal BA pool increases colonic RORγ+ Treg cell counts and ameliorates host susceptibility to inflammatory colitis via BA nuclear receptors. Thus, a pan-genomic biliary network interaction between hosts and their bacterial symbionts can control host immunological homeostasis via the resulting metabolites.


Subject(s)
Bile Acids and Salts/metabolism , Gastrointestinal Microbiome , Homeostasis , Intestines/immunology , Intestines/microbiology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Bile Acids and Salts/chemistry , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
18.
PLoS Genet ; 19(1): e1010551, 2023 01.
Article in English | MEDLINE | ID: mdl-36656838

ABSTRACT

Human activities have precipitated a rise in the levels of introgressive gene flow among animals. The investigation of conspecific populations at different time points may shed light on the magnitude of human-mediated introgression. We used the red junglefowl Gallus gallus, the wild ancestral form of the chicken, as our study system. As wild junglefowl and domestic chickens readily admix, conservationists fear that domestic introgression into junglefowl may compromise their wild genotype. By contrasting the whole genomes of 51 chickens with 63 junglefowl from across their natural range, we found evidence of a loss of the wild genotype across the Anthropocene. When comparing against the genomes of junglefowl from approximately a century ago using rigorous ancient-DNA protocols, we discovered that levels of domestic introgression are not equal among and within modern wild populations, with the percentage of domestic ancestry around 20-50%. We identified a number of domestication markers in which chickens are deeply differentiated from historic junglefowl regardless of breed and/or geographic provenance, with eight genes under selection. The latter are involved in pathways dealing with development, reproduction and vision. The wild genotype is an allelic reservoir that holds most of the genetic diversity of G. gallus, a species which is immensely important to human society. Our study provides fundamental genomic infrastructure to assist in efforts to prevent a further loss of the wild genotype through introgression of domestic alleles.


Subject(s)
Chickens , Genetics, Population , Genome , Animals , Chickens/genetics , Gene Flow , Genome/genetics , Genotype , Phylogeny
19.
N Engl J Med ; 386(2): 116-127, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34942067

ABSTRACT

BACKGROUND: Population-based data from the United States on the effectiveness of the three coronavirus disease 2019 (Covid-19) vaccines currently authorized by the Food and Drug Administration are limited. Whether declines in effectiveness are due to waning immunity, the B.1.617.2 (delta) variant of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), or other causes is unknown. METHODS: We used data for 8,690,825 adults in New York State to assess the effectiveness of the BNT162b2, mRNA-1273, and Ad26.COV2.S vaccines against laboratory-confirmed Covid-19 and hospitalization with Covid-19 (i.e., Covid-19 diagnosed at or after admission). We compared cohorts defined according to vaccine product received, age, and month of full vaccination with age-specific unvaccinated cohorts by linking statewide testing, hospital, and vaccine registry databases. We assessed vaccine effectiveness against Covid-19 from May 1 through September 3, 2021, and against hospitalization with Covid-19 from May 1 through August 31, 2021. RESULTS: There were 150,865 cases of Covid-19 and 14,477 hospitalizations with Covid-19. During the week of May 1, 2021, when the delta variant made up 1.8% of the circulating variants, the median vaccine effectiveness against Covid-19 was 91.3% (range, 84.1 to 97.0) for BNT162b2, 96.9% (range, 93.7 to 98.0) for mRNA-1273, and 86.6% (range, 77.8 to 89.7) for Ad26.COV2.S. Subsequently, effectiveness declined contemporaneously in all cohorts, from a median of 93.4% (range, 77.8 to 98.0) during the week of May 1 to a nadir of 73.5% (range, 13.8 to 90.0) around July 10, when the prevalence of the delta variant was 85.3%. By the week of August 28, when the prevalence of the delta variant was 99.6%, the effectiveness was 74.2% (range, 63.4 to 86.8). Effectiveness against hospitalization with Covid-19 among adults 18 to 64 years of age remained almost exclusively greater than 86%, with no apparent time trend. Effectiveness declined from May through August among persons 65 years of age or older who had received BNT162b2 (from 94.8 to 88.6%) or mRNA-1273 (from 97.1 to 93.7%). The effectiveness of Ad26.COV2.S was lower than that of the other vaccines, with no trend observed over time (range, 80.0 to 90.6%). CONCLUSIONS: The effectiveness of the three vaccines against Covid-19 declined after the delta variant became predominant. The effectiveness against hospitalization remained high, with modest declines limited to BNT162b2 and mRNA-1273 recipients 65 years of age or older.


Subject(s)
2019-nCoV Vaccine mRNA-1273 , Ad26COVS1 , BNT162 Vaccine , COVID-19/prevention & control , Hospitalization/statistics & numerical data , Vaccine Efficacy , Adolescent , Adult , Age Factors , Aged , COVID-19/epidemiology , COVID-19/virology , Cohort Studies , Humans , Incidence , Middle Aged , New York/epidemiology , SARS-CoV-2 , Young Adult
20.
Article in English | MEDLINE | ID: mdl-38868942

ABSTRACT

BACKGROUND: The chromatin-remodeling enzymes BRG1 (brahma-related gene 1) and CHD4 (chromodomain helicase DNA-binding protein 4) independently regulate the transcription of genes critical for vascular development, but their coordinated impact on vessels in late-stage embryos has not been explored. METHODS: In this study, we genetically deleted endothelial Brg1 and Chd4 in mixed background mice (Brg1fl/fl;Chd4fl/fl;VE-Cadherin-Cre+), and littermates that were negative for Cre recombinase were used as controls. Tissues were analyzed by immunostaining, immunoblot, and flow cytometry. Quantitative reverse transcription polymerase chain reaction was used to determine gene expression, and chromatin immunoprecipitation revealed gene targets of BRG1 and CHD4 in cultured endothelial cells. RESULTS: We found Brg1/Chd4 double mutants grew normally but died soon after birth with small and compact lungs. Despite having normal cellular composition, distal air sacs of the mutant lungs displayed diminished ECM (extracellular matrix) components and TGFß (transforming growth factor-ß) signaling, which typically promotes ECM synthesis. Transcripts for collagen- and elastin-related genes and the TGFß ligand Tgfb1 were decreased in mutant lung endothelial cells, but genetic deletion of endothelial Tgfb1 failed to recapitulate the small lungs and ECM defects seen in Brg1/Chd4 mutants. We instead found several ECM genes to be direct targets of BRG1 and CHD4 in cultured endothelial cells. CONCLUSIONS: Collectively, our data highlight essential roles for endothelial chromatin-remodeling enzymes in promoting ECM deposition in the distal lung tissue during the saccular stage of embryonic lung development.

SELECTION OF CITATIONS
SEARCH DETAIL