Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Science ; 381(6660): 851-857, 2023 08 25.
Article in English | MEDLINE | ID: mdl-37616368

ABSTRACT

The intestinal microbiota regulates mammalian lipid absorption, metabolism, and storage. We report that the microbiota reprograms intestinal lipid metabolism in mice by repressing the expression of long noncoding RNA (lncRNA) Snhg9 (small nucleolar RNA host gene 9) in small intestinal epithelial cells. Snhg9 suppressed the activity of peroxisome proliferator-activated receptor γ (PPARγ)-a central regulator of lipid metabolism-by dissociating the PPARγ inhibitor sirtuin 1 from cell cycle and apoptosis protein 2 (CCAR2). Forced expression of Snhg9 in the intestinal epithelium of conventional mice impaired lipid absorption, reduced body fat, and protected against diet-induced obesity. The microbiota repressed Snhg9 expression through an immune relay encompassing myeloid cells and group 3 innate lymphoid cells. Our findings thus identify an unanticipated role for a lncRNA in microbial control of host metabolism.


Subject(s)
Gastrointestinal Microbiome , Intestines , Lipid Metabolism , PPAR gamma , RNA, Long Noncoding , Sirtuin 1 , Animals , Mice , Immunity, Innate , Lipid Metabolism/genetics , Lymphocytes/immunology , PPAR gamma/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Sirtuin 1/metabolism , Cell Cycle Proteins/metabolism , Apoptosis Regulatory Proteins/metabolism , Myeloid Cells/immunology , Intestines/metabolism , Intestines/microbiology , Adipose Tissue/microbiology , Humans
2.
Elife ; 122023 04 26.
Article in English | MEDLINE | ID: mdl-37159507

ABSTRACT

Peristaltic movement of the intestine propels food down the length of the gastrointestinal tract to promote nutrient absorption. Interactions between intestinal macrophages and the enteric nervous system regulate gastrointestinal motility, yet we have an incomplete understanding of the molecular mediators of this crosstalk. Here, we identify complement component 1q (C1q) as a macrophage product that regulates gut motility. Macrophages were the predominant source of C1q in the mouse intestine and most extraintestinal tissues. Although C1q mediates the complement-mediated killing of bacteria in the bloodstream, we found that C1q was not essential for the immune defense of the intestine. Instead, C1q-expressing macrophages were located in the intestinal submucosal and myenteric plexuses where they were closely associated with enteric neurons and expressed surface markers characteristic of nerve-adjacent macrophages in other tissues. Mice with a macrophage-specific deletion of C1qa showed changes in enteric neuronal gene expression, increased neurogenic activity of peristalsis, and accelerated intestinal transit. Our findings identify C1q as a key regulator of gastrointestinal motility and provide enhanced insight into the crosstalk between macrophages and the enteric nervous system.


Subject(s)
Complement C1q , Enteric Nervous System , Mice , Animals , Complement C1q/metabolism , Gastrointestinal Motility/physiology , Macrophages/metabolism , Gastrointestinal Tract
3.
Science ; 374(6568): eabe6723, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34735226

ABSTRACT

A diverse group of antimicrobial proteins (AMPs) helps protect the mammalian intestine from varied microbial challenges. We show that small proline-rich protein 2A (SPRR2A) is an intestinal antibacterial protein that is phylogenetically unrelated to previously discovered mammalian AMPs. In this study, SPRR2A was expressed in Paneth cells and goblet cells and selectively killed Gram-positive bacteria by disrupting their membranes. SPRR2A shaped intestinal microbiota composition, restricted bacterial association with the intestinal surface, and protected against Listeria monocytogenes infection. SPRR2A differed from other intestinal AMPs in that it was induced by type 2 cytokines produced during helminth infection. Moreover, SPRR2A protected against helminth-induced bacterial invasion of intestinal tissue. Thus, SPRR2A is a distinctive AMP triggered by type 2 immunity that protects the intestinal barrier during helminth infection.


Subject(s)
Cornified Envelope Proline-Rich Proteins/metabolism , Gastrointestinal Microbiome , Gram-Positive Bacteria/physiology , Intestinal Mucosa/metabolism , Intestines/microbiology , Nematospiroides dubius , Strongylida Infections/immunology , Animals , Bacterial Load , Cell Membrane/metabolism , Cell Membrane Permeability , Cornified Envelope Proline-Rich Proteins/genetics , Cytokines/metabolism , Disease Susceptibility , Goblet Cells/metabolism , Humans , Immunity, Innate , Intestinal Mucosa/microbiology , Listeria monocytogenes/physiology , Listeriosis/microbiology , Mice , Microbial Viability , Paneth Cells/metabolism , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/metabolism , Strongylida Infections/metabolism , Strongylida Infections/microbiology
4.
Science ; 373(6561): eabf9232, 2021 Sep 17.
Article in English | MEDLINE | ID: mdl-34529485

ABSTRACT

Vitamin A and its derivative retinol are essential for the development of intestinal adaptive immunity. Retinoic acid (RA)­producing myeloid cells are central to this process, but how myeloid cells acquire retinol for conversion to RA is unknown. Here, we show that serum amyloid A (SAA) proteins­retinol-binding proteins induced in intestinal epithelial cells by the microbiota­deliver retinol to myeloid cells. We identify low-density lipoprotein (LDL) receptor­related protein 1 (LRP1) as an SAA receptor that endocytoses SAA-retinol complexes and promotes retinol acquisition by RA-producing intestinal myeloid cells. Consequently, SAA and LRP1 are essential for vitamin A­dependent immunity, including B and T cell homing to the intestine and immunoglobulin A production. Our findings identify a key mechanism by which vitamin A promotes intestinal immunity.


Subject(s)
Adaptive Immunity , Intestinal Mucosa/immunology , Intestine, Small/immunology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Myeloid Cells/metabolism , Serum Amyloid A Protein/metabolism , Vitamin A/metabolism , Animals , B-Lymphocytes/immunology , CD11c Antigen/analysis , CD4-Positive T-Lymphocytes/immunology , Cell Line , Endocytosis , Gene Deletion , Humans , Immunoglobulin A/biosynthesis , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestine, Small/cytology , Intestine, Small/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Mice , Mice, Inbred C57BL , Myeloid Cells/immunology , Protein Binding , Retinol-Binding Proteins/metabolism , Salmonella Infections, Animal/immunology , Salmonella typhimurium , Serum Amyloid A Protein/genetics , Th17 Cells/immunology
5.
Cell ; 184(16): 4154-4167.e12, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34324837

ABSTRACT

Environmental light cycles entrain circadian feeding behaviors in animals that produce rhythms in exposure to foodborne bacteria. Here, we show that the intestinal microbiota generates diurnal rhythms in innate immunity that synchronize with feeding rhythms to anticipate microbial exposure. Rhythmic expression of antimicrobial proteins was driven by daily rhythms in epithelial attachment by segmented filamentous bacteria (SFB), members of the mouse intestinal microbiota. Rhythmic SFB attachment was driven by the circadian clock through control of feeding rhythms. Mechanistically, rhythmic SFB attachment activated an immunological circuit involving group 3 innate lymphoid cells. This circuit triggered oscillations in epithelial STAT3 expression and activation that produced rhythmic antimicrobial protein expression and caused resistance to Salmonella Typhimurium infection to vary across the day-night cycle. Thus, host feeding rhythms synchronize with the microbiota to promote rhythms in intestinal innate immunity that anticipate exogenous microbial exposure.


Subject(s)
Circadian Clocks/physiology , Circadian Rhythm/physiology , Gastrointestinal Microbiome , Immunity, Innate , Animals , Antimicrobial Cationic Peptides/metabolism , Bacterial Adhesion , Cell Adhesion , Epithelial Cells/microbiology , Feeding Behavior , Intestine, Small/microbiology , Intestine, Small/ultrastructure , Lymphocytes/metabolism , Mice, Inbred C57BL , Muramidase/metabolism , Pancreatitis-Associated Proteins/metabolism , STAT3 Transcription Factor/metabolism , Salmonella Infections, Animal/microbiology , Signal Transduction
6.
Science ; 365(6460): 1428-1434, 2019 09 27.
Article in English | MEDLINE | ID: mdl-31604271

ABSTRACT

Circadian rhythmicity is a defining feature of mammalian metabolism that synchronizes metabolic processes to day-night light cycles. Here, we show that the intestinal microbiota programs diurnal metabolic rhythms in the mouse small intestine through histone deacetylase 3 (HDAC3). The microbiota induced expression of intestinal epithelial HDAC3, which was recruited rhythmically to chromatin, and produced synchronized diurnal oscillations in histone acetylation, metabolic gene expression, and nutrient uptake. HDAC3 also functioned noncanonically to coactivate estrogen-related receptor α, inducing microbiota-dependent rhythmic transcription of the lipid transporter gene Cd36 and promoting lipid absorption and diet-induced obesity. Our findings reveal that HDAC3 integrates microbial and circadian cues for regulation of diurnal metabolic rhythms and pinpoint a key mechanism by which the microbiota controls host metabolism.


Subject(s)
Circadian Rhythm , Epithelial Cells/metabolism , Gastrointestinal Microbiome , Histone Deacetylases/metabolism , Intestine, Small/metabolism , Acetylation , Animals , CD36 Antigens/metabolism , Chromatin/metabolism , Colon , Diet, High-Fat , Germ-Free Life , Intestine, Small/cytology , Jet Lag Syndrome , Lipid Metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Receptors, Estrogen/metabolism , ERRalpha Estrogen-Related Receptor
7.
Proc Natl Acad Sci U S A ; 116(38): 19077-19082, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31484771

ABSTRACT

Serum amyloid A (SAA) proteins are strongly induced in the liver by systemic infection and in the intestine by bacterial colonization. In infected mice, SAA proteins circulate in association with the vitamin A derivative retinol, suggesting that SAAs transport retinol during infection. Here we illuminate a structural basis for the retinol-SAA interaction. In the bloodstream of infected mice, most SAA is complexed with high-density lipoprotein (HDL). However, we found that the majority of the circulating retinol was associated with the small fraction of SAA proteins that circulate without binding to HDL, thus identifying free SAA as the predominant retinol-binding form in vivo. We then determined the crystal structure of retinol-bound mouse SAA3 at a resolution of 2.2 Å. Retinol-bound SAA3 formed a novel asymmetric trimeric assembly that was generated by the hydrophobic packing of the conserved amphipathic helices α1 and α3. This hydrophobic packing created a retinol-binding pocket in the center of the trimer, which was confirmed by mutagenesis studies. Together, these findings illuminate the molecular basis for retinol transport by SAA proteins during infection.


Subject(s)
Salmonella typhimurium/metabolism , Serum Amyloid A Protein/chemistry , Serum Amyloid A Protein/metabolism , Typhoid Fever/metabolism , Vitamin A/metabolism , Vitamins/metabolism , Animals , Crystallography, X-Ray , Mice , Mice, Knockout , Models, Molecular , Mutation , Protein Conformation , Serum Amyloid A Protein/genetics , Typhoid Fever/virology
8.
Proc Natl Acad Sci U S A ; 116(22): 10911-10916, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31097581

ABSTRACT

Vitamin A is a dietary component that is essential for the development of intestinal immunity. Vitamin A is absorbed and converted to its bioactive derivatives retinol and retinoic acid by the intestinal epithelium, yet little is known about how epithelial cells regulate vitamin A-dependent intestinal immunity. Here we show that epithelial cell expression of the transcription factor retinoic acid receptor ß (RARß) is essential for vitamin A-dependent intestinal immunity. Epithelial RARß activated vitamin A-dependent expression of serum amyloid A (SAA) proteins by binding directly to Saa promoters. In accordance with the known role of SAAs in regulating Th17 cell effector function, epithelial RARß promoted IL-17 production by intestinal Th17 cells. More broadly, epithelial RARß was required for the development of key vitamin A-dependent adaptive immune responses, including CD4+ T-cell homing to the intestine and the development of IgA-producing intestinal B cells. Our findings provide insight into how the intestinal epithelium senses dietary vitamin A status to regulate adaptive immunity, and highlight the role of epithelial cells in regulating intestinal immunity in response to diet.


Subject(s)
Immunity, Mucosal/physiology , Intestinal Mucosa/metabolism , Receptors, Retinoic Acid/metabolism , Serum Amyloid A Protein/metabolism , Vitamin A/metabolism , Animals , Cell Line , Gastrointestinal Microbiome/physiology , Hep G2 Cells , Humans , Mice , Receptors, Retinoic Acid/genetics , Serum Amyloid A Protein/genetics
9.
Cell Host Microbe ; 25(6): 777-788.e8, 2019 Jun 12.
Article in English | MEDLINE | ID: mdl-31101494

ABSTRACT

Vitamin A deficiency increases susceptibility to skin infection. However, the mechanisms by which vitamin A regulates skin immunity remain unclear. Here, we show that resistin-like molecule α (RELMα), a small secreted cysteine-rich protein, is expressed by epidermal keratinocytes and sebocytes and serves as an antimicrobial protein that is required for vitamin-A-dependent resistance to skin infection. RELMα was induced by microbiota colonization of the murine skin, was bactericidal in vitro, and was protected against bacterial infection of the skin in vivo. RELMα expression required dietary vitamin A and was induced by the therapeutic vitamin A analog isotretinoin, which protected against skin infection in a RELMα-dependent manner. The RELM family member Resistin was expressed in human skin, was induced by vitamin A analogs, and killed skin bacteria, indicating a conserved function for RELM proteins in skin innate immunity. Our findings provide insight into how vitamin A promotes resistance to skin infection.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Immunologic Factors/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Skin Diseases, Bacterial/prevention & control , Skin/immunology , Vitamin A/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/metabolism , Humans , Mice , Resistin/metabolism , Skin Diseases, Bacterial/immunology , Transcriptional Activation/drug effects
10.
Proc Natl Acad Sci U S A ; 114(42): 11027-11033, 2017 10 17.
Article in English | MEDLINE | ID: mdl-28973871

ABSTRACT

The mammalian intestine is colonized by trillions of bacteria that perform essential metabolic functions for their hosts. The mutualistic nature of this relationship depends on maintaining spatial segregation between these bacteria and the intestinal epithelial surface. This segregation is achieved in part by the presence of a dense mucus layer at the epithelial surface and by the production of antimicrobial proteins that are secreted by epithelial cells into the mucus layer. Here, we show that resistin-like molecule ß (RELMß) is a bactericidal protein that limits contact between Gram-negative bacteria and the colonic epithelial surface. Mouse and human RELMß selectively killed Gram-negative bacteria by forming size-selective pores that permeabilized bacterial membranes. In mice lacking RELMß, Proteobacteria were present in the inner mucus layer and invaded mucosal tissues. Another RELM family member, human resistin, was also bactericidal, suggesting that bactericidal activity is a conserved function of the RELM family. Our findings thus identify the RELM family as a unique family of bactericidal proteins and show that RELMß promotes host-bacterial mutualism by regulating the spatial segregation between the microbiota and the intestinal epithelium.


Subject(s)
Gastrointestinal Microbiome , Gram-Negative Bacteria , Hormones, Ectopic/physiology , Intestinal Mucosa/microbiology , Animals , Humans , Immunity, Innate , Intercellular Signaling Peptides and Proteins , Intestinal Mucosa/immunology , Lipid Metabolism , Mice , Resistin/physiology , Symbiosis
11.
Science ; 357(6354): 912-916, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28860383

ABSTRACT

The intestinal microbiota has been identified as an environmental factor that markedly affects energy storage and body-fat accumulation in mammals, yet the underlying mechanisms remain unclear. Here we show that the microbiota regulates body composition through the circadian transcription factor NFIL3. Nfil3 transcription oscillates diurnally in intestinal epithelial cells, and the amplitude of the circadian oscillation is controlled by the microbiota through group 3 innate lymphoid cells, STAT3 (signal transducer and activator of transcription 3), and the epithelial cell circadian clock. NFIL3 controls expression of a circadian lipid metabolic program and regulates lipid absorption and export in intestinal epithelial cells. These findings provide mechanistic insight into how the intestinal microbiota regulates body composition and establish NFIL3 as an essential molecular link among the microbiota, the circadian clock, and host metabolism.


Subject(s)
Basic-Leucine Zipper Transcription Factors/physiology , Body Composition/physiology , Circadian Clocks/physiology , Gastrointestinal Microbiome/physiology , Intestines/microbiology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Circadian Rhythm , Diet, High-Fat/adverse effects , Germ-Free Life , Glucose Tolerance Test , Insulin Resistance , Intestines/physiology , Lipid Metabolism/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Obesity/genetics , Obesity/microbiology , Promoter Regions, Genetic , STAT3 Transcription Factor/metabolism , Transcription, Genetic
12.
Science ; 357(6355): 1047-1052, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28751470

ABSTRACT

Intestinal Paneth cells limit bacterial invasion by secreting antimicrobial proteins, including lysozyme. However, invasive pathogens can disrupt the Golgi apparatus, interfering with secretion and compromising intestinal antimicrobial defense. Here we show that during bacterial infection, lysozyme is rerouted via secretory autophagy, an autophagy-based alternative secretion pathway. Secretory autophagy was triggered in Paneth cells by bacteria-induced endoplasmic reticulum (ER) stress, required extrinsic signals from innate lymphoid cells, and limited bacterial dissemination. Secretory autophagy was disrupted in Paneth cells of mice harboring a mutation in autophagy gene Atg16L1 that confers increased risk for Crohn's disease in humans. Our findings identify a role for secretory autophagy in intestinal defense and suggest why Crohn's disease is associated with genetic mutations that affect both the ER stress response and autophagy.


Subject(s)
Endoplasmic Reticulum Stress/immunology , Muramidase/metabolism , Paneth Cells/immunology , Paneth Cells/metabolism , Salmonella Infections/immunology , Salmonella enterica , Animals , Autophagy/genetics , Autophagy-Related Proteins , Carrier Proteins/genetics , Carrier Proteins/metabolism , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/microbiology , Endoplasmic Reticulum Stress/genetics , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Mutation , Paneth Cells/enzymology , Salmonella Infections/genetics
13.
Elife ; 32014 Oct 13.
Article in English | MEDLINE | ID: mdl-25310240

ABSTRACT

Innate lymphoid cells (ILCs) are recently identified lymphocytes that limit infection and promote tissue repair at mucosal surfaces. However, the pathways underlying ILC development remain unclear. Here we show that the transcription factor NFIL3 directs the development of a committed bone marrow precursor that differentiates into all known ILC lineages. NFIL3 was required in the common lymphoid progenitor (CLP), and was essential for the differentiation of αLP, a bone marrow cell population that gives rise to all known ILC lineages. Clonal differentiation studies revealed that CXCR6(+) cells within the αLP population differentiate into all ILC lineages but not T- and B-cells. We further show that NFIL3 governs ILC development by directly regulating expression of the transcription factor TOX. These findings establish that NFIL3 directs the differentiation of a committed ILC precursor that gives rise to all ILC lineages and provide insight into the defining role of NFIL3 in ILC development.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Immunity, Innate , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Animals , B-Lymphocytes/cytology , Basic-Leucine Zipper Transcription Factors/deficiency , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Lineage , Citrobacter rodentium , Disease Susceptibility/immunology , Disease Susceptibility/microbiology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/microbiology , Homeodomain Proteins/metabolism , Host-Pathogen Interactions/immunology , Mice , Receptors, CXCR/metabolism , Receptors, CXCR6 , T-Lymphocytes/cytology
14.
Elife ; 3: e03206, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25073702

ABSTRACT

Retinol plays a vital role in the immune response to infection, yet proteins that mediate retinol transport during infection have not been identified. Serum amyloid A (SAA) proteins are strongly induced in the liver by systemic infection and in the intestine by bacterial colonization, but their exact functions remain unclear. Here we show that mouse and human SAAs are retinol binding proteins. Mouse and human SAAs bound retinol with nanomolar affinity, were associated with retinol in vivo, and limited the bacterial burden in tissues after acute infection. We determined the crystal structure of mouse SAA3 at a resolution of 2 Å, finding that it forms a tetramer with a hydrophobic binding pocket that can accommodate retinol. Our results thus identify SAAs as a family of microbe-inducible retinol binding proteins, reveal a unique protein architecture involved in retinol binding, and suggest how retinol is circulated during infection.


Subject(s)
Retinol-Binding Proteins/chemistry , Salmonella Infections/metabolism , Serum Amyloid A Protein/chemistry , Vitamin A/metabolism , Animals , Biological Transport , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Hep G2 Cells , Humans , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/microbiology , Kinetics , Liver/immunology , Liver/metabolism , Liver/microbiology , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Protein Multimerization , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Retinol-Binding Proteins/genetics , Retinol-Binding Proteins/immunology , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/physiology , Serum Amyloid A Protein/genetics , Serum Amyloid A Protein/immunology , Tissue Culture Techniques , Vitamin A/administration & dosage
15.
PLoS One ; 9(5): e96544, 2014.
Article in English | MEDLINE | ID: mdl-24824433

ABSTRACT

CNS inflammation is a hallmark of neurodegenerative disease, and recent studies suggest that the inflammatory response may contribute to neuronal demise. In particular, increased tumor necrosis factor (TNF) signaling is implicated in the pathology of both Parkinson's disease (PD) and Alzheimer's disease (AD). We have previously shown that localized gene delivery of dominant negative TNF to the degenerating brain region can limit pathology in animal models of PD and AD. TNF is upregulated in Huntington's disease (HD), like in PD and AD, but it is unknown whether TNF signaling contributes to neuronal degeneration in HD. We used in vivo gene delivery to test whether selective reduction of soluble TNF signaling could attenuate medium spiny neuron (MSN) degeneration in the YAC128 transgenic (TG) mouse model of Huntington's disease (HD). AAV vectors encoding cDNA for dominant-negative tumor necrosis factor (DN-TNF) or GFP (control) were injected into the striatum of young adult wild type WT and YAC128 TG mice and achieved 30-50% target coverage. Expression of dominant negative TNF protein was confirmed immunohistologically and biochemically and was maintained as mice aged to one year, but declined significantly over time. However, the extent of striatal DN-TNF gene transfer achieved in our studies was not sufficient to achieve robust effects on neuroinflammation, rescue degenerating MSNs or improve motor function in treated mice. Our findings suggest that alternative drug delivery strategies should be explored to determine whether greater target coverage by DN-TNF protein might afford some level of neuroprotection against HD-like pathology and/or that soluble TNF signaling may not be the primary driver of striatal neuroinflammation and MSN loss in YAC128 TG mice.


Subject(s)
Corpus Striatum/metabolism , Genetic Therapy/methods , Huntington Disease/therapy , Neurons/metabolism , Tumor Necrosis Factor-alpha/genetics , Animals , Corpus Striatum/pathology , Disease Models, Animal , Gene Transfer Techniques , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Motor Skills/physiology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Nerve Degeneration/therapy , Neurons/pathology , Rotarod Performance Test , Treatment Outcome , Tumor Necrosis Factor-alpha/metabolism
16.
Science ; 342(6159): 727-30, 2013 Nov 08.
Article in English | MEDLINE | ID: mdl-24202171

ABSTRACT

Circadian clocks regulate numerous physiological processes that vary across the day-night (diurnal) cycle, but if and how the circadian clock regulates the adaptive immune system is mostly unclear. Interleukin-17-producing CD4(+) T helper (T(H)17) cells are proinflammatory immune cells that protect against bacterial and fungal infections at mucosal surfaces. Their lineage specification is regulated by the orphan nuclear receptor RORγt. We show that the transcription factor NFIL3 suppresses T(H)17 cell development by directly binding and repressing the Rorγt promoter. NFIL3 links T(H)17 cell development to the circadian clock network through the transcription factor REV-ERBα. Accordingly, TH17 lineage specification varies diurnally and is altered in Rev-erbα(-/-) mice. Light-cycle disruption elevated intestinal T(H)17 cell frequencies and increased susceptibility to inflammatory disease. Thus, lineage specification of a key immune cell is under direct circadian control.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/genetics , Circadian Clocks/immunology , Gene Expression Regulation , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Th17 Cells/cytology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , CLOCK Proteins/genetics , Cell Lineage/genetics , Circadian Clocks/genetics , Germ-Free Life , HEK293 Cells , Humans , Intestine, Small/immunology , Intestine, Small/microbiology , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Promoter Regions, Genetic
17.
J Neuroinflammation ; 10: 50, 2013 Apr 28.
Article in English | MEDLINE | ID: mdl-23622116

ABSTRACT

BACKGROUND: Complex interactions involving genetic susceptibility and environmental factors are thought to underlie the pathogenesis of Parkinson's disease (PD). Although the role of inflammatory processes in modulating risk for development of PD has yet to be fully understood, prospective studies suggest that chronic use of NSAIDs reduce the incidence of PD. Loss-of-function mutations in the DJ-1 gene cause a rare form of familial PD with an autosomal recessive pattern of inheritance; however, DJ-1-/- mice do not display nigrostriatal pathway degeneration, suggesting that additional factors such as inflammation may be needed to induce neurodegeneration on the background of DJ-1 gene mutations. Neuroinflammation causes oxidative stress and, based on evidence that DJ-1 plays a protective role against oxidative stress, we investigated whether DJ-1-/- mice display increased vulnerability to inflammation-induced nigral degeneration. METHODS: We exposed adult wild-type and DJ-1-/- mice to repeated intranasal administration of soluble TNF (inTNF) or repeated intraperitoneal injections of low-dose lipopolysaccharide (LPS) or saline vehicle. We measured locomotor performance using a variety of behavior tasks, striatal dopamine (DA) content by HPLC, DA neuron (TH+ cells) and total neuron (NeuN+ cells) number in the substantia nigra pars compacta and ventral tegmental area by unbiased stereology, number of Iba1-positive microglia, and mRNA levels of inflammatory and oxidative stress genes by quantitative PCR in the midbrain, cortex and isolated peritoneal macrophages of DJ-1-/- and wild-type mice. RESULTS: We found that chronic LPS injections induced similar neuroinflammatory responses in the midbrains of DJ-1-/- mice and wild-type mice and neither group developed locomotor deficits or nigral degeneration. inTNF administration did not appear to induce neuroinflammatory responses in LPS-treated wild-type or DJ-1-/- mice. The lack of vulnerability to inflammation-induced nigral degeneration was not due to enhanced anti-oxidant gene responses in the midbrains of DJ-1-/- mice which, in fact, displayed a blunted response relative to that of wild-type mice. Peripheral macrophages from wild-type and DJ-1-/- mice displayed similar basal and LPS-induced inflammatory and oxidative stress markers in vitro. CONCLUSIONS: Our studies indicate that DJ-1-/- mice do not display increased vulnerability to inflammation-related nigral degeneration in contrast to what has been reported for 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine. We conclude that either DJ-1 does not have a critical role in protecting DA neurons against inflammation-induced oxidative stress and/or there is compensatory gene expression in the midbrain of DJ-1-/- mice that renders them resistant to the cytotoxic effects triggered by chronic peripheral inflammation.


Subject(s)
Inflammation/pathology , Motor Activity/physiology , Nerve Degeneration/pathology , Oncogene Proteins/physiology , Substantia Nigra/pathology , Administration, Intranasal , Animals , Behavior, Animal/drug effects , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Immunohistochemistry , Inflammation/chemically induced , Injections, Intraperitoneal , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Oncogene Proteins/genetics , Oxidative Stress/physiology , Peroxiredoxins , Postural Balance/drug effects , Protein Deglycase DJ-1 , Psychomotor Performance/drug effects , Real-Time Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/pharmacology , Tyrosine 3-Monooxygenase/metabolism
18.
Science ; 334(6053): 255-8, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21998396

ABSTRACT

The mammalian intestine is home to ~100 trillion bacteria that perform important metabolic functions for their hosts. The proximity of vast numbers of bacteria to host intestinal tissues raises the question of how symbiotic host-bacterial relationships are maintained without eliciting potentially harmful immune responses. Here, we show that RegIIIγ, a secreted antibacterial lectin, is essential for maintaining a ~50-micrometer zone that physically separates the microbiota from the small intestinal epithelial surface. Loss of host-bacterial segregation in RegIIIγ(-/-) mice was coupled to increased bacterial colonization of the intestinal epithelial surface and enhanced activation of intestinal adaptive immune responses by the microbiota. Together, our findings reveal that RegIIIγ is a fundamental immune mechanism that promotes host-bacterial mutualism by regulating the spatial relationships between microbiota and host.


Subject(s)
Gram-Negative Bacteria/physiology , Gram-Positive Bacteria/physiology , Intestinal Mucosa/microbiology , Intestine, Small/microbiology , Metagenome , Proteins/metabolism , Adaptive Immunity , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Load , Gram-Negative Bacteria/immunology , Gram-Positive Bacteria/immunology , Homeostasis , Immunoglobulin A/analysis , Intestinal Mucosa/immunology , Intestine, Small/immunology , Lectins, C-Type/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Pancreatitis-Associated Proteins , Symbiosis , T-Lymphocytes, Helper-Inducer/immunology
19.
Proc Natl Acad Sci U S A ; 108(21): 8743-8, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555560

ABSTRACT

The mammalian gastrointestinal tract harbors thousands of bacterial species that include symbionts as well as potential pathogens. The immune responses that limit access of these bacteria to underlying tissue remain poorly defined. Here we show that γδ intraepithelial lymphocytes (γδ IEL) of the small intestine produce innate antimicrobial factors in response to resident bacterial "pathobionts" that penetrate the intestinal epithelium. γδ IEL activation was dependent on epithelial cell-intrinsic MyD88, suggesting that epithelial cells supply microbe-dependent cues to γδ IEL. Finally, γδ T cells protect against invasion of intestinal tissues by resident bacteria specifically during the first few hours after bacterial encounter, indicating that γδ IEL occupy a unique temporal niche among intestinal immune defenses. Thus, γδ IEL detect the presence of invading bacteria through cross-talk with neighboring epithelial cells and are an essential component of the hierarchy of immune defenses that maintain homeostasis with the intestinal microbiota.


Subject(s)
Homeostasis/immunology , Host-Pathogen Interactions/immunology , Intestinal Mucosa/immunology , Lymphocytes/immunology , Receptors, Antigen, T-Cell, gamma-delta/physiology , Animals , Bacteria/immunology , Cell Communication/immunology , Epithelial Cells , Immunity, Innate , Metagenome/immunology , Mice , Mice, Knockout
20.
Mol Ther ; 19(1): 46-52, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20959812

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder typified by the loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc). Recent evidence indicates that neuroinflammation may play a critical role in the pathogenesis of PD, particularly tumor necrosis factor (TNF). We have previously shown that soluble TNF (solTNF) is required to mediate robust degeneration induced by 6-hydroxydopamine (6-OHDA) or lipopolysaccharide. What remains unknown is whether TNF inhibition can attenuate the delayed and progressive phase of neurodegeneration. To test this, rats were injected in the SNpc with lentivirus encoding dominant-negative TNF (lenti-DN-TNF) 2 weeks after receiving a 6-OHDA lesion. Remarkably, when examined 5 weeks after the initial 6-OHDA lesion, no further loss of nigral DA neurons was observed. Lenti-DN-TNF also attenuated microglial activation. Together, these data suggest that TNF is likely a critical mediator of nigral DA neuron death during the delayed and progressive phase of neurodegeneration, and that microglia may be the principal cell type involved. These promising findings provide compelling reasons to perform DN-TNF gene transfer studies in nonhuman primates with the long-term goal of using it in the clinic to prevent the delayed and progressive degeneration of DA neurons that gives rise to motor symptoms in PD.


Subject(s)
Dopamine/metabolism , Genetic Therapy/methods , Neurons/pathology , Parkinson Disease/pathology , Parkinson Disease/therapy , Substantia Nigra/pathology , Tumor Necrosis Factor-alpha/genetics , Animals , Astrocytes/metabolism , Astrocytes/pathology , Cell Death/genetics , Disease Models, Animal , Female , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Oxidopamine , Parkinson Disease/genetics , Parkinson Disease/metabolism , Rats , Rats, Sprague-Dawley , Substantia Nigra/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...