Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cell ; 170(4): 649-663.e13, 2017 Aug 10.
Article in English | MEDLINE | ID: mdl-28802038

ABSTRACT

Elevated risk of developing Alzheimer's disease (AD) is associated with hypomorphic variants of TREM2, a surface receptor required for microglial responses to neurodegeneration, including proliferation, survival, clustering, and phagocytosis. How TREM2 promotes such diverse responses is unknown. Here, we find that microglia in AD patients carrying TREM2 risk variants and TREM2-deficient mice with AD-like pathology have abundant autophagic vesicles, as do TREM2-deficient macrophages under growth-factor limitation or endoplasmic reticulum (ER) stress.Ā Combined metabolomics and RNA sequencing (RNA-seq) linked this anomalous autophagy to defective mammalian target of rapamycin (mTOR) signaling, which affects ATP levels and biosynthetic pathways. Metabolic derailment and autophagy were offset inĀ vitro through Dectin-1, a receptor that elicits TREM2-like intracellular signals, and cyclocreatine, a creatine analog that can supply ATP. Dietary cyclocreatine tempered autophagy, restored microglial clustering around plaques, and decreased plaque-adjacent neuronal dystrophy in TREM2-deficient mice with amyloid-Ɵ pathology. Thus, TREM2 enables microglial responses during AD by sustaining cellular energetic and biosynthetic metabolism.


Subject(s)
Alzheimer Disease/pathology , Energy Metabolism , Membrane Glycoproteins/metabolism , Microglia/metabolism , Receptors, Immunologic/metabolism , AMP-Activated Protein Kinases/metabolism , Alzheimer Disease/metabolism , Animals , Autophagy , Creatinine/analogs & derivatives , Creatinine/metabolism , Disease Models, Animal , Humans , Lectins, C-Type/metabolism , Macrophages/metabolism , Membrane Glycoproteins/genetics , Mice , Microglia/pathology , Neurites/metabolism , Plaque, Amyloid/metabolism , Receptors, Immunologic/genetics , TOR Serine-Threonine Kinases/metabolism
2.
Cell ; 160(3): 447-60, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25619688

ABSTRACT

Decreases in the diversity of enteric bacterial populations are observed in patients with Crohn's disease (CD) and ulcerative colitis (UC). Less is known about the virome in these diseases. We show that the enteric virome is abnormal in CD and UC patients. In-depth analysis of preparations enriched for free virions in the intestine revealed that CD and UC were associated with a significant expansion of Caudovirales bacteriophages. The viromes of CD and UC patients were disease and cohort specific. Importantly, it did not appear that expansion and diversification of the enteric virome was secondary to changes in bacterial populations. These data support a model in which changes in the virome may contribute to intestinal inflammation and bacterial dysbiosis. We conclude that the virome is a candidate for contributing to, or being a biomarker for, human inflammatory bowel disease and speculate that the enteric virome may play a role in other diseases.


Subject(s)
Caudovirales/isolation & purification , Colitis, Ulcerative/virology , Crohn Disease/virology , Dysbiosis/virology , Microviridae/isolation & purification , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Case-Control Studies , Caudovirales/genetics , Cohort Studies , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colitis, Ulcerative/therapy , Crohn Disease/microbiology , Crohn Disease/pathology , Crohn Disease/therapy , Dysbiosis/microbiology , Dysbiosis/pathology , Dysbiosis/therapy , Feces/microbiology , Feces/virology , Humans , Metagenome , Microviridae/genetics
3.
Cell ; 151(2): 253-66, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23063120

ABSTRACT

Pathogenic simian immunodeficiency virus (SIV) infection is associated with enteropathy, which likely contributes to AIDS progression. To identify candidate etiologies for AIDS enteropathy, we used next-generation sequencing to define the enteric virome during SIV infection in nonhuman primates. Pathogenic, but not nonpathogenic, SIV infection was associated with significant expansion of the enteric virome. We identified at least 32 previously undescribed enteric viruses during pathogenic SIV infection and confirmed their presence by using viral culture and PCR testing. We detected unsuspected mucosal adenovirus infection associated with enteritis as well as parvovirus viremia in animals with advanced AIDS, indicating the pathogenic potential of SIV-associated expansion of the enteric virome. No association between pathogenic SIV infection and the family-level taxonomy of enteric bacteria was detected. Thus, enteric viral infections may contribute to AIDS enteropathy and disease progression. These findings underline the importance of metagenomic analysis of the virome for understanding AIDS pathogenesis.


Subject(s)
Caliciviridae/isolation & purification , Intestines/virology , Parvoviridae/isolation & purification , Picornaviridae/isolation & purification , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Animals , Caliciviridae/classification , Caliciviridae/genetics , Chlorocebus aethiops , Feces/microbiology , Feces/virology , Intestines/microbiology , Molecular Sequence Data , Parvoviridae/classification , Parvoviridae/genetics , Phylogeny , Picornaviridae/classification , Picornaviridae/genetics , Polymerase Chain Reaction , Simian Acquired Immunodeficiency Syndrome/microbiology , Simian Immunodeficiency Virus/pathogenicity
4.
Nature ; 528(7583): 565-9, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26649827

ABSTRACT

Mycobacterium tuberculosis, a major global health threat, replicates in macrophages in part by inhibiting phagosome-lysosome fusion, until interferon-ƎĀ³ (IFNƎĀ³) activates the macrophage to traffic M. tuberculosis to the lysosome. How IFNƎĀ³ elicits this effect is unknown, but many studies suggest a role for macroautophagy (herein termed autophagy), a process by which cytoplasmic contents are targeted for lysosomal degradation. The involvement of autophagy has been defined based on studies in cultured cells where M. tuberculosis co-localizes with autophagy factors ATG5, ATG12, ATG16L1, p62, NDP52, BECN1 and LC3 (refs 2-6), stimulation of autophagy increases bacterial killing, and inhibition of autophagy increases bacterial survival. Notably, these studies reveal modest (~1.5-3-fold change) effects on M. tuberculosis replication. By contrast, mice lacking ATG5 in monocyte-derived cells and neutrophils (polymorponuclear cells, PMNs) succumb to M. tuberculosis within 30 days, an extremely severe phenotype similar to mice lacking IFNƎĀ³ signalling. Importantly, ATG5 is the only autophagy factor that has been studied during M. tuberculosis infection in vivo and autophagy-independent functions of ATG5 have been described. For this reason, we used a genetic approach to elucidate the role for multiple autophagy-related genes and the requirement for autophagy in resistance to M. tuberculosis infection in vivo. Here we show that, contrary to expectation, autophagic capacity does not correlate with the outcome of M. tuberculosis infection. Instead, ATG5 plays a unique role in protection against M. tuberculosis by preventing PMN-mediated immunopathology. Furthermore, while Atg5 is dispensable in alveolar macrophages during M. tuberculosis infection, loss of Atg5 in PMNs can sensitize mice to M. tuberculosis. These findings shift our understanding of the role of ATG5 during M. tuberculosis infection, reveal new outcomes of ATG5 activity, and shed light on early events in innate immunity that are required to regulate disease pathology and bacterial replication.


Subject(s)
Microtubule-Associated Proteins/metabolism , Mycobacterium tuberculosis , Neutrophils/immunology , Tuberculosis/immunology , Tuberculosis/pathology , Animals , Autophagy/genetics , Autophagy-Related Protein 5 , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Immunity, Innate/immunology , Interferon-gamma/deficiency , Interferon-gamma/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Male , Mice , Microtubule-Associated Proteins/deficiency , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/physiology , Neutrophils/metabolism , Tuberculosis/microbiology
5.
bioRxiv ; 2024 Feb 17.
Article in English | MEDLINE | ID: mdl-37292968

ABSTRACT

Background & Aims: Human intestinal enteroids (HIEs) are gaining recognition as physiologically relevant models of the intestinal epithelium. While HIEs from adults are used extensively in biomedical research, few studies have used HIEs from infants. Considering the dramatic developmental changes that occur during infancy, it is important to establish models that represent infant intestinal characteristics and physiological responses. Methods: We established jejunal HIEs from infant surgical samples and performed comparisons to jejunal HIEs from adults using RNA sequencing (RNA-Seq) and morphologic analyses. We validated differences in key pathways through functional studies and determined if these cultures recapitulate known features of the infant intestinal epithelium. Results: RNA-Seq analysis showed significant differences in the transcriptome of infant and adult HIEs, including differences in genes and pathways associated with cell differentiation and proliferation, tissue development, lipid metabolism, innate immunity, and biological adhesion. Validating these results, we observed a higher abundance of cells expressing specific enterocyte, goblet cell and enteroendocrine cell markers in differentiated infant HIE monolayers, and greater numbers of proliferative cells in undifferentiated 3D cultures. Compared to adult HIEs, infant HIEs portray characteristics of an immature gastrointestinal epithelium including significantly shorter cell height, lower epithelial barrier integrity, and lower innate immune responses to infection with an oral poliovirus vaccine. Conclusions: HIEs established from infant intestinal tissues reflect characteristics of the infant gut and are distinct from adult cultures. Our data support the use of infant HIEs as an ex-vivo model to advance studies of infant-specific diseases and drug discovery for this population.

6.
mBio ; 15(8): e0131624, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38953637

ABSTRACT

Human intestinal enteroids (HIEs) are gaining recognition as physiologically relevant models of the intestinal epithelium. While HIEs from adults are used extensively in biomedical research, few studies have used HIEs from infants. Considering the dramatic developmental changes that occur during infancy, it is important to establish models that represent infant intestinal characteristics and physiological responses. We established jejunal HIEs from infant surgical samples and performed comparisons to jejunal HIEs from adults using RNA sequencing (RNA-Seq) and morphologic analyses. We then validated differences in key pathways through functional studies and determined whether these cultures recapitulate known features of the infant intestinal epithelium. RNA-Seq analysis showed significant differences in the transcriptome of infant and adult HIEs, including differences in genes and pathways associated with cell differentiation and proliferation, tissue development, lipid metabolism, innate immunity, and biological adhesion. Validating these results, we observed a higher abundance of cells expressing specific enterocyte, goblet cell, and enteroendocrine cell markers in differentiated infant HIE monolayers, and greater numbers of proliferative cells in undifferentiated 3D cultures. Compared to adult HIEs, infant HIEs portray characteristics of an immature gastrointestinal epithelium including significantly shorter cell height, lower epithelial barrier integrity, and lower innate immune responses to infection with an oral poliovirus vaccine. HIEs established from infant intestinal tissues reflect characteristics of the infant gut and are distinct from adult cultures. Our data support the use of infant HIEs as an ex vivo model to advance studies of infant-specific diseases and drug discovery for this population. IMPORTANCE: Tissue or biopsy stem cell-derived human intestinal enteroids are increasingly recognized as physiologically relevant models of the human gastrointestinal epithelium. While enteroids from adults and fetal tissues have been extensively used for studying many infectious and non-infectious diseases, there are few reports on enteroids from infants. We show that infant enteroids exhibit both transcriptomic and morphological differences compared to adult cultures. They also differ in functional responses to barrier disruption and innate immune responses to infection, suggesting that infant and adult enteroids are distinct model systems. Considering the dramatic changes in body composition and physiology that begin during infancy, tools that appropriately reflect intestinal development and diseases are critical. Infant enteroids exhibit key features of the infant gastrointestinal epithelium. This study is significant in establishing infant enteroids as age-appropriate models for infant intestinal physiology, infant-specific diseases, and responses to pathogens.


Subject(s)
Intestinal Mucosa , Humans , Infant , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Adult , Cell Differentiation , Jejunum/cytology , Jejunum/immunology , Transcriptome , Organoids , Immunity, Innate , Female , Male , Infant, Newborn , Enterocytes
7.
bioRxiv ; 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38352333

ABSTRACT

Respiratory syncytial virus (RSV) is a common cause of respiratory infections, causing significant morbidity and mortality, especially in young children. Why RSV infection in children is more severe as compared to healthy adults is not fully understood. In the present study, we infect both pediatric and adult human nose organoid-air liquid interface (HNO-ALIs) cell lines with two contemporary RSV isolates and demonstrate how they differ in virus replication, induction of the epithelial cytokine response, cell injury, and remodeling. Pediatric HNO-ALIs were more susceptible to early RSV replication, elicited a greater overall cytokine response, demonstrated enhanced mucous production, and manifested greater cellular damage compared to their adult counterparts. Adult HNO-ALIs displayed enhanced mucus production and robust cytokine response that was well controlled by superior regulatory cytokine response and possibly resulted in lower cellular damage than in pediatric lines. Taken together, our data suggest substantial differences in how pediatric and adult upper respiratory tract epithelium responds to RSV infection. These differences in epithelial cellular response can lead to poor mucociliary clearance and predispose infants to a worse respiratory outcome of RSV infection.

8.
J Virol ; 86(24): 13515-23, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23035219

ABSTRACT

Human noroviruses (HuNoV) are the major cause of epidemic, nonbacterial gastroenteritis in the world. The short course of HuNoV-induced symptoms has implicated innate immunity in control of norovirus (NoV) infection. Studies using murine norovirus (MNV) confirm the importance of innate immune responses during NoV infection. Type I alpha and beta interferons (IFN-α/Ɵ) limit HuNoV replicon function, restrict MNV replication in cultured cells, and control MNV replication in vivo. Therefore, the cell types and transcription factors involved in antiviral immune responses and IFN-α/Ɵ-mediated control of NoV infection are important to define. We used mice with floxed alleles of the IFNAR1 chain of the IFN-α/Ɵ receptor to identify cells expressing lysozyme M or CD11c as cells that respond to IFN-α/Ɵ to restrict MNV replication in vivo. Furthermore, we show that the transcription factors IRF-3 and IRF-7 work in concert to initiate unique and overlapping antiviral responses to restrict MNV replication in vivo. IRF-3 and IRF-7 restrict MNV replication in both cultured macrophages and dendritic cells, are required for induction of IFN-α/Ɵ in macrophages but not dendritic cells, and are dispensable for the antiviral effects of IFN-α/Ɵ that block MNV replication. These studies suggest that expression of the IFN-α/Ɵ receptor on macrophages/neutrophils and dendritic cells, as well as of IRF-3 and IRF-7, is critical for innate immune responses to NoV infection.


Subject(s)
Interferon Regulatory Factor-3/physiology , Interferon Regulatory Factor-7/physiology , Interferon Type I/physiology , Norovirus/physiology , Virus Replication/physiology , Animals , Base Sequence , Cell Line , DNA Primers , Dendritic Cells/immunology , Immunity, Innate , Macrophages/immunology , Mice , Norovirus/immunology , Reverse Transcriptase Polymerase Chain Reaction
9.
Blood ; 117(5): 1530-9, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-21123823

ABSTRACT

Heterozygous deletions spanning chromosome 5q31.2 occur frequently in the myelodysplastic syndromes (MDS) and are highly associated with progression to acute myeloid leukemia (AML) when p53 is mutated. Mutagenesis screens in zebrafish and mice identified Hspa9 as a del(5q31.2) candidate gene that may contribute to MDS and AML pathogenesis, respectively. To test whether HSPA9 haploinsufficiency recapitulates the features of ineffective hematopoiesis observed in MDS, we knocked down the expression of HSPA9 in primary human hematopoietic cells and in a murine bone marrow-transplantation model using lentivirally mediated gene silencing. Knockdown of HSPA9 in human cells significantly delayed the maturation of erythroid precursors, but not myeloid or megakaryocytic precursors, and suppressed cell growth by 6-fold secondary to an increase in apoptosis and a decrease in the cycling of cells compared with control cells. Erythroid precursors, B lymphocytes, and the bone marrow progenitors c-kit(+)/lineage(-)/Sca-1(+) (KLS) and megakaryocyte/erythrocyte progenitor (MEP) were significantly reduced in a murine Hspa9-knockdown model. These abnormalities suggest that cooperating gene mutations are necessary for del(5q31.2) MDS cells to gain clonal dominance in the bone marrow. Our results demonstrate that Hspa9 haploinsufficiency alters the hematopoietic progenitor pool in mice and contributes to abnormal hematopoiesis.


Subject(s)
Carrier Proteins/physiology , Chromosome Deletion , Chromosomes, Mammalian/genetics , HSP70 Heat-Shock Proteins/physiology , Hematopoietic Stem Cells/pathology , Hematopoietic System/physiology , Myelodysplastic Syndromes/etiology , Animals , Apoptosis , Blotting, Western , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cells, Cultured , Fetal Blood/cytology , Fetal Blood/metabolism , Flow Cytometry , Haploinsufficiency , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Tumor Suppressor Protein p53/physiology
10.
Mol Cell Neurosci ; 49(3): 271-81, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22198539

ABSTRACT

Huntington's disease (HD) is a fatal, autosomal dominant neurodegenerative disorder caused by an expanded trinucleotide (CAG) repeat in exon 1 of the huntingtin gene (Htt). This expansion creates a toxic polyglutamine tract in the huntingtin protein (HTT). Currently, there is no treatment for either the progression or prevention of the disease. RNA interference (RNAi) technology has shown promise in transgenic mouse models of HD by reducing expression of mutant HTT and slowing disease progression. The advancement of RNAi therapies to human clinical trials is hampered by problems delivering RNAi to affected neurons in a robust and sustainable manner. Mesenchymal stem cells (MSC) have demonstrated a strong safety profile in both completed and numerous ongoing clinical trials. MSC exhibit a number of innate therapeutic effects, such as immune system modulation, homing to injury, and cytokine release into damaged microenvironments. The ability of MSC to transfer larger molecules and even organelles suggested their potential usefulness as delivery vehicles for therapeutic RNA inhibition. In a series of model systems we have found evidence that MSC can transfer RNAi targeting both reporter genes and mutant huntingtin in neural cell lines. MSC expressing shRNA antisense to GFP were found to decrease expression of GFP in SH-SY5Y cells after co-culture when assayed by flow cytometry. Additionally MSC expressing shRNA antisense to HTT were able to decrease levels of mutant HTT expressed in both U87 and SH-SY5Y target cells when assayed by Western blot and densitometry. These results are encouraging for expanding the therapeutic abilities of both RNAi and MSC for future treatments of Huntington's disease.


Subject(s)
Genetic Vectors , Mesenchymal Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , RNA Interference/physiology , Cell Line , Coculture Techniques , Down-Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Lentivirus/genetics
11.
Science ; 381(6657): 502-508, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37535745

ABSTRACT

The mammalian gut secretes a family of multifunctional peptides that affect appetite, intestinal secretions, and motility whereas others regulate the microbiota. We have found that peptide YY (PYY1-36), but not endocrine PYY3-36, acts as an antimicrobial peptide (AMP) expressed by gut epithelial paneth cells (PC). PC-PYY is packaged into secretory granules and is secreted into and retained by surface mucus, which optimizes PC-PYY activity. Although PC-PYY shows some antibacterial activity, it displays selective antifungal activity against virulent Candida albicans hyphae-but not the yeast form. PC-PYY is a cationic molecule that interacts with the anionic surfaces of fungal hyphae to cause membrane disruption and transcriptional reprogramming that selects for the yeast phenotype. Hence, PC-PYY is an antifungal AMP that contributes to the maintenance of gut fungal commensalism.


Subject(s)
Antifungal Agents , Antimicrobial Peptides , Candida , Paneth Cells , Peptide Fragments , Peptide YY , Animals , Antifungal Agents/metabolism , Antimicrobial Peptides/metabolism , Candida/drug effects , Candida/physiology , Paneth Cells/metabolism , Peptide Fragments/metabolism , Peptide YY/metabolism , Symbiosis , Humans , Mice
12.
J Clin Invest ; 133(21)2023 11 01.
Article in English | MEDLINE | ID: mdl-37909332

ABSTRACT

Cronkhite-Canada Syndrome (CCS) is a rare, noninherited polyposis syndrome affecting 1 in every million individuals. Despite over 50 years of CCS cases, the etiopathogenesis and optimal treatment for CCS remains unknown due to the rarity of the disease and lack of model systems. To better understand the etiology of CCS, we generated human intestinal organoids (HIOs) from intestinal stem cells isolated from 2 patients. We discovered that CCS HIOs are highly proliferative and have increased numbers of enteroendocrine cells producing serotonin (also known as 5-hydroxytryptamine or 5HT). These features were also confirmed in patient tissue biopsies. Recombinant 5HT increased proliferation of non-CCS donor HIOs and inhibition of 5HT production in the CCS HIOs resulted in decreased proliferation, suggesting a link between local epithelial 5HT production and control of epithelial stem cell proliferation. This link was confirmed in genetically engineered HIOs with an increased number of enteroendocrine cells. This work provides a new mechanism to explain the pathogenesis of CCS and illustrates the important contribution of HIO cultures to understanding disease etiology and in the identification of novel therapies. Our work demonstrates the principle of using organoids for personalized medicine and sheds light on how intestinal hormones can play a role in intestinal epithelial proliferation.


Subject(s)
Colorectal Neoplasms , Intestinal Polyposis , Humans , Serotonin , Intestines , Organoids/pathology , Colorectal Neoplasms/pathology , Intestinal Polyposis/genetics , Intestinal Polyposis/pathology
13.
bioRxiv ; 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37398483

ABSTRACT

We describe the epidemiology and clinical characteristics of 29 patients with cancer and diarrhea in whom Enteroaggregative Escherichia coli (EAEC) was initially identified by GI BioFire panel multiplex. E. coli strains were successfully isolated from fecal cultures in 14 of 29 patients. Six of the 14 strains were identified as EAEC and 8 belonged to other diverse E. coli groups of unknown pathogenesis. We investigated these strains by their adherence to human intestinal organoids, cytotoxic responses, antibiotic resistance profile, full sequencing of their genomes, and annotation of their functional virulome. Interestingly, we discovered novel and enhanced adherence and aggregative patterns for several diarrheagenic pathotypes that were not previously seen when co-cultured with immortalized cell lines. EAEC isolates displayed exceptional adherence and aggregation to human colonoids compared not only to diverse GI E. coli , but also compared to prototype strains of other diarrheagenic E. coli . Some of the diverse E. coli strains that could not be classified as a conventional pathotype also showed an enhanced aggregative and cytotoxic response. Notably, we found a high carriage rate of antibiotic resistance genes in both EAEC strains and diverse GI E. coli isolates and observed a positive correlation between adherence to colonoids and the number of metal acquisition genes carried in both EAEC and the diverse E. coli strains. This work indicates that E. coli from cancer patients constitute strains of remarkable pathotypic and genomic divergence, including strains of unknown disease etiology with unique virulomes. Future studies will allow for the opportunity to re-define E. coli pathotypes with greater diagnostic accuracy and into more clinically relevant groupings.

14.
Mol Ther ; 19(3): 584-93, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21119622

ABSTRACT

Induced pluripotent stem cells (iPSCs) have radically advanced the field of regenerative medicine by making possible the production of patient-specific pluripotent stem cells from adult individuals. By developing iPSCs to treat HIV, there is the potential for generating a continuous supply of therapeutic cells for transplantation into HIV-infected patients. In this study, we have used human hematopoietic stem cells (HSCs) to generate anti-HIV gene expressing iPSCs for HIV gene therapy. HSCs were dedifferentiated into continuously growing iPSC lines with four reprogramming factors and a combination anti-HIV lentiviral vector containing a CCR5 short hairpin RNA (shRNA) and a human/rhesus chimeric TRIM5α gene. Upon directed differentiation of the anti-HIV iPSCs toward the hematopoietic lineage, a robust quantity of colony-forming CD133(+) HSCs were obtained. These cells were further differentiated into functional end-stage macrophages which displayed a normal phenotypic profile. Upon viral challenge, the anti-HIV iPSC-derived macrophages exhibited strong protection from HIV-1 infection. Here, we demonstrate the ability of iPSCs to develop into HIV-1 resistant immune cells and highlight the potential use of iPSCs for HIV gene and cellular therapies.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Induced Pluripotent Stem Cells/metabolism , Macrophages/cytology , Macrophages/immunology , AC133 Antigen , Adult , Antigens, CD/metabolism , Antigens, CD34/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Glycoproteins/metabolism , HEK293 Cells , HIV Infections/virology , Humans , Induced Pluripotent Stem Cells/cytology , Peptides/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, CCR5/genetics , Receptors, CCR5/metabolism
15.
Cell Host Microbe ; 30(6): 809-823.e6, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35439436

ABSTRACT

Gut microbial diurnal oscillations are important diet-dependent drivers of host circadian rhythms and metabolism ensuring optimal energy balance. However, the interplay between diet, microbes, and host factors sustaining intestinal oscillations is complex and poorly understood. Here, using a mouse model, we report the host C-type lectin antimicrobial peptide Reg3ƎĀ³ works with key ileal microbes to orchestrate these interactions in a bidirectional manner and does not correlate with the intestinal core circadian clock. High-fat diet is the primary driver of microbial oscillators that impair host metabolic homeostasis, resulting in arrhythmic host Reg3ƎĀ³ expression that secondarily drives abundance and oscillation of key gut microbes. This illustrates transkingdom coordination of biological rhythms primarily influenced by diet and reciprocal sensor-effector signals between host and microbial components, ultimately driving metabolism. Restoring the gut microbiota's capacity to sense dietary signals mediated by specific host factors such as Reg3ƎĀ³ could be harnessed to improve metabolic dysfunction.


Subject(s)
Circadian Clocks , Gastrointestinal Microbiome , Circadian Rhythm , Diet , Diet, High-Fat/adverse effects , Lipid Metabolism
16.
Cell Host Microbe ; 19(5): 713-9, 2016 May 11.
Article in English | MEDLINE | ID: mdl-27107939

ABSTRACT

Immune responses differ between laboratory mice and humans. Chronic infection with viruses and parasites are common in humans, but are absent in laboratory mice, and thus represent potential contributors to inter-species differences in immunity. To test this, we sequentially infected laboratory mice with herpesviruses, influenza, and an intestinal helminth and compared their blood immune signatures to mock-infected mice before and after vaccination against yellow fever virus (YFV-17D). Sequential infection altered pre- and post-vaccination gene expression, cytokines, and antibodies in blood. Sequential pathogen exposure induced gene signatures that recapitulated those seen in blood from pet store-raised versus laboratory mice, and adult versus cord blood in humans. Therefore, basal and vaccine-induced murine immune responses are altered by infection with agents common outside of barrier facilities. This raises the possibility that we can improve mouse models of vaccination and immunity by selective microbial exposure of laboratory animals to mimic that of humans.


Subject(s)
Helminthiasis/immunology , Herpesviridae Infections/immunology , Herpesviridae/immunology , Intestinal Diseases, Parasitic/immunology , Yellow Fever Vaccine/immunology , Yellow Fever/immunology , Yellow Fever/prevention & control , Yellow fever virus/immunology , Animals , Antibodies/blood , Antibodies, Viral/immunology , Coinfection/immunology , Coinfection/parasitology , Coinfection/virology , Cytokines/blood , Disease Models, Animal , Fetal Blood/immunology , Gene Expression , Helminthiasis/prevention & control , Helminthiasis/virology , Herpesviridae Infections/prevention & control , Humans , Immunity, Innate , Immunoglobulin G/blood , Influenza, Human/immunology , Influenza, Human/prevention & control , Intestinal Diseases, Parasitic/prevention & control , Intestinal Diseases, Parasitic/virology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/parasitology , Orthomyxoviridae Infections/prevention & control , Yellow Fever/parasitology , Yellow Fever/virology , Yellow Fever Vaccine/pharmacology
17.
Science ; 352(6289): 1116-20, 2016 May 27.
Article in English | MEDLINE | ID: mdl-27230380

ABSTRACT

Inflammatory bowel disease (IBD) is associated with risk variants in the human genome and dysbiosis of the gut microbiome, though unifying principles for these findings remain largely undescribed. The human commensal Bacteroides fragilis delivers immunomodulatory molecules to immune cells via secretion of outer membrane vesicles (OMVs). We reveal that OMVs require IBD-associated genes, ATG16L1 and NOD2, to activate a noncanonical autophagy pathway during protection from colitis. ATG16L1-deficient dendritic cells do not induce regulatory T cells (T(regs)) to suppress mucosal inflammation. Immune cells from human subjects with a major risk variant in ATG16L1 are defective in T(reg) responses to OMVs. We propose that polymorphisms in susceptibility genes promote disease through defects in "sensing" protective signals from the microbiome, defining a potentially critical gene-environment etiology for IBD.


Subject(s)
Bacteroides fragilis/immunology , Carrier Proteins/genetics , Gastrointestinal Microbiome/immunology , Gene-Environment Interaction , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Nod2 Signaling Adaptor Protein/genetics , Adult , Aged , Animals , Autophagy/immunology , Autophagy-Related Proteins , Dendritic Cells/immunology , Extracellular Vesicles/immunology , Female , Genetic Predisposition to Disease , Genome, Human , Humans , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Polymorphism, Genetic , T-Lymphocytes, Regulatory/immunology
18.
Cell Host Microbe ; 19(1): 102-13, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26764600

ABSTRACT

Mutations in the autophagy gene EPG5 are linked to the multisystem human disease Vici syndrome, which is characterized in part by pulmonary abnormalities, including recurrent infections. We found that Epg5-deficient mice exhibited elevated baseline innate immune cellular and cytokine-based lung inflammation and were resistant to lethal influenza virus infection. Lung transcriptomics, bone marrow transplantation experiments, and analysis of cellular cytokine expression indicated that Epg5 plays a role in lung physiology through its function in macrophages. Deletion of other autophagy genes including Atg14, Fip200, Atg5, and Atg7 in myeloid cells also led to elevated basal lung inflammation and influenza resistance. This suggests that Epg5 and other Atg genes function in macrophages to limit innate immune inflammation in the lung. Disruption of this normal homeostatic dampening of lung inflammation results in increased resistance to influenza, suggesting that normal homeostatic mechanisms that limit basal tissue inflammation support some infectious diseases.


Subject(s)
Immunity, Innate , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/immunology , Pneumonia/immunology , Proteins/immunology , Animals , Autophagy-Related Protein 7 , Autophagy-Related Proteins , Homeostasis , Humans , Influenza, Human/genetics , Influenza, Human/virology , Macrophages/immunology , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/immunology , Pneumonia/genetics , Pneumonia/virology , Proteins/genetics , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/immunology
19.
Sci Rep ; 6: 23326, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26987296

ABSTRACT

CLEC16A is in a locus genetically linked to autoimmune diseases including multiple sclerosis, but the function of this gene in the nervous system is unknown. Here we show that two mouse strains carrying independent Clec16a mutations developed neurodegenerative disease characterized by motor impairments and loss of Purkinje cells. Neurons from Clec16a-mutant mice exhibited increased expression of the autophagy substrate p62, accumulation of abnormal intra-axonal membranous structures bearing the autophagy protein LC3, and abnormal Golgi morphology. Multiple aspects of endocytosis, lysosome and Golgi function were normal in Clec16a-deficient murine embryonic fibroblasts and HeLa cells. However, these cells displayed abnormal bulk autophagy despite unimpaired autophagosome formation. Cultured Clec16a-deficient cells exhibited a striking accumulation of LC3 and LAMP-1 positive autolysosomes containing undigested cytoplasmic contents. Therefore Clec16a, an autophagy protein that is critical for autolysosome function and clearance, is required for Purkinje cell survival.


Subject(s)
Lectins, C-Type/genetics , Lysosomes/physiology , Monosaccharide Transport Proteins/genetics , Motor Neuron Disease/pathology , Mutation , Purkinje Cells/cytology , Animals , Autophagy , Cell Survival , Cells, Cultured , Golgi Apparatus/pathology , HeLa Cells , Humans , Lectins, C-Type/metabolism , Mice , Monosaccharide Transport Proteins/metabolism , Motor Neuron Disease/genetics
20.
Science ; 347(6219): 266-9, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25431490

ABSTRACT

The capacity of human norovirus (NoV), which causes >90% of global epidemic nonbacterial gastroenteritis, to infect a subset of people persistently may contribute to its spread. How such enteric viruses establish persistent infections is not well understood. We found that antibiotics prevented persistent murine norovirus (MNoV) infection, an effect that was reversed by replenishment of the bacterial microbiota. Antibiotics did not prevent tissue infection or affect systemic viral replication but acted specifically in the intestine. The receptor for the antiviral cytokine interferon-λ, Ifnlr1, as well as the transcription factors Stat1 and Irf3, were required for antibiotics to prevent viral persistence. Thus, the bacterial microbiome fosters enteric viral persistence in a manner counteracted by specific components of the innate immune system.


Subject(s)
Caliciviridae Infections/virology , Cytokines/physiology , Gastroenteritis/virology , Intestines/microbiology , Microbiota , Norovirus/physiology , Symbiosis , Animals , Anti-Bacterial Agents/pharmacology , Caliciviridae Infections/drug therapy , Caliciviridae Infections/immunology , Caliciviridae Infections/microbiology , Female , Gastroenteritis/drug therapy , Gastroenteritis/immunology , Gastroenteritis/microbiology , Intestines/virology , Male , Mice, Inbred C57BL , Mice, Knockout , Microbiota/drug effects , Norovirus/immunology , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Signal Transduction , Viral Load , Virus Replication , Virus Shedding
SELECTION OF CITATIONS
SEARCH DETAIL