Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
2.
J Immunol ; 206(5): 936-940, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33504619

ABSTRACT

Dysregulated IL-17 expression is central to the pathogenesis of several inflammatory disorders, including ulcerative colitis. We have shown earlier that SUMOylation of ROR-γt, the transcription factor for IL-17, regulates colonic inflammation. In this study, we show that the expression of Ubc9, the E2 enzyme that targets ROR-γt for SUMOylation, is significantly reduced in the colonic mucosa of ulcerative colitis patients. Mechanistically, we demonstrate that hypoxia-inducible factor 1α (HIF-1α) binds to a CpG island within the Ubc9 gene promoter, resulting in its hypermethylation and reduced Ubc9 expression. CRISPR-Cas9-mediated inhibition of HIF-1α normalized Ubc9 and attenuated IL-17 expression in Th17 cells and reduced diseases severity in Rag1 -/- mice upon adoptive transfer. Collectively, our study reveals a novel epigenetic mechanism of regulation of ROR-γt that could be exploited in inflammatory diseases.


Subject(s)
Colitis, Ulcerative/genetics , DNA Methylation/genetics , Hypoxia/genetics , Interleukin-17/genetics , Promoter Regions, Genetic/genetics , Ubiquitin-Conjugating Enzymes/genetics , Animals , Colitis, Ulcerative/pathology , Colon/pathology , Humans , Hypoxia/pathology , Inflammation/genetics , Inflammation/pathology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Th17 Cells
3.
J Clin Immunol ; 41(2): 285-293, 2021 02.
Article in English | MEDLINE | ID: mdl-33392852

ABSTRACT

POLA1 encodes the catalytic unit of DNA polymerase α, which together with the Primase complex launches the DNA replication process. While complete deficiency of this essential gene is presumed to be lethal, at least two conditions due to partial POLA1 deficiency have been described. The first genetic syndrome to be mapped to POLA1 was X-linked reticulate pigmentary disorder (XLPDR, MIM #301220), a rare syndrome characterized by skin hyperpigmentation, sterile multiorgan inflammation, recurrent infections, and distinct facial features. XLPDR has been shown to be accompanied by profound activation of type I interferon signaling, but unlike other interferonopathies, it is not associated with autoantibodies or classical autoimmunity. Rather, it is accompanied by marked Natural Killer (NK) cell dysfunction, which may explain the recurrent infections seen in this syndrome. To date, all XLPDR cases are caused by the same recurrent intronic mutation, which results in gene missplicing. Several hypomorphic mutations in POLA1, distinct from the XLPDR intronic mutation, have been recently reported and these mutations associate with a separate condition, van Esch-O'Driscoll syndrome (VEODS, MIM #301030). This condition results in growth retardation, microcephaly, hypogonadism, and in some cases, overlapping immunological features to those seen in XLPDR. This review summarizes our current understanding of the clinical manifestations of POLA1 gene mutations with an emphasis on its immunological consequences, as well as recent advances in understanding of its pathophysiologic basis and potential therapeutic options.


Subject(s)
DNA Polymerase I/deficiency , DNA Polymerase I/genetics , Immune System Diseases/genetics , Intellectual Disability/genetics , Nervous System Malformations/genetics , Animals , Genes, X-Linked/genetics , Humans , Mutation/genetics
4.
Life Sci Alliance ; 4(3)2021 03.
Article in English | MEDLINE | ID: mdl-33372038

ABSTRACT

Paneth cells (PCs) are small intestinal epithelial cells that secrete antimicrobial peptides and growth factors, such as Wnt ligands. Intriguingly, the context in which PC-derived Wnt secretion is relevant in vivo remains unknown as intestinal epithelial ablation of Wnt does not affect homeostatic proliferation or restitution after irradiation injury. Considering the importance of growth factors in tumor development, we explored here the role of PCs in intestinal carcinogenesis using a genetic model of PC depletion through conditional expression of diphtheria toxin-α subunit. PC depletion in Apc Min mice impaired adenoma development in the small intestine and led to decreased Wnt3 expression in small bowel adenomas. To determine if PC-derived Wnt3 was required for adenoma development, we examined tumor formation after PC-specific ablation of Wnt3 We found that this was sufficient to decrease small intestinal adenoma formation; moreover, organoids derived from these tumors displayed slower growth capacity. Overall, we report that PC-derived Wnt3 is required to sustain early tumorigenesis in the small bowel and identify a clear role for PC-derived Wnt production in intestinal pathology.


Subject(s)
Adenoma/metabolism , Carcinogenesis/metabolism , Colorectal Neoplasms/metabolism , Intercellular Signaling Peptides and Proteins/deficiency , Intestine, Small/metabolism , Paneth Cells/metabolism , Wnt3 Protein/deficiency , Adenoma/genetics , Animals , Carcinogenesis/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/genetics , Disease Models, Animal , Female , Intercellular Signaling Peptides and Proteins/genetics , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organoids/metabolism , Signal Transduction/genetics , Wnt3 Protein/genetics
5.
JCI Insight ; 4(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31672938

ABSTRACT

X-linked reticulate pigmentary disorder (XLPDR, Mendelian Inheritance in Man #301220) is a rare syndrome characterized by recurrent infections and sterile multiorgan inflammation. The syndrome is caused by an intronic mutation in POLA1, the gene encoding the catalytic subunit of DNA polymerase-α (Pol-α), which is responsible for Okazaki fragment synthesis during DNA replication. Reduced POLA1 expression in this condition triggers spontaneous type I interferon expression, which can be linked to the autoinflammatory manifestations of the disease. However, the history of recurrent infections in this syndrome is as yet unexplained. Here we report that patients with XLPDR have reduced NK cell cytotoxic activity and decreased numbers of NK cells, particularly differentiated, stage V, cells (CD3-CD56dim). This phenotype is reminiscent of hypomorphic mutations in MCM4, which encodes a component of the minichromosome maintenance (MCM) helicase complex that is functionally linked to Pol-α during the DNA replication process. We find that POLA1 deficiency leads to MCM4 depletion and that both can impair NK cell natural cytotoxicity and show that this is due to a defect in lytic granule polarization. Altogether, our study provides mechanistic connections between Pol-α and the MCM complex and demonstrates their relevance in NK cell function.


Subject(s)
Amyloidosis, Familial/immunology , Genetic Diseases, X-Linked/immunology , Killer Cells, Natural/immunology , Pigmentation Disorders/immunology , Skin Diseases, Genetic/immunology , Amyloidosis, Familial/genetics , Cytotoxicity, Immunologic , DNA Repair , Genetic Diseases, X-Linked/genetics , Humans , K562 Cells , Minichromosome Maintenance Complex Component 4/genetics , Pigmentation Disorders/genetics , Recombination, Genetic , Skin Diseases, Genetic/genetics
6.
Elife ; 82019 10 30.
Article in English | MEDLINE | ID: mdl-31663849

ABSTRACT

Inflammatory bowel disease (IBD) affects 1.5-3.0 million people in the United States. IBD is genetically determined and many common risk alleles have been identified. Yet, a large proportion of genetic predisposition remains unexplained. In this study, we report the identification of an ultr arare missense variant (NM_006998.3:c.230G > A;p.Arg77His) in the SCGN gene causing Mendelian early-onset ulcerative colitis. SCGN encodes a calcium sensor that is exclusively expressed in neuroendocrine lineages, including enteroendocrine cells and gut neurons. SCGN interacts with the SNARE complex, which is required for vesicle fusion with the plasma membrane. We show that the SCGN mutation identified impacted the localization of the SNARE complex partner, SNAP25, leading to impaired hormone release. Finally, we show that mouse models of Scgn deficiency recapitulate impaired hormone release and susceptibility to DSS-induced colitis. Altogether, these studies demonstrate that functional deficiency in SCGN can result in intestinal inflammation and implicates the neuroendocrine cellular compartment in IBD.


Subject(s)
Colitis, Ulcerative/genetics , Genetic Predisposition to Disease , Secretagogins/deficiency , Animals , Cell Membrane/metabolism , Cytoplasmic Vesicles/metabolism , Disease Models, Animal , Humans , Membrane Fusion , Mice , Mutation, Missense , Protein Transport , SNARE Proteins/metabolism , Secretagogins/genetics , Synaptosomal-Associated Protein 25/metabolism
7.
J Exp Med ; 216(10): 2378-2393, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31358565

ABSTRACT

Chronic inflammation and gut microbiota dysbiosis, in particular the bloom of genotoxin-producing E. coli strains, are risk factors for the development of colorectal cancer. Here, we sought to determine whether precision editing of gut microbiota metabolism and composition could decrease the risk for tumor development in mouse models of colitis-associated colorectal cancer (CAC). Expansion of experimentally introduced E. coli strains in the azoxymethane/dextran sulfate sodium colitis model was driven by molybdoenzyme-dependent metabolic pathways. Oral administration of sodium tungstate inhibited E. coli molybdoenzymes and selectively decreased gut colonization with genotoxin-producing E. coli and other Enterobacteriaceae. Restricting the bloom of Enterobacteriaceae decreased intestinal inflammation and reduced the incidence of colonic tumors in two models of CAC, the azoxymethane/dextran sulfate sodium colitis model and azoxymethane-treated, Il10-deficient mice. We conclude that metabolic targeting of protumoral Enterobacteriaceae during chronic inflammation is a suitable strategy to prevent the development of malignancies arising from gut microbiota dysbiosis.


Subject(s)
Colitis/microbiology , Colorectal Neoplasms/microbiology , Dysbiosis/microbiology , Gastrointestinal Microbiome , Neoplasms, Experimental/microbiology , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/genetics , Dextran Sulfate/toxicity , Dysbiosis/chemically induced , Dysbiosis/genetics , Escherichia coli/growth & development , Interleukin-10/deficiency , Mice , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/genetics
8.
J Crohns Colitis ; 13(11): 1474-1478, 2019 Oct 28.
Article in English | MEDLINE | ID: mdl-31104071

ABSTRACT

The genetic basis of inflammatory bowel disease remains to be elucidated completely. Here we report on a patient with inflammatory bowel disease who has mosaic tetrasomy of the short arm of chromosome 9, a genomic region that harbours the type I interferon gene cluster. We show that increased interferon activation is present in peripheral blood and intestinal tissue from this patient, similar to previous reports of autoinflammatory organ damage driven by interferon activation in other patients with this chromosomal abnormality. To our knowledge, this is the first case of tetrasomy 9p-associated interferonopathy driving intestinal inflammation and highlights the role that type-I interferon pathways can play in the pathogenesis of intestinal inflammation.


Subject(s)
Aneuploidy , Inflammatory Bowel Diseases/genetics , Antigens/metabolism , Child , Chromosomes, Human, Pair 9 , Cytoskeletal Proteins/metabolism , Female , Humans , In Situ Hybridization, Fluorescence , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Mosaicism
9.
Am J Hum Genet ; 104(5): 957-967, 2019 05 02.
Article in English | MEDLINE | ID: mdl-31006512

ABSTRACT

Replicating the human genome efficiently and accurately is a daunting challenge involving the duplication of upward of three billion base pairs. At the core of the complex machinery that achieves this task are three members of the B family of DNA polymerases: DNA polymerases α, δ, and ε. Collectively these multimeric polymerases ensure DNA replication proceeds at optimal rates approaching 2 × 103 nucleotides/min with an error rate of less than one per million nucleotides polymerized. The majority of DNA replication of undamaged DNA is conducted by DNA polymerases δ and ε. The DNA polymerase α-primase complex performs limited synthesis to initiate the replication process, along with Okazaki-fragment synthesis on the discontinuous lagging strand. An increasing number of human disorders caused by defects in different components of the DNA-replication apparatus have been described to date. These are clinically diverse and involve a wide range of features, including variable combinations of growth delay, immunodeficiency, endocrine insufficiencies, lipodystrophy, and cancer predisposition. Here, by using various complementary approaches, including classical linkage analysis, targeted next-generation sequencing, and whole-exome sequencing, we describe distinct missense and splice-impacting mutations in POLA1 in five unrelated families presenting with an X-linked syndrome involving intellectual disability, proportionate short stature, microcephaly, and hypogonadism. POLA1 encodes the p180 catalytic subunit of DNA polymerase α-primase. A range of replicative impairments could be demonstrated in lymphoblastoid cell lines derived from affected individuals. Our findings describe the presentation of pathogenic mutations in a catalytic component of a B family DNA polymerase member, DNA polymerase α.


Subject(s)
DNA Polymerase I/genetics , DNA Primase/genetics , Genetic Diseases, X-Linked/etiology , Growth Disorders/etiology , Hypogonadism/etiology , Intellectual Disability/etiology , Microcephaly/etiology , Mutation , Adolescent , Adult , Child , Child, Preschool , Female , Genetic Diseases, X-Linked/pathology , Genotype , Growth Disorders/pathology , Humans , Hypogonadism/pathology , Infant , Intellectual Disability/pathology , Male , Microcephaly/pathology , Middle Aged , Pedigree , Exome Sequencing
10.
Cell Rep ; 24(9): 2342-2355, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30157428

ABSTRACT

Physiologic microbe-host interactions in the intestine require the maintenance of the microbiota in a luminal compartment through a complex interplay between epithelial and immune cells. However, the roles of mucosal myeloid cells in this process remain incompletely understood. In this study, we identified that decreased myeloid cell phagocytic activity promotes colon tumorigenesis. We show that this is due to bacterial accumulation in the lamina propria and present evidence that the underlying mechanism is bacterial induction of prostaglandin production by myeloid cells. Moreover, we show that similar events in the normal colonic mucosa lead to reductions in Tuft cells, goblet cells, and the mucus barrier of the colonic epithelium. These alterations are again linked to the induction of prostaglandin production in response to bacterial penetration of the mucosa. Altogether, our work highlights immune cell-epithelial cell interactions triggered by the microbiota that control intestinal immunity, epithelial differentiation, and carcinogenesis.


Subject(s)
Carcinogenesis/metabolism , Epithelial Cells/immunology , Intestines/physiopathology , Microbiota/physiology , Myeloid Cells/metabolism , Animals , Humans , Mice
11.
Nat Immunol ; 17(5): 495-504, 2016 May.
Article in English | MEDLINE | ID: mdl-27019227

ABSTRACT

Aberrant nucleic acids generated during viral replication are the main trigger for antiviral immunity, and mutations that disrupt nucleic acid metabolism can lead to autoinflammatory disorders. Here we investigated the etiology of X-linked reticulate pigmentary disorder (XLPDR), a primary immunodeficiency with autoinflammatory features. We discovered that XLPDR is caused by an intronic mutation that disrupts the expression of POLA1, which encodes the catalytic subunit of DNA polymerase-α. Unexpectedly, POLA1 deficiency resulted in increased production of type I interferons. This enzyme is necessary for the synthesis of RNA:DNA primers during DNA replication and, strikingly, we found that POLA1 is also required for the synthesis of cytosolic RNA:DNA, which directly modulates interferon activation. Together this work identifies POLA1 as a critical regulator of the type I interferon response.


Subject(s)
DNA Polymerase I/metabolism , DNA/biosynthesis , Interferon Type I/metabolism , RNA/biosynthesis , Base Sequence , Cells, Cultured , Cytosol/metabolism , DNA/genetics , DNA Polymerase I/genetics , Family Health , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Male , Microscopy, Confocal , Mutation , Oligonucleotide Array Sequence Analysis , Pedigree , Pigmentation Disorders/genetics , Pigmentation Disorders/metabolism , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction
12.
Stud Hist Philos Biol Biomed Sci ; 59: 125-34, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26965225

ABSTRACT

The question whether or not "viruses are alive" has caused considerable debate over many years. Yet, the question is effectively without substance because the answer depends entirely on the definition of life or the state of "being alive" that is bound to be arbitrary. In contrast, the status of viruses among biological entities is readily defined within the replicator paradigm. All biological replicators form a continuum along the selfishness-cooperativity axis, from the completely selfish to fully cooperative forms. Within this range, typical, lytic viruses represent the selfish extreme whereas temperate viruses and various mobile elements occupy positions closer to the middle of the range. Selfish replicators not only belong to the biological realm but are intrinsic to any evolving system of replicators. No such system can evolve without the emergence of parasites, and moreover, parasites drive the evolution of biological complexity at multiple levels. The history of life is a story of parasite-host coevolution that includes both the incessant arms race and various forms of cooperation. All organisms are communities of interacting, coevolving replicators of different classes. A complete theory of replicator coevolution remains to be developed, but it appears likely that not only the differentiation between selfish and cooperative replicators but the emergence of the entire range of replication strategies, from selfish to cooperative, is intrinsic to biological evolution.


Subject(s)
Virus Physiological Phenomena , Biological Evolution , Life , Virus Replication
13.
Methods Mol Biol ; 1280: 15-24, 2015.
Article in English | MEDLINE | ID: mdl-25736741

ABSTRACT

The NF-κB signaling pathway is a primary regulator of inflammation that has been implicated in the pathogenesis of immune disorders and cancer. This signaling network is strictly regulated; in a nonactivated state, NF-κB transcription factors are sequestered in the cytoplasm by the IκB family of proteins. Various pro-inflammatory stimuli result in the phosphorylation and subsequent ubiquitination of IκBs. These events lead to rapid degradation of IκB and allow translocation of the transcription factors to the nucleus. Therefore, ubiquitination and degradation of IκBs are critical steps in NF-κB pathway activation and can serve as a quantitative parameter to assess pathway activation. In this article, we present a detailed protocol for the quantification of in vivo ubiquitination and turnover of IκB-α in response to a variety of cellular stimuli.


Subject(s)
I-kappa B Proteins/metabolism , Ubiquitination , Animals , Blotting, Western/methods , Cell Line , Humans , Immunoprecipitation/methods , Mice , NF-KappaB Inhibitor alpha , Proteolysis , Transcription Factor RelA/metabolism , Ubiquitins/metabolism
14.
Methods Mol Biol ; 1280: 371-81, 2015.
Article in English | MEDLINE | ID: mdl-25736761

ABSTRACT

Nuclear factor-kappa B (NF-κB) is a family of transcription factors that regulate immune responses, cell proliferation, differentiation, and survival. Activity of the NF-κΒ pathway on a cellular level is tightly controlled through various mechanisms, one of which is the ubiquitin-dependent degradation of chromatin-bound NF-κB subunits. In general, the ubiquitination of NF-κB regulates the duration of gene transcription activated in response to inflammatory signals. In this article, we present protocols to examine the in vivo ubiquitination status of RelA, a critical protein of the NF-κB family.


Subject(s)
NF-kappa B/metabolism , Transcription Factor RelA/metabolism , Ubiquitination , Blotting, Western/methods , Cell Line , Gene Expression , Humans , Immunoprecipitation/methods , NF-kappa B/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Transcription Factor RelA/genetics , Ubiquitin/genetics , Ubiquitin/metabolism
15.
Mol Biol Cell ; 26(1): 91-103, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25355947

ABSTRACT

COMMD1 deficiency results in defective copper homeostasis, but the mechanism for this has remained elusive. Here we report that COMMD1 is directly linked to early endosomes through its interaction with a protein complex containing CCDC22, CCDC93, and C16orf62. This COMMD/CCDC22/CCDC93 (CCC) complex interacts with the multisubunit WASH complex, an evolutionarily conserved system, which is required for endosomal deposition of F-actin and cargo trafficking in conjunction with the retromer. Interactions between the WASH complex subunit FAM21, and the carboxyl-terminal ends of CCDC22 and CCDC93 are responsible for CCC complex recruitment to endosomes. We show that depletion of CCC complex components leads to lack of copper-dependent movement of the copper transporter ATP7A from endosomes, resulting in intracellular copper accumulation and modest alterations in copper homeostasis in humans with CCDC22 mutations. This work provides a mechanistic explanation for the role of COMMD1 in copper homeostasis and uncovers additional genes involved in the regulation of copper transporter recycling.


Subject(s)
Actin Cytoskeleton , Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphatases/metabolism , Cation Transport Proteins/metabolism , Microfilament Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Movement , Copper/metabolism , Copper-Transporting ATPases , Cytoplasm/metabolism , Endosomes/metabolism , HEK293 Cells , HeLa Cells , Homeostasis , Humans , Mice , Mutation , Neoplasm Proteins/metabolism , Proteins/genetics , Proteins/metabolism , Transport Vesicles/metabolism , Vesicular Transport Proteins
16.
Biol Direct ; 10: 25, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25433677

ABSTRACT

Our body harbors hundreds of microbial species and contains many more bacterial than human cells. These microbes are not passive riders but rather a vital component of the organism. The human microbiota affects our health in multiple ways, both positively and negatively. One of the new attractive directions in microbiome biology is the "microbiome-brain axis". Several groups of researchers have described the ability of the gut microbiota to communicate with the brain and thus modulate human behavior. These limited experimental data became the foundation of the "biomeme hypothesis" of possible microbial origin of some religious rituals that has recently appeared in Biology Direct. Here I propose a critical analysis of this hypothesis. I conclude that there is no evidence of the microbial origin of religious practices but there are strong indications of their psychological and social roots.


Subject(s)
Microbiota , Models, Biological , Bacteria/metabolism , Ceremonial Behavior , Decision Making , Humans , Interpersonal Relations , Religion
17.
Methods Mol Biol ; 1177: 193-209, 2014.
Article in English | MEDLINE | ID: mdl-24943324

ABSTRACT

The identification of true interacting partners of any given bait can be plagued by the nonspecific purification of irrelevant proteins. To avoid this problem, Tandem Affinity Purification (TAP) is a widely used procedure in molecular biology as this reduces the chance of nonspecific proteins being present in the final preparation. In this approach, two different affinity tags are fused to the protein bait. Herein, we review in detail a variation on the TAP procedure that we have previously developed, where the affinity moieties are placed on two different proteins that form a complex in vivo. This variation, which we refer to as Bimolecular Affinity Purification (BAP), is suited for the identification of specific molecular complexes marked by the presence of two known proteins. We have utilized BAP for characterization of molecular complexes and evaluation of proteins interaction. Another application of BAP is the isolation of ubiquitin-like proteins (UBL)-modified fractions of a given protein and characterization of the lysine-acceptor site and structure of UBL-chains.


Subject(s)
Chromatography, Affinity/methods , Multiprotein Complexes/isolation & purification , Ubiquitins/isolation & purification , Humans , Molecular Biology/methods , Multiprotein Complexes/chemistry , Protein Interaction Mapping/methods , Tandem Mass Spectrometry/methods , Ubiquitins/chemistry
18.
Gastroenterology ; 147(1): 184-195.e3, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24727021

ABSTRACT

BACKGROUND & AIMS: Activation of the transcription factor nuclear factor-κB (NF-κB) has been associated with the development of inflammatory bowel disease (IBD). Copper metabolism MURR1 domain containing 1 (COMMD1), a regulator of various transport pathways, has been shown to limit NF-κB activation. We investigated the roles of COMMD1 in the pathogenesis of colitis in mice and IBD in human beings. METHODS: We created mice with a specific disruption of Commd1 in myeloid cells (Mye-knockout [K/O] mice); we analyzed immune cell populations and functions and expression of genes regulated by NF-κB. Sepsis was induced in Mye-K/O and wild-type mice by cecal ligation and puncture or intraperitoneal injection of lipopolysaccharide (LPS), colitis was induced by administration of dextran sodium sulfate, and colitis-associated cancer was induced by administration of dextran sodium sulfate and azoxymethane. We measured levels of COMMD1 messenger RNA in colon biopsy specimens from 29 patients with IBD and 16 patients without (controls), and validated findings in an independent cohort (17 patients with IBD and 22 controls). We searched for polymorphisms in or near COMMD1 that were associated with IBD using data from the International IBD Genetics Consortium and performed quantitative trait locus analysis. RESULTS: In comparing gene expression patterns between myeloid cells from Mye-K/O and wild-type mice, we found that COMMD1 represses expression of genes induced by LPS. Mye-K/O mice had more intense inflammatory responses to LPS and developed more severe sepsis and colitis, with greater mortality. More Mye-K/O mice with colitis developed colon dysplasia and tumors than wild-type mice. We observed a reduced expression of COMMD1 in colon biopsy specimens and circulating leukocytes from patients with IBD. We associated single-nucleotide variants near COMMD1 with reduced expression of the gene and linked them with increased risk for ulcerative colitis. CONCLUSIONS: Expression of COMMD1 by myeloid cells has anti-inflammatory effects. Reduced expression or function of COMMD1 could be involved in the pathogenesis of IBD.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Colitis/prevention & control , Colitis/physiopathology , Colonic Neoplasms/prevention & control , Colonic Neoplasms/physiopathology , Inflammation/genetics , Inflammation/physiopathology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Azoxymethane/adverse effects , Biopsy , Case-Control Studies , Colitis/chemically induced , Colon/metabolism , Colon/pathology , Colonic Neoplasms/chemically induced , Dextran Sulfate/adverse effects , Disease Models, Animal , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Mice , Mice, Knockout , NF-kappa B/metabolism , Polymorphism, Single Nucleotide/genetics , RNA, Messenger/metabolism
19.
Autophagy ; 9(7): 1121-6, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23615436

ABSTRACT

Autophagy is a process in which a eukaryotic (but not prokaryotic) cell destroys its own components through the lysosomal machinery. This tightly regulated process is essential for normal cell growth, development, and homeostasis, serving to maintain a balance between synthesis and degradation, resulting in the recycling of cellular products. Here we try to expand the concept of autophagy and define it as a general mechanism of regulation encompassing various levels of the biosphere. Interestingly, one of the consequences of such an approach is that we must presume an existence of the autophagic processes in the prokaryotic domain.


Subject(s)
Autophagy , Animals , Eukaryotic Cells/cytology , Humans , Models, Biological , Prokaryotic Cells/cytology
20.
J Clin Invest ; 123(5): 2244-56, 2013 May.
Article in English | MEDLINE | ID: mdl-23563313

ABSTRACT

NF-κB is a master regulator of inflammation and has been implicated in the pathogenesis of immune disorders and cancer. Its regulation involves a variety of steps, including the controlled degradation of inhibitory IκB proteins. In addition, the inactivation of DNA-bound NF-κB is essential for its regulation. This step requires a factor known as copper metabolism Murr1 domain-containing 1 (COMMD1), the prototype member of a conserved gene family. While COMMD proteins have been linked to the ubiquitination pathway, little else is known about other family members. Here we demonstrate that all COMMD proteins bind to CCDC22, a factor recently implicated in X-linked intellectual disability (XLID). We showed that an XLID-associated CCDC22 mutation decreased CCDC22 protein expression and impaired its binding to COMMD proteins. Moreover, some affected individuals displayed ectodermal dysplasia, a congenital condition that can result from developmental NF-κB blockade. Indeed, patient-derived cells demonstrated impaired NF-κB activation due to decreased IκB ubiquitination and degradation. In addition, we found that COMMD8 acted in conjunction with CCDC22 to direct the degradation of IκB proteins. Taken together, our results indicate that CCDC22 participates in NF-κB activation and that its deficiency leads to decreased IκB turnover in humans, highlighting an important regulatory component of this pathway.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation , NF-kappa B/metabolism , Proteins/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Chromosomes, Human, X , Ectodermal Dysplasia/metabolism , Genetic Linkage , HEK293 Cells , HeLa Cells , Humans , I-kappa B Proteins/metabolism , Inflammation , Microscopy, Fluorescence , Mutation , NF-KappaB Inhibitor alpha , Neoplasms/metabolism , Protein Structure, Tertiary , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...