Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Ther ; 32(6): 1643-1657, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38582963

ABSTRACT

Gene therapy in hematopoietic stem and progenitor cells (HSPCs) shows great potential for the treatment of inborn metabolic diseases. Typical HSPC gene therapy approaches rely on constitutive promoters to express a therapeutic transgene, which is associated with multiple disadvantages. Here, we propose a novel promoterless intronic gene editing approach that triggers transgene expression only after cellular differentiation into the myeloid lineage. We integrated a splicing-competent eGFP cassette into the first intron of CD11b and observed expression of eGFP in the myeloid lineage but minimal to no expression in HSPCs or differentiated non-myeloid lineages. In vivo, edited HSPCs successfully engrafted in immunodeficient mice and displayed transgene expression in the myeloid compartment of multiple tissues. Using the same approach, we expressed alpha-L-iduronidase (IDUA), the defective enzyme in Mucopolysaccharidosis type I, and observed a 10-fold supraendogenous IDUA expression exclusively after myeloid differentiation. Edited cells efficiently populated bone marrow, blood, and spleen of immunodeficient mice, and retained the capacity to secrete IDUA ex vivo. Importantly, cells edited with the eGFP and IDUA transgenes were also found in the brain. This approach may unlock new therapeutic strategies for inborn metabolic and neurological diseases that require the delivery of therapeutics in brain.


Subject(s)
Gene Editing , Hematopoietic Stem Cells , Introns , Myeloid Cells , Transcription Activator-Like Effector Nucleases , Transgenes , Animals , Gene Editing/methods , Mice , Hematopoietic Stem Cells/metabolism , Humans , Myeloid Cells/metabolism , Transcription Activator-Like Effector Nucleases/genetics , Transcription Activator-Like Effector Nucleases/metabolism , Cell Differentiation/genetics , Genetic Therapy/methods , Iduronidase/genetics , Iduronidase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Gene Expression , Cell Lineage/genetics , CD11b Antigen/genetics , CD11b Antigen/metabolism , Hematopoietic Stem Cell Transplantation/methods , Mucopolysaccharidosis I/therapy , Mucopolysaccharidosis I/genetics
2.
Infect Immun ; 86(11)2018 11.
Article in English | MEDLINE | ID: mdl-30104215

ABSTRACT

Helminths have coevolved with their hosts, resulting in the development of specialized host immune mechanisms and parasite-specific regulatory products. Identification of new pathways that regulate helminth infection could provide a better understanding of host-helminth interaction and may identify new therapeutic targets for helminth infection. Here we identify the endocannabinoid system as a new mechanism that influences host immunity to helminths. Endocannabinoids are lipid-derived signaling molecules that control important physiologic processes, such as feeding behavior and metabolism. Following murine infection with Nippostrongylus brasiliensis, an intestinal nematode with a life cycle similar to that of hookworms, we observed increased levels of endocannabinoids (2-arachidonoylglycerol [2-AG] or anandamide [AEA]) and the endocannabinoid-like molecule oleoylethanolamine (OEA) in infected lung and intestine. To investigate endocannabinoid function in helminth infection, we employed pharmacological inhibitors of cannabinoid subtype receptors 1 and 2 (CB1R and CB2R). Compared to findings for vehicle-treated mice, inhibition of CB1R but not CB2R resulted in increased N. brasiliensis worm burden and egg output, associated with significantly decreased expression of the T helper type 2 cytokine interleukin 5 (IL-5) in intestinal tissue and splenocyte cultures. Strikingly, bioinformatic analysis of genomic and transcriptome sequencing (RNA-seq) data sets identified putative genes encoding endocannabinoid biosynthetic and degradative enzymes in many parasitic nematodes. To test the novel hypothesis that helminth parasites produce their own endocannabinoids, we measured endocannabinoid levels in N. brasiliensis by mass spectrometry and quantitative PCR and found that N. brasiliensis parasites produced endocannabinoids, especially at the infectious larval stage. To our knowledge, this is the first report of helminth- and host-derived endocannabinoids that promote host immune responses and reduce parasite burden.


Subject(s)
Endocannabinoids/metabolism , Host-Pathogen Interactions , Immunologic Factors/metabolism , Nippostrongylus/growth & development , Nippostrongylus/metabolism , Strongylida Infections/immunology , Strongylida Infections/pathology , Animals , Cytokines/metabolism , Disease Models, Animal , Intestines/pathology , Leukocytes, Mononuclear/immunology , Lung/pathology , Mass Spectrometry , Mice , Nippostrongylus/chemistry , Parasite Egg Count , Parasite Load
3.
J Leukoc Biol ; 104(4): 855-869, 2018 10.
Article in English | MEDLINE | ID: mdl-29992625

ABSTRACT

Resistin-like molecule α (RELMα) is a highly secreted protein in type 2 (Th2) cytokine-induced inflammation including helminth infection and allergy. In infection with Nippostrongylus brasiliensis (Nb), RELMα dampens Th2 inflammatory responses. RELMα is expressed by immune cells, and by epithelial cells (EC); however, the functional impact of immune versus EC-derived RELMα is unknown. We generated bone marrow (BM) chimeras that were RELMα deficient (RELMα-/- ) in BM or non BM cells and infected them with Nb. Non BM RELMα-/- chimeras had comparable inflammatory responses and parasite burdens to RELMα+/+ mice. In contrast, both RELMα-/- and BM RELMα-/- mice exhibited increased Nb-induced lung and intestinal inflammation, correlated with elevated Th2 cytokines and Nb killing. CD11c+ lung macrophages were the dominant BM-derived source of RELMα and can mediate Nb killing. Therefore, we employed a macrophage-worm co-culture system to investigate whether RELMα regulates macrophage-mediated Nb killing. Compared to RELMα+/+ macrophages, RELMα-/- macrophages exhibited increased binding to Nb and functionally impaired Nb development. Supplementation with recombinant RELMα partially reversed this phenotype. Gene expression analysis revealed that RELMα decreased cell adhesion and Fc receptor signaling pathways, which are associated with macrophage-mediated helminth killing. Collectively, these studies demonstrate that BM-derived RELMα is necessary and sufficient to dampen Nb immune responses, and identify that one mechanism of action of RELMα is through inhibiting macrophage recruitment and interaction with Nb. Our findings suggest that RELMα acts as an immune brake that provides mutually beneficial effects for the host and parasite by limiting tissue damage and delaying parasite expulsion.


Subject(s)
Intercellular Signaling Peptides and Proteins/physiology , Strongylida Infections/immunology , Adenosine Triphosphate/metabolism , Alveolar Epithelial Cells/metabolism , Animals , Cell Adhesion , Cells, Cultured , Coculture Techniques , Dendritic Cells/metabolism , Female , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Macrophages, Alveolar/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nippostrongylus/isolation & purification , Nippostrongylus/ultrastructure , Radiation Chimera , Recombinant Proteins/metabolism , Strongylida Infections/parasitology , Th2 Cells/immunology
4.
ASN Neuro ; 10: 1759091418782304, 2018.
Article in English | MEDLINE | ID: mdl-30016877

ABSTRACT

Continuous exposure to aerosolized fine (particle size ≤2.5 µm) and ultrafine (particle size ≤0.1 µm) particulates can trigger innate inflammatory responses in the lung and brain depending on particle composition. Most studies of manmade toxicants use inhalation exposure routes, whereas most studies of allergens use soluble solutions administered via intranasal or injection routes. Here, we tested whether continuous inhalation exposure to aerosolized Alternaria alternata particulates (a common fungal allergen associated with asthma) would induce innate inflammatory responses in the lung and brain. By designing a new environmental chamber able to control particle size distribution and mass concentration, we continuously exposed adult mice to aerosolized ultrafine Alternaria particulates for 96 hr. Despite induction of innate immune responses in the lung, induction of innate immune responses in whole brain samples was not detected by quantitative polymerase chain reaction or flow cytometry. However, exposure did trigger decreases in Arginase 1, inducible nitric oxide synthase, and tumor necrosis factor alpha mRNA in the brainstem samples containing the central nervous system respiratory circuit (the dorsal respiratory group, ventral respiratory group, and the pre-Bötzinger and Bötzinger complexes). In addition, a significant decrease in the percentage of Toll-like receptor 2-expressing brainstem microglia was detected by flow cytometry. Histologic analysis revealed a significant decrease in Iba1 but not glial fibrillary acidic protein immunoreactivity in both the brainstem and the hippocampus. Together these data indicate that inhalation exposure to a natural fungal allergen under conditions sufficient to induce lung inflammation surprisingly causes reductions in baseline expression of select innate immune molecules (similar to that observed during endotoxin tolerance) in the region of the central nervous system controlling respiration.


Subject(s)
Allergens/toxicity , Brain Stem/metabolism , Fungi/chemistry , Immunity, Innate/physiology , Pneumonia/etiology , Pneumonia/pathology , Animals , Antigens, CD/metabolism , Arginase/metabolism , Disease Models, Animal , Inhalation Exposure , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , NADPH Oxidase 2/metabolism , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/metabolism
5.
Proc Natl Acad Sci U S A ; 114(48): E10399-E10408, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29133417

ABSTRACT

Helminths trigger multiple immunomodulatory pathways that can protect from sepsis. Human resistin (hRetn) is an immune cell-derived protein that is highly elevated in helminth infection and sepsis. However, the function of hRetn in sepsis, or whether hRetn influences helminth protection against sepsis, is unknown. Employing hRetn-expressing transgenic mice (hRETNTg+) and recombinant hRetn, we identify a therapeutic function for hRetn in lipopolysaccharide (LPS)-induced septic shock. hRetn promoted helminth-induced immunomodulation, with increased survival of Nippostrongylus brasiliensis (Nb)-infected hRETNTg+ mice after a fatal LPS dose compared with naive mice or Nb-infected hRETNTg- mice. Employing immunoprecipitation assays, hRETNTg+Tlr4-/- mice, and human immune cell culture, we demonstrate that hRetn binds the LPS receptor Toll-like receptor 4 (TLR4) through its N terminal and modulates STAT3 and TBK1 signaling, triggering a switch from proinflammatory to anti-inflammatory responses. Further, we generate hRetn N-terminal peptides that are able to block LPS proinflammatory function. Together, our studies identify a critical role for hRetn in blocking LPS function with important clinical significance in helminth-induced immunomodulation and sepsis.


Subject(s)
Lipopolysaccharides/metabolism , Resistin/immunology , Shock, Septic/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Animals , Biological Therapy/methods , Disease Models, Animal , Female , Gram-Negative Bacteria/immunology , Gram-Negative Bacteria/metabolism , Humans , Lipopolysaccharide Receptors/immunology , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nippostrongylus/immunology , Protective Agents , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , STAT3 Transcription Factor/metabolism , Shock, Septic/microbiology , Shock, Septic/therapy , Signal Transduction/immunology
6.
Front Immunol ; 8: 335, 2017.
Article in English | MEDLINE | ID: mdl-28424687

ABSTRACT

During chronic infection, memory T cells acquire a unique phenotype and become dependent on different survival signals than those needed for memory T cells generated during an acute infection. The distinction between the role of effector and memory T cells in an environment of persistent antigen remains unclear. Here, in the context of chronic Toxoplasma gondii infection, we demonstrate that a population of CD8 T cells exhibiting a tissue-resident memory (TRM) phenotype accumulates within the brain. We show that this population is distributed throughout the brain in both parenchymal and extraparenchymal spaces. Furthermore, this population is transcriptionally distinct and exhibits a transcriptional signature consistent with the TRM observed in acute viral infections. Finally, we establish that the CD103+ TRM population has an intrinsic capacity to produce both IFN-γ and TNF-α, cytokines critical for parasite control within the central nervous system (CNS). The contribution of this population to pro-inflammatory cytokine production suggests an important role for TRM in protective and ongoing immune responses in the infected CNS. Accession number: GSE95105.

7.
Infect Immun ; 84(4): 1100-1111, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26831469

ABSTRACT

Resistin-like molecules (RELMs) are highly expressed following helminth infection, where they impact both the host and helminth. While RELMα (Retnla) impairs helminth expulsion by inhibiting protective Th2 immunity, RELMß (Retnlb) can promote its expulsion. We employed Retnla(-/-) and Retnlb(-/-) mice to delineate the function of both proteins following infection with Nippostrongylus brasiliensis, a hookworm that infects the lung and intestine. Whereas wild-type (WT) and Retnlb(-/-)mice exhibited equivalent infection-induced inflammation, Retnla(-/-) mice suffered a heightened inflammatory response, including increased mortality, weight loss, and lung inflammation. In the intestine, Retnla(-/-)mice had low parasite egg burdens compared to those of WT mice, while Retnlb(-/-) mice exhibited high egg burdens, suggesting that RELMα and RELMß have functionally distinct effects on immunity and inflammation to N. brasiliensis To test the importance of both proteins, we generated Retnla(-/-) Retnlb(-/-) mice. Infected Retnla(-/-)Retnlb(-/-) mice exhibited similar responses to Retnla(-/-) mice, including increased mortality and lung inflammation. This inflammatory response in Retnla(-/-) Retnlb(-/-) mice negatively impacted N. brasiliensis fitness, as demonstrated by significantly lower worm ATP levels and decreased intestinal worm burden and fecundity. Lung cytokine analysis revealed that Retnla(-/-) and Retnla(-/-) Retnlb(-/-) mice expressed significantly increased levels of interleukin-4 (IL-4). Finally, we generated Retnla(-/-) mice on the Rag(-/-) background and observed that the effects of RELMα were abrogated in the absence of adaptive immunity. Together, these data demonstrate that RELMα but not RELMß significantly impacts the immune response toN. brasiliensis infection by downregulating the Th2 adaptive immune response in the lung, which protects the host but allows improved parasite fitness.


Subject(s)
Gene Expression Regulation/physiology , Hormones, Ectopic/metabolism , Inflammation/parasitology , Intercellular Signaling Peptides and Proteins/metabolism , Strongylida Infections/metabolism , Animals , CD4-Positive T-Lymphocytes , Down-Regulation , Hormones, Ectopic/genetics , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Lung Diseases/metabolism , Lung Diseases/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nippostrongylus , Strongylida Infections/genetics , Strongylida Infections/parasitology
8.
PLoS Pathog ; 11(1): e1004579, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25568944

ABSTRACT

Parasitic helminth infections can be associated with lifelong morbidity such as immune-mediated organ failure. A better understanding of the host immune response to helminths could provide new avenues to promote parasite clearance and/or alleviate infection-associated morbidity. Murine resistin-like molecules (RELM) exhibit pleiotropic functions following helminth infection including modulating the host immune response; however, the relevance of human RELM proteins in helminth infection is unknown. To examine the function of human resistin (hResistin), we utilized transgenic mice expressing the human resistin gene (hRetnTg+). Following infection with the helminth Nippostrongylus brasiliensis (Nb), hResistin expression was significantly upregulated in infected tissue. Compared to control hRetnTg- mice, hRetnTg+ mice suffered from exacerbated Nb-induced inflammation characterized by weight loss and increased infiltration of inflammatory monocytes in the lung, along with elevated Nb egg burdens and delayed parasite expulsion. Genome-wide transcriptional profiling of the infected tissue revealed that hResistin promoted expression of proinflammatory cytokines and genes downstream of toll-like receptor signaling. Moreover, hResistin preferentially bound lung monocytes, and exogenous treatment of mice with recombinant hResistin promoted monocyte recruitment and proinflammatory cytokine expression. In human studies, increased serum resistin was associated with higher parasite load in individuals infected with soil-transmitted helminths or filarial nematode Wuchereria bancrofti, and was positively correlated with proinflammatory cytokines. Together, these studies identify human resistin as a detrimental factor induced by multiple helminth infections, where it promotes proinflammatory cytokines and impedes parasite clearance. Targeting the resistin/proinflammatory cytokine immune axis may provide new diagnostic or treatment strategies for helminth infection and associated immune-mediated pathology.


Subject(s)
Helminthiasis , Inflammation Mediators/metabolism , Macrophages/metabolism , Monocytes/metabolism , Parasitemia/genetics , Resistin/genetics , Animals , Cytokines/metabolism , Female , Helminthiasis/genetics , Helminthiasis/immunology , Helminthiasis/metabolism , Helminthiasis/parasitology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/parasitology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/immunology , Nippostrongylus/immunology , Parasitemia/immunology , Parasitemia/metabolism , Rats , Rats, Sprague-Dawley , Resistin/metabolism , Strongylida Infections/genetics , Strongylida Infections/immunology , Strongylida Infections/metabolism , Strongylida Infections/parasitology , Up-Regulation/genetics
9.
Gut Microbes ; 5(3): 363-8, 2014.
Article in English | MEDLINE | ID: mdl-24831469

ABSTRACT

Citrobacter rodentium infection is a murine model of pathogenic Escherichia coli infection that allows investigation of the cellular and molecular mechanisms involved in host-protective immunity and bacterial-induced intestinal inflammation. We recently demonstrated that following C. rodentium infection, the absence of Resistin-Like Molecule (RELM) α resulted in attenuated Th17 cell responses and reduced intestinal inflammation with minimal effects on bacterial clearance. In this addendum, we investigated the cytokine modulatory effects of RELMα and RELMα expression in the intestinal mucosa following C. rodentium infection. We show that in addition to promoting Th17 cytokine responses, RELMα inhibits Th2 cytokine expression and Th2-cytokine effector macrophage responses in the C. rodentium-infected colons. Second, utilizing reporter C. rodentium, we examined RELMα expression and macrophage recruitment at the host pathogen interface. We observed infection-induced macrophage infiltration and RELMα expression by intestinal epithelial cells. The influence of infection-induced RELMα on macrophage recruitment in the intestine is discussed.


Subject(s)
Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Th17 Cells/immunology , Animals , Colon/pathology , Cytokines/metabolism , Disease Models, Animal , Enterobacteriaceae Infections/microbiology , Gene Expression Profiling , Intercellular Signaling Peptides and Proteins/genetics , Macrophages/immunology , Mice, Inbred C57BL
10.
Curr Immunol Rev ; 9(3): 147-156, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-24772059

ABSTRACT

The role of macrophages in homeostatic conditions and the immune system range from clearing debris to recognizing and killing pathogens. While classically activated macrophages (CAMacs) are induced by T helper type 1 (Th1) cytokines and exhibit microbicidal properties, Th2 cytokines promote alternative activation of macrophages (AAMacs). AAMacs contribute to the killing of helminth parasites and mediate additional host-protective processes such as regulating inflammation and wound healing. Yet, other parasites susceptible to Th1 type responses can exploit alternative activation of macrophages to diminish Th1 immune responses and prolong infection. In this review, we will delineate the factors that mediate alternative activation (e.g. Th2 cytokines and chitin) and the resulting downstream signaling events (e.g. STAT6 signaling). Next, the specific AAMac-derived factors (e.g. Arginase1) that contribute to resistance or susceptibility to parasitic infections will be summarized. Finally, we will conclude with the discussion of additional AAMac functions beyond immunity to parasites, including the regulation of inflammation, wound healing and the regulation of metabolic disorders.

SELECTION OF CITATIONS
SEARCH DETAIL
...