Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Science ; 381(6665): 1461-1467, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37769084

ABSTRACT

Candida auris is an emerging fungal pathogen responsible for health care-associated outbreaks that arise from persistent surface and skin colonization. We characterized the arsenal of adhesins used by C. auris and discovered an uncharacterized adhesin, Surface Colonization Factor (Scf1), and a conserved adhesin, Iff4109, that are essential for the colonization of inert surfaces and mammalian hosts. SCF1 is apparently specific to C. auris, and its expression mediates adhesion to inert and biological surfaces across isolates from all five clades. Unlike canonical fungal adhesins, which function through hydrophobic interactions, Scf1 relies on exposed cationic residues for surface association. SCF1 is required for C. auris biofilm formation, skin colonization, virulence in systemic infection, and colonization of inserted medical devices.


Subject(s)
Candida auris , Candidiasis, Invasive , Fungal Proteins , Microfilament Proteins , Animals , Humans , Candida auris/genetics , Candida auris/pathogenicity , Virulence , Candidiasis, Invasive/microbiology , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Protein Domains , Hydrophobic and Hydrophilic Interactions , Mice
2.
PLoS Biol ; 21(5): e3001822, 2023 05.
Article in English | MEDLINE | ID: mdl-37205709

ABSTRACT

Candida albicans is a frequent colonizer of human mucosal surfaces as well as an opportunistic pathogen. C. albicans is remarkably versatile in its ability to colonize diverse host sites with differences in oxygen and nutrient availability, pH, immune responses, and resident microbes, among other cues. It is unclear how the genetic background of a commensal colonizing population can influence the shift to pathogenicity. Therefore, we examined 910 commensal isolates from 35 healthy donors to identify host niche-specific adaptations. We demonstrate that healthy people are reservoirs for genotypically and phenotypically diverse C. albicans strains. Using limited diversity exploitation, we identified a single nucleotide change in the uncharacterized ZMS1 transcription factor that was sufficient to drive hyper invasion into agar. We found that SC5314 was significantly different from the majority of both commensal and bloodstream isolates in its ability to induce host cell death. However, our commensal strains retained the capacity to cause disease in the Galleria model of systemic infection, including outcompeting the SC5314 reference strain during systemic competition assays. This study provides a global view of commensal strain variation and within-host strain diversity of C. albicans and suggests that selection for commensalism in humans does not result in a fitness cost for invasive disease.


Subject(s)
Candida albicans , Symbiosis , Humans , Candida albicans/genetics , Transcription Factors/genetics , Gene Expression Regulation
3.
Nat Commun ; 12(1): 4939, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34400627

ABSTRACT

Pain is a central feature of soft tissue trauma, which under certain contexts, results in aberrant osteochondral differentiation of tissue-specific stem cells. Here, the role of sensory nerve fibers in this abnormal cell fate decision is investigated using a severe extremity injury model in mice. Soft tissue trauma results in NGF (Nerve growth factor) expression, particularly within perivascular cell types. Consequently, NGF-responsive axonal invasion occurs which precedes osteocartilaginous differentiation. Surgical denervation impedes axonal ingrowth, with significant delays in cartilage and bone formation. Likewise, either deletion of Ngf or two complementary methods to inhibit its receptor TrkA (Tropomyosin receptor kinase A) lead to similar delays in axonal invasion and osteochondral differentiation. Mechanistically, single-cell sequencing suggests a shift from TGFß to FGF signaling activation among pre-chondrogenic cells after denervation. Finally, analysis of human pathologic specimens and databases confirms the relevance of NGF-TrkA signaling in human disease. In sum, NGF-mediated TrkA-expressing axonal ingrowth drives abnormal osteochondral differentiation after soft tissue trauma. NGF-TrkA signaling inhibition may have dual therapeutic use in soft tissue trauma, both as an analgesic and negative regulator of aberrant stem cell differentiation.


Subject(s)
Cell Differentiation , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Signal Transduction , Wounds and Injuries/metabolism , Animals , Axons/metabolism , Cartilage/metabolism , Mice , Mice, Inbred C57BL , Nerve Growth Factor/genetics , Osteogenesis , Stem Cells/metabolism , Wounds and Injuries/pathology
4.
Stem Cell Reports ; 16(3): 626-640, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33606989

ABSTRACT

Heterotopic ossification (HO) is a form of pathological cell-fate change of mesenchymal stem/precursor cells (MSCs) that occurs following traumatic injury, limiting range of motion in extremities and causing pain. MSCs have been shown to differentiate to form bone; however, their lineage and aberrant processes after trauma are not well understood. Utilizing a well-established mouse HO model and inducible lineage-tracing mouse (Hoxa11-CreERT2;ROSA26-LSL-TdTomato), we found that Hoxa11-lineage cells represent HO progenitors specifically in the zeugopod. Bioinformatic single-cell transcriptomic and epigenomic analyses showed Hoxa11-lineage cells are regionally restricted mesenchymal cells that, after injury, gain the potential to undergo differentiation toward chondrocytes, osteoblasts, and adipocytes. This study identifies Hoxa11-lineage cells as zeugopod-specific ectopic bone progenitors and elucidates the fate specification and multipotency that mesenchymal cells acquire after injury. Furthermore, this highlights homeobox patterning genes as useful tools to trace region-specific progenitors and enable location-specific gene deletion.


Subject(s)
Bone and Bones/metabolism , Cell Differentiation , Cell Lineage , Mesenchymal Stem Cells/metabolism , Ossification, Heterotopic/genetics , Ossification, Heterotopic/metabolism , Osteogenesis , Adipocytes/metabolism , Animals , Chondrocytes/metabolism , Disease Models, Animal , Ectopic Gene Expression , Epigenomics , Female , Gene Expression Profiling , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Ossification, Heterotopic/pathology , Osteoblasts/metabolism , Single-Cell Analysis , Tendons/metabolism
5.
FASEB J ; 34(12): 15753-15770, 2020 12.
Article in English | MEDLINE | ID: mdl-33089917

ABSTRACT

Ischemia reperfusion (IR) injury results in devastating skeletal muscle fibrosis. Here, we recapitulate this injury with a mouse model of hindlimb IR injury which leads to skeletal muscle fibrosis. Injury resulted in extensive immune infiltration with robust neutrophil extracellular trap (NET) formation in the skeletal muscle, however, direct targeting of NETs via the peptidylarginine deiminase 4 (PAD4) mechanism was insufficient to reduce muscle fibrosis. Circulating levels of IL-10 and TNFα were significantly elevated post injury, indicating toll-like receptor (TLR) signaling may be involved in muscle injury. Administration of hydroxychloroquine (HCQ), a small molecule inhibitor of TLR7/8/9, following injury reduced NET formation, IL-10, and TNFα levels and ultimately mitigated muscle fibrosis and improved myofiber regeneration following IR injury. HCQ treatment decreased fibroadipogenic progenitor cell proliferation and partially inhibited ERK1/2 phosphorylation in the injured tissue, suggesting it may act through a combination of TLR7/8/9 and ERK signaling mechanisms. We demonstrate that treatment with FDA-approved HCQ leads to decreased muscle fibrosis and increased myofiber regeneration following IR injury, suggesting short-term HCQ treatment may be a viable treatment to prevent muscle fibrosis in ischemia reperfusion and traumatic extremity injury.


Subject(s)
Extracellular Traps/metabolism , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Neutrophils/metabolism , Reperfusion Injury/metabolism , Signal Transduction/physiology , Toll-Like Receptors/metabolism , Animals , Cell Proliferation/physiology , Disease Models, Animal , Fibrosis/metabolism , Interleukin-10/metabolism , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Protein-Arginine Deiminase Type 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
J Clin Invest ; 130(10): 5444-5460, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32673290

ABSTRACT

Cells sense the extracellular environment and mechanical stimuli and translate these signals into intracellular responses through mechanotransduction, which alters cell maintenance, proliferation, and differentiation. Here we use a mouse model of trauma-induced heterotopic ossification (HO) to examine how cell-extrinsic forces impact mesenchymal progenitor cell (MPC) fate. After injury, single-cell (sc) RNA sequencing of the injury site reveals an early increase in MPC genes associated with pathways of cell adhesion and ECM-receptor interactions, and MPC trajectories to cartilage and bone. Immunostaining uncovers active mechanotransduction after injury with increased focal adhesion kinase signaling and nuclear translocation of transcriptional coactivator TAZ, inhibition of which mitigates HO. Similarly, joint immobilization decreases mechanotransductive signaling, and completely inhibits HO. Joint immobilization decreases collagen alignment and increases adipogenesis. Further, scRNA sequencing of the HO site after injury with or without immobilization identifies gene signatures in mobile MPCs correlating with osteogenesis, and signatures from immobile MPCs with adipogenesis. scATAC-seq in these same MPCs confirm that in mobile MPCs, chromatin regions around osteogenic genes are open, whereas in immobile MPCs, regions around adipogenic genes are open. Together these data suggest that joint immobilization after injury results in decreased ECM alignment, altered MPC mechanotransduction, and changes in genomic architecture favoring adipogenesis over osteogenesis, resulting in decreased formation of HO.


Subject(s)
Extremities/injuries , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/physiology , Ossification, Heterotopic/etiology , Restraint, Physical , Acyltransferases , Adipogenesis/genetics , Animals , Cell Differentiation , Cell Lineage , Disease Models, Animal , Extracellular Matrix/metabolism , Focal Adhesion Kinase 1/deficiency , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Humans , Male , Mechanotransduction, Cellular/genetics , Mechanotransduction, Cellular/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ossification, Heterotopic/pathology , Ossification, Heterotopic/physiopathology , Osteogenesis/genetics , Restraint, Physical/adverse effects , Restraint, Physical/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
7.
J Immunol ; 204(8): 2203-2215, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32161098

ABSTRACT

Myeloid cells are critical to the development of fibrosis following muscle injury; however, the mechanism of their role in fibrosis formation remains unclear. In this study, we demonstrate that myeloid cell-derived TGF-ß1 signaling is increased in a profibrotic ischemia reperfusion and cardiotoxin muscle injury model. We found that myeloid-specific deletion of Tgfb1 abrogates the fibrotic response in this injury model and reduces fibro/adipogenic progenitor cell proliferation while simultaneously enhancing muscle regeneration, which is abrogated by adaptive transfer of normal macrophages. Similarly, a murine TGFBRII-Fc ligand trap administered after injury significantly reduced muscle fibrosis and improved muscle regeneration. This study ultimately demonstrates that infiltrating myeloid cell TGF-ß1 is responsible for the development of traumatic muscle fibrosis, and its blockade offers a promising therapeutic target for preventing muscle fibrosis after ischemic injury.


Subject(s)
Fibrosis/immunology , Fibrosis/pathology , Macrophages/immunology , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Myeloid Cells/immunology , Transforming Growth Factor beta1/immunology , Animals , Cardiotoxins , Fibrosis/complications , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/pathology , Phenotype , Reperfusion Injury/chemically induced , Reperfusion Injury/complications , Reperfusion Injury/immunology
8.
Nat Commun ; 11(1): 722, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32024825

ABSTRACT

Heterotopic ossification (HO) is an aberrant regenerative process with ectopic bone induction in response to musculoskeletal trauma, in which mesenchymal stem cells (MSC) differentiate into osteochondrogenic cells instead of myocytes or tenocytes. Despite frequent cases of hospitalized musculoskeletal trauma, the inflammatory responses and cell population dynamics that regulate subsequent wound healing and tissue regeneration are still unclear. Here we examine, using a mouse model of trauma-induced HO, the local microenvironment of the initial post-injury inflammatory response. Single cell transcriptome analyses identify distinct monocyte/macrophage populations at the injury site, with their dynamic changes over time elucidated using trajectory analyses. Mechanistically, transforming growth factor beta-1 (TGFß1)-producing monocytes/macrophages are associated with HO and aberrant chondrogenic progenitor cell differentiation, while CD47-activating peptides that reduce systemic macrophage TGFß levels and help ameliorate HO. Our data thus implicate CD47 activation as a therapeutic approach for modulating monocyte/macrophage phenotypes, MSC differentiation and HO formation during wound healing.


Subject(s)
Burns/pathology , Monocytes/pathology , Ossification, Heterotopic/pathology , Wound Healing/physiology , Animals , CD47 Antigen/metabolism , Cell Differentiation , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation , Macrophages/pathology , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology , Mice, Inbred C57BL , Mice, Transgenic , Peptides/pharmacology , Phagocytosis , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
9.
Bone Res ; 7: 36, 2019.
Article in English | MEDLINE | ID: mdl-31840004

ABSTRACT

Heterotopic ossification (HO) is a debilitating condition characterized by the pathologic formation of ectopic bone. HO occurs commonly following orthopedic surgeries, burns, and neurologic injuries. While surgical excision may provide palliation, the procedure is often burdened with significant intra-operative blood loss due to a more robust contribution of blood supply to the pathologic bone than to native bone. Based on these clinical observations, we set out to examine the role of vascular signaling in HO. Vascular endothelial growth factor A (VEGFA) has previously been shown to be a crucial pro-angiogenic and pro-osteogenic cue during normal bone development and homeostasis. Our findings, using a validated mouse model of HO, demonstrate that HO lesions are highly vascular, and that VEGFA is critical to ectopic bone formation, despite lacking a contribution of endothelial cells within the developing anlagen.

10.
Am J Pathol ; 188(11): 2464-2473, 2018 11.
Article in English | MEDLINE | ID: mdl-30142335

ABSTRACT

Heterotopic ossification (HO) occurs secondary to trauma, causing pain and functional limitations. Identification of the cells that contribute to HO is critical to the development of therapies. Given that innate immune cells and mesenchymal stem cells are known contributors to HO, we sought to define the contribution of these populations to HO and to identify what, if any, contribution circulating populations have to HO. A shared circulation was obtained using a parabiosis model, established between an enhanced green fluorescent protein-positive/luciferase+ donor and a same-strain nonreporter recipient mouse. The nonreporter mouse received Achilles tendon transection and dorsal burn injury to induce HO formation. Bioluminescence imaging and immunostaining were performed to define the circulatory contribution of immune and mesenchymal cell populations. Histologic analysis showed circulating cells present throughout each stage of the developing HO anlagen. Circulating cells were present at the injury site during the inflammatory phase and proliferative period, with diminished contribution in mature HO. Immunostaining demonstrated that most early circulatory cells were from the innate immune system; only a small population of mesenchymal cells were present in the HO. We demonstrate the time course of the participation of circulatory cells in trauma-induced HO and identify populations of circulating cells present in different stages of HO. These findings further elucidate the relative contribution of local and systemic cell populations to HO.


Subject(s)
Burns/complications , Disease Models, Animal , Inflammation/pathology , Mesenchymal Stem Cells/pathology , Ossification, Heterotopic/pathology , Animals , Female , Inflammation/blood , Inflammation/etiology , Mice , Mice, Inbred C57BL , Ossification, Heterotopic/blood , Ossification, Heterotopic/etiology , Osteogenesis , Signal Transduction
11.
Microb Ecol ; 72(3): 621-32, 2016 10.
Article in English | MEDLINE | ID: mdl-27341838

ABSTRACT

All plants form symbioses with endophytic fungi, which affect host plant health and function. Most endophytic fungi are horizontally transmitted, and consequently, local environment and geographic location greatly influence endophyte community composition. Growing evidence also suggests that identity of the plant host (e.g., species, genotype) can be important in shaping endophyte communities. However, little is known about how disturbances to plants affect their fungal symbiont communities. The goal of this study was to test if disturbances, from both natural and anthropogenic sources, can alter endophyte communities independent of geographic location or plant host identity. Using the plant species white snakeroot (Ageratina altissima; Asteraceae), we conducted two experiments that tested the effect of perturbation on endophyte communities. First, we examined endophyte response to leaf mining insect activity, a natural perturbation, in three replicate populations. Second, for one population, we applied fungicide to plant leaves to test endophyte community response to an anthropogenic perturbation. Using culture-based methods and Sanger sequencing of fungal isolates, we then examined abundance, diversity, and community structure of endophytic fungi in leaves subjected to perturbations by leaf mining and fungicide application. Our results show that plant host individual and geographic location are the major determinants of endophyte community composition even in the face of perturbations. Unexpectedly, we found that leaf mining did not impact endophyte communities in white snakeroot, but fungicide treatment resulted in small but significant changes in endophyte community structure. Together, our results suggest that endophyte communities are highly resistant to biotic and anthropogenic disturbances.


Subject(s)
Endophytes , Face/microbiology , Fungi/classification , Herbivory , Plants/classification , Plants/microbiology , Symbiosis , Animals , Biodiversity , Endophytes/classification , Endophytes/drug effects , Endophytes/isolation & purification , Fungi/drug effects , Fungi/isolation & purification , Indiana , Insecta/microbiology , Microbiota/physiology , Mining , Pesticides , Phylogeny , Plant Leaves/microbiology , Soil Microbiology , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...