Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ana M. Misic is active.

Publication


Featured researches published by Ana M. Misic.


Nature | 2016

Glial-cell-derived neuroregulators control type 3 innate lymphoid cells and gut defence

Sales Ibiza; Bethania García-Cassani; Hélder Ribeiro; Tânia Carvalho; Luís Pereira de Almeida; Rute Marques; Ana M. Misic; Casey Bartow-McKenney; Denise M. Larson; William J. Pavan; Gérard Eberl; Elizabeth A. Grice; Henrique Veiga-Fernandes

Group 3 innate lymphoid cells (ILC3) are major regulators of inflammation and infection at mucosal barriers. ILC3 development is thought to be programmed, but how ILC3 perceive, integrate and respond to local environmental signals remains unclear. Here we show that ILC3 in mice sense their environment and control gut defence as part of a glial–ILC3–epithelial cell unit orchestrated by neurotrophic factors. We found that enteric ILC3 express the neuroregulatory receptor RET. ILC3-autonomous Ret ablation led to decreased innate interleukin-22 (IL-22), impaired epithelial reactivity, dysbiosis and increased susceptibility to bowel inflammation and infection. Neurotrophic factors directly controlled innate Il22 downstream of the p38 MAPK/ERK-AKT cascade and STAT3 activation. Notably, ILC3 were adjacent to neurotrophic-factor-expressing glial cells that exhibited stellate-shaped projections into ILC3 aggregates. Glial cells sensed microenvironmental cues in a MYD88-dependent manner to control neurotrophic factors and innate IL-22. Accordingly, glial-intrinsic Myd88 deletion led to impaired production of ILC3-derived IL-22 and a pronounced propensity towards gut inflammation and infection. Our work sheds light on a novel multi-tissue defence unit, revealing that glial cells are central hubs of neuron and innate immune regulation by neurotrophic factor signals.


Mbio | 2015

The shared microbiota of humans and companion animals as evaluated from Staphylococcus carriage sites

Ana M. Misic; Meghan F. Davis; Amanda S. Tyldsley; Brendan P. Hodkinson; Pam Tolomeo; Baofeng Hu; Irving Nachamkin; Ebbing Lautenbach; Daniel O. Morris; Elizabeth A. Grice

BackgroundStaphylococcus aureus and other coagulase-positive staphylococci (CPS) colonize skin and mucous membrane sites and can cause skin and soft tissue infections (SSTIs) in humans and animals. Factors modulating methicillin-resistant S. aureus (MRSA) colonization and infection in humans remain unclear, including the role of the greater microbial community and environmental factors such as contact with companion animals. In the context of a parent study evaluating the households of outpatients with community MRSA SSTI, the objectives of this study were 1) to characterize the microbiota that colonizes typical coagulase-positive Staphylococcus spp. carriage sites in humans and their companion pets, 2) to analyze associations between Staphylococcus infection and carriage and the composition and diversity of microbial communities, and 3) to analyze factors that influence sharing of microbiota between pets and humans.ResultsWe enrolled 25 households containing 56 pets and 30 humans. Sampling locations were matched to anatomical sites cultured by the parent study for MRSA and other CPS. Bacterial microbiota were characterized by sequencing of 16S ribosomal RNA genes. Household membership was strongly associated with microbial communities, in both humans and pets. Pets were colonized with a greater relative abundance of Proteobacteria, whereas people were colonized with greater relative abundances of Firmicutes and Actinobacteria. We did not detect differences in microbiota associated with MRSA SSTI, or carriage of MRSA, S. aureus or CPS. Humans in households without pets were more similar to each other than humans in pet-owning households, suggesting that companion animals may play a role in microbial transfer. We examined changes in microbiota over a 3-month time period and found that pet staphylococcal carriage sites were more stable than human carriage sites.ConclusionsWe characterized and identified patterns of microbiota sharing and stability between humans and companion animals. While we did not detect associations with MRSA SSTI, or carriage of MRSA, S. aureus or CPS in this small sample size, larger studies are warranted to fully explore how microbial communities may be associated with and contribute to MRSA and/or CPS colonization, infection, and recurrence.


Scientific Reports | 2017

Stress during pregnancy alters temporal and spatial dynamics of the maternal and offspring microbiome in a sex-specific manner

Eldin Jašarević; Christopher D. Howard; Ana M. Misic; Daniel P. Beiting; Tracy L. Bale

The microbiome is a regulator of host immunity, metabolism, neurodevelopment, and behavior. During early life, bacterial communities within maternal gut and vaginal compartments can have an impact on directing these processes. Maternal stress experience during pregnancy may impact offspring development by altering the temporal and spatial dynamics of the maternal microbiome during pregnancy. To examine the hypothesis that maternal stress disrupts gut and vaginal microbial dynamics during critical prenatal and postnatal windows, we used high-resolution 16S rRNA marker gene sequencing to examine outcomes in our mouse model of early prenatal stress. Consistent with predictions, maternal fecal communities shift across pregnancy, a process that is disrupted by stress. Vaginal bacterial community structure and composition exhibit lasting disruption following stress exposure. Comparison of maternal and offspring microbiota revealed that similarities in bacterial community composition was predicted by a complex interaction between maternal body niche and offspring age and sex. Importantly, early prenatal stress influenced offspring bacterial community assembly in a temporal and sex-specific manner. Taken together, our results demonstrate that early prenatal stress may influence offspring development through converging modifications to gut microbial composition during pregnancy and transmission of dysbiotic vaginal microbiome at birth.


Genome Announcements | 2015

Complete Genome Sequence and Methylome of Staphylococcus schleiferi, an Important Cause of Skin and Ear Infections in Veterinary Medicine.

Ana M. Misic; Christine L. Cain; Daniel O. Morris; Shelley C. Rankin; Daniel P. Beiting

ABSTRACT Staphylococcus schleiferi, a Gram-positive and coagulase-variable organism, is an opportunistic human pathogen and a major cause of skin and soft tissue infections in dogs. Here, we report the first S. schleiferi genome sequence and methylome from four canine clinical isolates.


mSphere | 2016

Divergent Isoprenoid Biosynthesis Pathways in Staphylococcus Species Constitute a Drug Target for Treating Infections in Companion Animals

Ana M. Misic; Christine L. Cain; Daniel O. Morris; Shelley C. Rankin; Daniel P. Beiting

Drug-resistant Staphylococcus species are a major concern in human and veterinary medicine. There is a need for new antibiotics that exhibit a selective effect in treating infections in companion and livestock animals and that would not be used to treat human bacterial infections. We have identified fosmidomycin as an antibiotic that selectively targets certain Staphylococcus species that are often encountered in skin infections in cats and dogs. These findings expand our understanding of Staphylococcus evolution and may have direct implications for treating staphylococcal infections in veterinary medicine. ABSTRACT Staphylococcus species are a leading cause of skin and soft tissue infections in humans and animals, and the antibiotics used to treat these infections are often the same. Methicillin- and multidrug-resistant staphylococcal infections are becoming more common in human and veterinary medicine. From a “One Health” perspective, this overlap in antibiotic use and resistance raises concerns over the potential spread of antibiotic resistance genes. Whole-genome sequencing and comparative genomics analysis revealed that Staphylococcus species use divergent pathways to synthesize isoprenoids. Species frequently associated with skin and soft tissue infections in companion animals, including S. schleiferi and S. pseudintermedius, use the nonmevalonate pathway. In contrast, S. aureus, S. epidermidis, and S. lugdunensis use the mevalonate pathway. The antibiotic fosmidomycin, an inhibitor of the nonmevalonate pathway, was effective in killing canine clinical staphylococcal isolates but had no effect on the growth or survival of S. aureus and S. epidermidis. These data identify an essential metabolic pathway in Staphylococcus that differs among members of this genus and suggest that drugs such as fosmidomycin, which targets enzymes in the nonmevalonate pathway, may be an effective treatment for certain staphylococcal infections. IMPORTANCE Drug-resistant Staphylococcus species are a major concern in human and veterinary medicine. There is a need for new antibiotics that exhibit a selective effect in treating infections in companion and livestock animals and that would not be used to treat human bacterial infections. We have identified fosmidomycin as an antibiotic that selectively targets certain Staphylococcus species that are often encountered in skin infections in cats and dogs. These findings expand our understanding of Staphylococcus evolution and may have direct implications for treating staphylococcal infections in veterinary medicine.


Veterinary Microbiology | 2015

Genome sequencing reveals strain dynamics of methicillin-resistant Staphylococcus aureus in the same household in the context of clinical disease in a person and a dog

Meghan F. Davis; Ana M. Misic; Daniel O. Morris; John T. Moss; Pam Tolomeo; Daniel P. Beiting; Irving Nachamkin; Ebbing Lautenbach; Shelley C. Rankin

The strain dynamics of methicillin-resistant Staphylococcus aureus (MRSA) isolates from people and the household dog were investigated. The isolates were identified in the context of a randomized controlled trial that tested household-wide decolonization of people. Genotypic comparison of MRSA isolates obtained from two household members, the dog, and home surfaces over a three-month period failed to implicate the pet or the home environment in recurrent colonization of the household members. However, it did implicate the pets bed in exposure of the dog prior to the dogs infection. Whole genome sequencing was performed to differentiate the isolates. This report also describes introduction of diverse strains of MRSA into the household within six weeks of cessation of harmonized decolonization treatment of people and treatment for infection in the dog. These findings suggest that community sources outside the home may be important for recurrent MRSA colonization or infection.


bioRxiv | 2017

Aging-associated dysbiosis increases susceptibility to enteric viral infection in Drosophila

Christine Sansone; Jonathan D. Cohen; Beth Gold; Wenhan Zhu; Ana M. Misic; Daniel P. Beiting; Sebastian E. Winter; Sara Cherry

Age is associated with increased susceptibility to enteric infections, but the molecular mechanisms are unclear. We find that aged Drosophila are more susceptible to enteric viral infections and that this increase in susceptibility is due to the aged microbiota, since depletion of the microbiota or reconstitution with a young microbiome suppressed infection. Metagenomic analysis of the aged microbiome revealed dysbiosis with an increased abundance in reactive oxygen species (ROS) producing pathways. This aged microbiota drives intestinal ROS production and we could restore immune function in old flies by reducing ROS genetically or pharmacologically. Moreover, we found that reconstitution of old flies with a cocktail of commensals, including L. fructivorans and heat-killed A. pomorum, could fully restore immunity. Altogether, these findings provide a mechanistic link between age-dependent dysbiosis and antiviral immunity and show that we can restore innate protection in aged animals, suggesting that this is a treatable and reversible state.


Nature Neuroscience | 2018

The maternal vaginal microbiome partially mediates the effects of prenatal stress on offspring gut and hypothalamus

Eldin Jašarević; Christopher D. Howard; Kathleen E. Morrison; Ana M. Misic; Tiffany Weinkopff; Phillip Scott; Christopher A. Hunter; Daniel P. Beiting; Tracy L. Bale

Early prenatal stress disrupts maternal-to-offspring microbiota transmission and has lasting effects on metabolism, physiology, cognition, and behavior in male mice. Here we show that transplantation of maternal vaginal microbiota from stressed dams into naive pups delivered by cesarean section had effects that partly resembled those seen in prenatally stressed males. However, transplantation of control maternal vaginal microbiota into prenatally stressed pups delivered by cesarean section did not rescue the prenatal-stress phenotype. Prenatal stress was associated with alterations in the fetal intestinal transcriptome and niche, as well as with changes in the adult gut that were altered by additional stress exposure in adulthood. Further, maternal vaginal transfer also partially mediated the effects of prenatal stress on hypothalamic gene expression, as observed after chronic stress in adulthood. These findings suggest that the maternal vaginal microbiota contribute to the lasting effects of prenatal stress on gut and hypothalamus in male mice.Maternal stress during pregnancy is a risk factor for neurodevelopmental disorders. Jasarevic and colleagues show that the maternal vaginal microbiota partially mediates the lasting effects of prenatal stress on the gut and hypothalamus in mice.


Advances in wound care | 2014

The Wound Microbiome: Modern Approaches to Examining the Role of Microorganisms in Impaired Chronic Wound Healing

Ana M. Misic; Sue E. Gardner; Elizabeth A. Grice


Journal of Investigative Dermatology | 2016

Longitudinal Evaluation of the Skin Microbiome and Association with Microenvironment and Treatment in Canine Atopic Dermatitis.

Charles W. Bradley; Daniel O. Morris; Shelley C. Rankin; Christine L. Cain; Ana M. Misic; Timothy Houser; Elizabeth A. Mauldin; Elizabeth A. Grice

Collaboration


Dive into the Ana M. Misic's collaboration.

Top Co-Authors

Avatar

Daniel P. Beiting

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Daniel O. Morris

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shelley C. Rankin

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Christine L. Cain

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Stephen D. Cole

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ebbing Lautenbach

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge