Network


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

Hotspot


Dive into the research topics where Kelli W. Williams is active.

Publication


Featured researches published by Kelli W. Williams.


JCI insight | 2016

Extrapulmonary Aspergillus infection in patients with CARD9 deficiency

Nikolaus Rieber; Roel P. Gazendam; Alexandra F. Freeman; Amy P. Hsu; Amanda L. Collar; Janyce A. Sugui; Rebecca A. Drummond; Chokechai Rongkavilit; Kevin W. Hoffman; Carolyn Henderson; Lily P. Clark; Markus Mezger; Muthulekha Swamydas; Maik Engeholm; Rebecca Schüle; Bettina Neumayer; Frank Ebel; Constantinos M. Mikelis; Stefania Pittaluga; Vinod K. Prasad; Anurag Singh; Joshua D. Milner; Kelli W. Williams; Jean K. Lim; Kyung J. Kwon-Chung; Steven M. Holland; Dominik Hartl; Taco W. Kuijpers; Michail S. Lionakis

Invasive pulmonary aspergillosis is a life-threatening mycosis that only affects patients with immunosuppression, chemotherapy-induced neutropenia, transplantation, or congenital immunodeficiency. We studied the clinical, genetic, histological, and immunological features of 2 unrelated patients without known immunodeficiency who developed extrapulmonary invasive aspergillosis at the ages of 8 and 18. One patient died at age 12 with progressive intra-abdominal aspergillosis. The other patient had presented with intra-abdominal candidiasis at age 9, and developed central nervous system aspergillosis at age 18 and intra-abdominal aspergillosis at age 25. Neither patient developed Aspergillus infection of the lungs. One patient had homozygous M1I CARD9 (caspase recruitment domain family member 9) mutation, while the other had homozygous Q295X CARD9 mutation; both patients lacked CARD9 protein expression. The patients had normal monocyte and Th17 cell numbers in peripheral blood, but their mononuclear cells exhibited impaired production of proinflammatory cytokines upon fungus-specific stimulation. Neutrophil phagocytosis, killing, and oxidative burst against Aspergillus fumigatus were intact, but neither patient accumulated neutrophils in infected tissue despite normal neutrophil numbers in peripheral blood. The neutrophil tissue accumulation defect was not caused by defective neutrophil-intrinsic chemotaxis, indicating that production of neutrophil chemoattractants in extrapulmonary tissue is impaired in CARD9 deficiency. Taken together, our results show that CARD9 deficiency is the first known inherited or acquired condition that predisposes to extrapulmonary Aspergillus infection with sparing of the lungs, associated with impaired neutrophil recruitment to the site of infection.


JCI insight | 2016

Transplantation of human skin microbiota in models of atopic dermatitis

Ian A. Myles; Kelli W. Williams; Jensen D. Reckhow; Momodou L. Jammeh; Nathan B. Pincus; Inka Sastalla; Danial Saleem; Kelly D. Stone; Sandip K. Datta

Atopic dermatitis (AD) is characterized by reduced barrier function, reduced innate immune activation, and susceptibility to Staphylococcus aureus. Host susceptibility factors are suggested by monogenic disorders associated with AD-like phenotypes and can be medically modulated. S. aureus contributes to AD pathogenesis and can be mitigated by antibiotics and bleach baths. Recent work has revealed that the skin microbiome differs significantly between healthy controls and patients with AD, including decreased Gram-negative bacteria in AD. However, little is known about the potential therapeutic benefit of microbiome modulation. To evaluate whether parameters of AD pathogenesis are altered after exposure to different culturable Gram-negative bacteria (CGN) collected from human skin, CGN were collected from healthy controls and patients with AD. Then, effects on cellular and culture-based models of immune, epithelial, and bacterial function were evaluated. Representative strains were evaluated in the MC903 mouse model of AD. We found that CGN taken from healthy volunteers but not from patients with AD were associated with enhanced barrier function, innate immunity activation, and control of S. aureus. Treatment with CGN from healthy controls improved outcomes in a mouse model of AD. These findings suggest that a live-biotherapeutic approach may hold promise for treatment of patients with AD.


Immunology and Allergy Clinics of North America | 2015

Eosinophilia Associated with Disorders of Immune Deficiency or Immune Dysregulation.

Kelli W. Williams; Joshua D. Milner; Alexandra F. Freeman

Increased serum eosinophil levels have been associated with multiple disorders of immune deficiency or immune dysregulation. Although primary immunodeficiency diseases are rare, it is important to consider these in the differential diagnosis of patients with eosinophilia. In this review, the clinical features, laboratory findings, diagnosis, and genetic basis of disease of several disorders of immune deficiency or dysregulation are discussed. The article includes autosomal dominant hyper IgE syndrome, DOCK8 deficiency, phosphoglucomutase 3 deficiency, ADA-SCID, Omenn syndrome, Wiskott-Aldrich syndrome, Loeys-Dietz syndrome, autoimmune lymphoproliferative syndrome, immunodysregulation, polyendocrinopathy, enteropathy, X-linked syndrome, Comel-Netherton syndrome, and severe dermatitis, multiple allergies, and metabolic wasting syndrome.


Immunology and Allergy Clinics of North America | 2015

Anaphylaxis and Urticaria

Kelli W. Williams; Hemant P. Sharma

Anaphylaxis and urticaria are common presenting allergic complaints. Affecting up to 2% of the population, anaphylaxis is a serious, life-threatening allergic reaction. Although not life-threatening, urticaria is a rash of transient, erythematous, pruritic wheals that can be bothersome and affects up to 25% of the population. All cases of anaphylaxis warrant thorough clinical evaluation by the allergist-immunologist, although most cases of urticaria are self-limited and do not require specialist referral. This article offers an overview of our current knowledge on the epidemiology, pathogenesis, triggers, diagnosis, and treatment of anaphylaxis and urticaria.


The Journal of Allergy and Clinical Immunology: In Practice | 2014

Telangiectasia macularis eruptiva perstans or highly vascularized urticaria pigmentosa

Kelli W. Williams; Dean D. Metcalfe; Calman Prussin; Melody C. Carter; Hirsh D. Komarow

o comment; Both, by both skin biopsy and clinical observation; Bx, skin biopsy; C sinophils, ISM, indolent systemic mastocytosis; L, lymphocytic; M, mastocytosis; M rted; SM, systemic mastocytosis; Y, yes. seen on skin biopsy specimen. This original report did not include histologic findings. Weber subsequently evaluated a second case of a patient with urticaria pigmentosa (UP) that closely resembled the index patient with TMEP and, based on these 2 cases, concluded that TMEP was a pigmentless variant of UP that presents primarily in adulthood. In subsequent reports, mast cell infiltration was noted histologically in both UP and TMEP; however, the gross appearance and perivascular distribution of mast cells and association with dilated vessels was said to distinguish TMEP from UP. Thus, TMEP now is usually referred to as a rare form of cutaneous mastocytosis in which the skin has flat telangiectatic macules in a generalized distribution. The relative lack of information on the consequences of a diagnosis of TMEP has led to confusion in clinical management. Based on these observations, we conducted a survey of patients who were evaluated for mastocytosis and who carried a diagnosis of TMEP to determine how the diagnosis of TMEP was being applied and if there were any consistent features to report. By using the National Institutes of Health Biomedical Translational Research Information System, we conducted a retrospective chart review of 299 subjects who were referred to the National Institute of Allergy and Infectious Diseases, with institutional review board approved mastocytosis protocols. All the subjects


BMC Microbiology | 2016

A method for culturing Gram-negative skin microbiota

Ian A. Myles; Jensen D. Reckhow; Kelli W. Williams; Inka Sastalla; Karen M. Frank; Sandip K. Datta

BackgroundCommensal Gram-negative (CGN) microbiota have been identified on human skin by DNA sequencing; however, methods to reliably culture viable Gram-negative skin organisms have not been previously described.ResultsThrough the use of selective antibiotics and minimal media we developed methods to culture CGN from skin swabs. We identified several previously uncharacterized CGN at the species level by optimizing growth conditions and limiting the inhibitory effects of nutrient shock, temperature, and bacterial competition, factors that may have previously limited CGN isolation from skin cultures.ConclusionsOur protocol will permit future functional studies on the influences of CGN on skin homeostasis and disease.


JCI insight | 2018

First-in-human topical microbiome transplantation with Roseomonas mucosa for atopic dermatitis

Ian A. Myles; Noah J. Earland; Erik D. Anderson; Ian N. Moore; Mark D. Kieh; Kelli W. Williams; Arhum Saleem; Natalia M. Fontecilla; Pamela Welch; Dirk A. Darnell; Lisa A. Barnhart; Ashleigh A. Sun; Gulbu Uzel; Sandip K. Datta

The underlying pathology of atopic dermatitis (AD) includes impaired skin barrier function, susceptibility to Staphylococcus aureus skin infection, immune dysregulation, and cutaneous dysbiosis. Our recent investigation into the potential role of Gram-negative skin bacteria in AD revealed that isolates of one particular commensal, Roseomonas mucosa, collected from healthy volunteers (HVs) improved outcomes in mouse and cell culture models of AD. In contrast, isolates of R. mucosa from patients with AD worsened outcomes in these models. These preclinical results suggested that interventions targeting the microbiome could provide therapeutic benefit for patients with AD. As a first test of this hypothesis in humans, 10 adult and 5 pediatric patients were enrolled in an open-label phase I/II safety and activity trial (the Beginning Assessment of Cutaneous Treatment Efficacy for Roseomonas in Atopic Dermatitis trial; BACTERiAD I/II). Treatment with R. mucosa was associated with significant decreases in measures of disease severity, topical steroid requirement, and S. aureus burden. There were no adverse events or treatment complications. We additionally evaluated differentiating bacterial metabolites and topical exposures that may contribute to the skin dysbiosis associated with AD and/or influence future microbiome-based treatments. These early results support continued evaluation of R. mucosa therapy with a placebo-controlled trial.


Journal of Clinical Investigation | 2018

TNF overproduction impairs epithelial staphylococcal response in hyper IgE syndrome

Ian A. Myles; Erik D. Anderson; Noah J. Earland; Kol A. Zarember; Inka Sastalla; Kelli W. Williams; Portia Gough; Ian N. Moore; Sundar Ganesan; Cedar J. Fowler; Arian Laurence; Mary Garofalo; Douglas B. Kuhns; Mark D. Kieh; Arhum Saleem; Pamela Welch; Dirk A. Darnell; John I. Gallin; Alexandra F. Freeman; Steven M. Holland; Sandip K. Datta

Autosomal dominant hyper IgE syndrome (AD-HIES), or Job’s syndrome, is a primary immune deficiency caused by dominant-negative mutations in STAT3. Recurrent Staphylococcus aureus skin abscesses are a defining feature of this syndrome. A widely held hypothesis that defects in peripheral Th17 differentiation confer this susceptibility has never been directly evaluated. To assess the cutaneous immune response in AD-HIES, we induced suction blisters in healthy volunteers (HVs) and patients with AD-HIES and then challenged the wound with lethally irradiated bacteria. We show that cutaneous production of IL-17A and IL-17F was normal in patients with AD-HIES. Overproduction of TNF-&agr; differentiated the responses in AD-HIES from HVs. This was associated with reduced IL-10 family signaling in blister-infiltrating cells and defective epithelial cell function. Mouse models of AD-HIES recapitulated these aberrant epithelial responses to S. aureus and involved defective epithelial-to-mesenchymal transition (EMT) rather than a failure of bacterial killing. Defective responses in mouse models of AD-HIES and primary keratinocyte cultures from patients with AD-HIES could be reversed by TNF-&agr; blockade and by drugs with reported modulatory effects on EMT. Our results identify these as potential therapeutic approaches in patients with AD-HIES suffering S. aureus infections.


Pathogenetics | 2017

Molecular Typing of Staphylococcus aureus Isolated from Patients with Autosomal Dominant Hyper IgE Syndrome

Inka Sastalla; Kelli W. Williams; Erik D. Anderson; Ian A. Myles; Jensen D. Reckhow; Marlene Espinoza-Moraga; Alexandra F. Freeman; Sandip K. Datta

Autosomal dominant hyper IgE syndrome (AD-HIES) is a primary immunodeficiency caused by a loss-of-function mutation in the Signal Transducer and Activator of Transcription 3 (STAT3). This immune disorder is clinically characterized by increased susceptibility to cutaneous and sinopulmonary infections, in particular with Candida and Staphylococcus aureus. It has recently been recognized that the skin microbiome of patients with AD-HIES is altered with an overrepresentation of certain Gram-negative bacteria and Gram-positive staphylococci. However, these alterations have not been characterized at the species- and strain-level. Since S. aureus infections are influenced by strain-specific expression of virulence factors, information on colonizing strain characteristics may provide insights into host-pathogen interactions and help guide management strategies for treatment and prophylaxis. The aim of this study was to determine whether the immunodeficiency of AD-HIES selects for unique strains of colonizing S. aureus. Using multi-locus sequence typing (MLST), protein A (spa) typing, and PCR-based detection of toxin genes, we performed a detailed analysis of the S. aureus isolates (n = 13) found on the skin of twenty-one patients with AD-HIES. We found a low diversity of sequence types, and an abundance of strains that expressed methicillin resistance, Panton-Valentine leukocidin (PVL), and staphylococcal enterotoxins K and Q (SEK, SEQ). Our results indicate that patients with AD-HIES may often carry antibiotic-resistant strains that harbor key virulence factors.


The Journal of Allergy and Clinical Immunology | 2017

Abnormal B-cell maturation in the bone marrow of patients with germline mutations in PIK3CD

Alina Dulau Florea; Raul C. Braylan; Kristian T. Schafernak; Kelli W. Williams; Janine Daub; Rakesh K. Goyal; Jennifer M. Puck; V. Koneti Rao; Stefania Pittaluga; Steven M. Holland; Gulbu Uzel; Katherine R. Calvo

Collaboration


Dive into the Kelli W. Williams's collaboration.

Top Co-Authors

Avatar

Alexandra F. Freeman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ian A. Myles

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Inka Sastalla

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Steven M. Holland

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Erik D. Anderson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jensen D. Reckhow

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Joshua D. Milner

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dirk A. Darnell

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gulbu Uzel

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge