Network


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

Hotspot


Dive into the research topics where Mary K. Estes is active.

Publication


Featured researches published by Mary K. Estes.


Science | 2016

Replication of human noroviruses in stem cell–derived human enteroids

Khalil Ettayebi; Sue E. Crawford; Kosuke Murakami; James R. Broughman; Umesh C. Karandikar; Victoria R. Tenge; Frederick H. Neill; Sarah E. Blutt; Xi-Lei Zeng; Lin Qu; Baijun Kou; Antone R. Opekun; Douglas G. Burrin; David Y. Graham; Sasirekha Ramani; Robert L. Atmar; Mary K. Estes

The major barrier to research and development of effective interventions for human noroviruses (HuNoVs) has been the lack of a robust and reproducible in vitro cultivation system. HuNoVs are the leading cause of gastroenteritis worldwide. We report the successful cultivation of multiple HuNoV strains in enterocytes in stem cell–derived, nontransformed human intestinal enteroid monolayer cultures. Bile, a critical factor of the intestinal milieu, is required for strain-dependent HuNoV replication. Lack of appropriate histoblood group antigen expression in intestinal cells restricts virus replication, and infectivity is abrogated by inactivation (e.g., irradiation, heating) and serum neutralization. This culture system recapitulates the human intestinal epithelium, permits human host-pathogen studies of previously noncultivatable pathogens, and allows the assessment of methods to prevent and treat HuNoV infections.


Journal of Biological Chemistry | 2016

Human Enteroids/Colonoids and Intestinal Organoids Functionally Recapitulate Normal Intestinal Physiology and Pathophysiology

Nicholas C. Zachos; Olga Kovbasnjuk; Jennifer Foulke-Abel; Julie In; Sarah E. Blutt; Hugo R. de Jonge; Mary K. Estes; Mark Donowitz

Identification of Lgr5 as the intestinal stem cell marker as well as the growth factors necessary to replicate adult intestinal stem cell division has led to the establishment of the methods to generate “indefinite” ex vivo primary intestinal epithelial cultures, termed “mini-intestines.” Primary cultures developed from isolated intestinal crypts or stem cells (termed enteroids/colonoids) and from inducible pluripotent stem cells (termed intestinal organoids) are being applied to study human intestinal physiology and pathophysiology with great expectations for translational applications, including regenerative medicine. Here we discuss the physiologic properties of these cultures, their current use in understanding diarrhea-causing host-pathogen interactions, and potential future applications.


Experimental Biology and Medicine | 2014

Human enteroids as an ex-vivo model of host–pathogen interactions in the gastrointestinal tract

Jennifer Foulke-Abel; Julie In; Olga Kovbasnjuk; Nicholas C. Zachos; Khalil Ettayebi; Sarah E. Blutt; Joseph M. Hyser; Xi Lei Zeng; Sue E. Crawford; James R. Broughman; Mary K. Estes; Mark Donowitz

Currently, 9 out of 10 experimental drugs fail in clinical studies. This has caused a 40% plunge in the number of drugs approved by the US Food and Drug Administration (FDA) since 2005. It has been suggested that the mechanistic differences between human diseases modeled in animals (mostly rodents) and the pathophysiology of human diseases might be one of the critical factors that contribute to drug failure in clinical trials. Rapid progress in the field of human stem cell technology has allowed the in-vitro recreation of human tissue that should complement and expand upon the limitations of cell and animal models currently used to study human diseases and drug toxicity. Recent success in the identification and isolation of human intestinal epithelial stem cells (Lgr5+) from the small intestine and colon has led to culture of functional intestinal epithelial units termed organoids or enteroids. Intestinal enteroids are comprised of all four types of normal epithelial cells and develop a crypt–villus differentiation axis. They demonstrate major intestinal physiologic functions, including Na+ absorption and Cl− secretion. This review discusses the recent progress in establishing human enteroids as a model of infectious diarrheal diseases such as cholera, rotavirus, and enterohemorrhagic Escherichia coli, and use of the enteroids to determine ways to correct the diarrhea-induced ion transport abnormalities via drug therapy.


Scientific Reports | 2017

Functional Coupling of Human Microphysiology Systems: Intestine, Liver, Kidney Proximal Tubule, Blood-Brain Barrier and Skeletal Muscle

Lawrence Vernetti; Albert Gough; Nicholas W. Baetz; Sarah E. Blutt; James R. Broughman; Jacquelyn A. Brown; Jennifer Foulke-Abel; Nesrin M. Hasan; Julie In; Edward J. Kelly; Olga Kovbasnjuk; Jonathan Repper; Nina Senutovitch; Janet Stabb; Catherine K. Yeung; Nick Zachos; Mark Donowitz; Mary K. Estes; Jonathan Himmelfarb; George A. Truskey; John P. Wikswo; D. Lansing Taylor

Organ interactions resulting from drug, metabolite or xenobiotic transport between organs are key components of human metabolism that impact therapeutic action and toxic side effects. Preclinical animal testing often fails to predict adverse outcomes arising from sequential, multi-organ metabolism of drugs and xenobiotics. Human microphysiological systems (MPS) can model these interactions and are predicted to dramatically improve the efficiency of the drug development process. In this study, five human MPS models were evaluated for functional coupling, defined as the determination of organ interactions via an in vivo-like sequential, organ-to-organ transfer of media. MPS models representing the major absorption, metabolism and clearance organs (the jejunum, liver and kidney) were evaluated, along with skeletal muscle and neurovascular models. Three compounds were evaluated for organ-specific processing: terfenadine for pharmacokinetics (PK) and toxicity; trimethylamine (TMA) as a potentially toxic microbiome metabolite; and vitamin D3. We show that the organ-specific processing of these compounds was consistent with clinical data, and discovered that trimethylamine-N-oxide (TMAO) crosses the blood-brain barrier. These studies demonstrate the potential of human MPS for multi-organ toxicity and absorption, distribution, metabolism and excretion (ADME), provide guidance for physically coupling MPS, and offer an approach to coupling MPS with distinct media and perfusion requirements.


Archive | 1999

A Viral Enterotoxin

Mary K. Estes; Andrew P. Morris

Acute infectious gastroenteritis is a major cause of infant morbidity in developed countries and of infant mortality in developing areas of the world. Rotavirus is recognized as the most important etiologic agent of infantile gastroenteritis, and studies of rotavirus serve as models to understand the complex interactions between enteric viruses and the multifunctional cells of the gastrointestinal tract. Understanding such interactions is significant for microbial pathogenesis because most (> 80%) infections are initiated at mucosal surfaces. Rotaviruses are pathogens that infect the mature enterocytes of the villi in the intestine and infection appears to be limited to these highly differentiated cells in immunologically competent hosts. In such hosts, infections are generally acute yet diarrheal disease can be severe and life-threatening. Disease generally is resolved within 2-5 days after infection if affected hosts receive adequate rehydration. In immunocompromised hosts, virus infections persist, virus can be detected extraintestinally and virus excretion may be detected for extended periods of time (many months). Rotaviruses infect almost all mammalian and some avian species and much of our understanding of rotavirus pathogenesis has come from studies in animal models, particularly in small animal models (mice and rabbits), but also in larger animals (cows and piglets). Studies in children are limited due to the difficulty and lack of clinical need of obtaining biopsies from infants and the inability to determine the precise time of natural infections. In all animal species where naïve animals can be infected, disease is age-dependent; for example, in mice and rabbits, diarrheal disease is the outcome of infections that occur only during the first two weeks of life (Ciarlet et al., 1998; Starkey et al., 1986; Ramig 1988; Ward et al., 1990; Burns et al., 1995), while animals remain susceptible to viral infection into adulthood. Rotavirus infections have been reported to occur repeatedly in humans from birth to old age, but the majority of infections after the first 2 years of life are asymptomatic or associated with mild gastrointestinal symptoms. The age-related resistance to rotavirus-induced diarrhea in humans is thought to be mediated primarily by acquired immunity, but it is not possible to directly test if humans also exhibit an age-dependent resistance to disease based on other factors such as intestinal development and maturation. Currently, our best understanding of the mechanisms of rotavirus pathogenesis rely on results obtained in animal models.


Proceedings of the National Academy of Sciences of the United States of America | 2017

A paradox of transcriptional and functional innate interferon responses of human intestinal enteroids to enteric virus infection

Kapil Saxena; Lukas M. Simon; Xi Lei Zeng; Sarah E. Blutt; Sue E. Crawford; Narayan P. Sastri; Umesh C. Karandikar; Nadim J. Ajami; Nicholas C. Zachos; Olga Kovbasnjuk; Mark Donowitz; Margaret E. Conner; Chad A. Shaw; Mary K. Estes

Significance Understanding host–enteric virus interactions has been limited by the inability to culture nontransformed small intestinal epithelial cells and to infect animal models with human viruses. We report epithelial responses in human small intestinal enteroid cultures from different individuals following infection with human rotavirus (HRV), a model enteric pathogen. RNA-sequencing and functional assays revealed type III IFN as the dominant transcriptional response that activates interferon-stimulated genes, but antagonism of the IFN response negates restriction of HRV replication. Exogenously added IFNs reduce HRV replication, with type I IFN being most effective. This highlights a paradox between the strong type III transcriptional response and the weaker functional role of type III IFN in human enteric viral restriction in human small intestinal cultures. The intestinal epithelium can limit enteric pathogens by producing antiviral cytokines, such as IFNs. Type I IFN (IFN-α/β) and type III IFN (IFN-λ) function at the epithelial level, and their respective efficacies depend on the specific pathogen and site of infection. However, the roles of type I and type III IFN in restricting human enteric viruses are poorly characterized as a result of the difficulties in cultivating these viruses in vitro and directly obtaining control and infected small intestinal human tissue. We infected nontransformed human intestinal enteroid cultures from multiple individuals with human rotavirus (HRV) and assessed the host epithelial response by using RNA-sequencing and functional assays. The dominant transcriptional pathway induced by HRV infection is a type III IFN-regulated response. Early after HRV infection, low levels of type III IFN protein activate IFN-stimulated genes. However, this endogenous response does not restrict HRV replication because replication-competent HRV antagonizes the type III IFN response at pre- and posttranscriptional levels. In contrast, exogenous IFN treatment restricts HRV replication, with type I IFN being more potent than type III IFN, suggesting that extraepithelial sources of type I IFN may be the critical IFN for limiting enteric virus replication in the human intestine.


Archive | 2017

Human Intestinal Enteroids: New Models to Study Gastrointestinal Virus Infections

Winnie Y. Zou; Sarah E. Blutt; Sue E. Crawford; Khalil Ettayebi; Xi-Lei Zeng; Kapil Saxena; Sasirekha Ramani; Umesh C. Karandikar; Nicholas C. Zachos; Mary K. Estes

Human rotavirus (HRV) and human norovirus (HuNoV) infections are recognized as the most common causes of epidemic and sporadic cases of gastroenteritis worldwide. The study of these two human gastrointestinal viruses is important for understanding basic virus-host interactions and mechanisms of pathogenesis and to establish models to evaluate vaccines and treatments. Despite the introduction of live-attenuated vaccines to prevent life-threatening HRV-induced disease, the burden of HRV illness remains significant in low-income and less-industrialized countries, and small animal models or ex vivo models to study HRV infections efficiently are lacking. Similarly, HuNoVs remained non-cultivatable until recently. With the advent of non-transformed human intestinal enteroid (HIE) cultures, we are now able to culture and study both clinically relevant HRV and HuNoV in a biologically relevant human system. Methods described here will allow investigators to use these new culture techniques to grow HRV and HuNoV and analyze new aspects of virus replication and pathogenesis.


Journal of Virology | 2016

Replication of Human Norovirus RNA in Mammalian Cells Reveals Lack of Interferon Response.

Lin Qu; Kosuke Murakami; James R. Broughman; Margarita K. Lay; Susana Guix; Victoria R. Tenge; Robert L. Atmar; Mary K. Estes

ABSTRACT Human noroviruses (HuNoVs), named after the prototype strain Norwalk virus (NV), are a leading cause of acute gastroenteritis outbreaks worldwide. Studies on the related murine norovirus (MNV) have demonstrated the importance of an interferon (IFN) response in host control of virus replication, but this remains unclear for HuNoVs. Despite the lack of an efficient cell culture infection system, transfection of stool-isolated NV RNA into mammalian cells leads to viral RNA replication and virus production. Using this system, we show here that NV RNA replication is sensitive to type I (α/β) and III (interleukin-29 [IL-29]) IFN treatment. However, in cells capable of a strong IFN response to Sendai virus (SeV) and poly(I·C), NV RNA replicates efficiently and generates double-stranded RNA without inducing a detectable IFN response. Replication of HuNoV genogroup GII.3 strain U201 RNA, generated from a reverse genetics system, also does not induce an IFN response. Consistent with a lack of IFN induction, NV RNA replication is enhanced neither by neutralization of type I/III IFNs through neutralizing antibodies or the soluble IFN decoy receptor B18R nor by short hairpin RNA (shRNA) knockdown of mitochondrial antiviral signaling protein (MAVS) or interferon regulatory factor 3 (IRF3) in the IFN induction pathways. In contrast to other positive-strand RNA viruses that block IFN induction by targeting MAVS for degradation, MAVS is not degraded in NV RNA-replicating cells, and an SeV-induced IFN response is not blocked. Together, these results indicate that HuNoV RNA replication in mammalian cells does not induce an IFN response, suggesting that the epithelial IFN response may play a limited role in host restriction of HuNoV replication. IMPORTANCE Human noroviruses (HuNoVs) are a leading cause of epidemic gastroenteritis worldwide. Due to lack of an efficient cell culture system and robust small-animal model, little is known about the innate host defense to these viruses. Studies on murine norovirus (MNV) have shown the importance of an interferon (IFN) response in host control of MNV replication, but this remains unclear for HuNoVs. Here, we investigated the IFN response to HuNoV RNA replication in mammalian cells using Norwalk virus stool RNA transfection, a reverse genetics system, IFN neutralization reagents, and shRNA knockdown methods. Our results show that HuNoV RNA replication in mammalian epithelial cells does not induce an IFN response, nor can it be enhanced by blocking the IFN response. These results suggest a limited role of the epithelial IFN response in host control of HuNoV RNA replication, providing important insights into our understanding of the host defense to HuNoVs that differs from that to MNV.


Journal of Virology | 1992

Expression, self-assembly, and antigenicity of the Norwalk virus capsid protein.

Xi Jiang; Min Wang; D. Y. Graham; Mary K. Estes


Virology | 1993

Sequence and Genomic Organization of Norwalk Virus

Xi Jiang; Min Wang; Kening Wang; Mary K. Estes

Collaboration


Dive into the Mary K. Estes's collaboration.

Top Co-Authors

Avatar

Sarah E. Blutt

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Sue E. Crawford

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark Donowitz

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Nicholas C. Zachos

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Olga Kovbasnjuk

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jennifer Foulke-Abel

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Julie In

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge