Network


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

Hotspot


Dive into the research topics where Julie In is active.

Publication


Featured researches published by Julie In.


Journal of Virology | 2016

Human Intestinal Enteroids: A New Model to Study Human Rotavirus Infection, Host Restriction and Pathophysiology

Kapil Saxena; Sarah E. Blutt; Khalil Ettayebi; Xi Lei Zeng; James R. Broughman; Sue E. Crawford; Umesh C. Karandikar; Narayan P. Sastri; Margaret E. Conner; Antone R. Opekun; David Y. Graham; Waqar A. Qureshi; Vadim Sherman; Jennifer Foulke-Abel; Julie In; Olga Kovbasnjuk; Nicholas C. Zachos; Mark Donowitz; Mary K. Estes

ABSTRACT Human gastrointestinal tract research is limited by the paucity of in vitro intestinal cell models that recapitulate the cellular diversity and complex functions of human physiology and disease pathology. Human intestinal enteroid (HIE) cultures contain multiple intestinal epithelial cell types that comprise the intestinal epithelium (enterocytes and goblet, enteroendocrine, and Paneth cells) and are physiologically active based on responses to agonists. We evaluated these nontransformed, three-dimensional HIE cultures as models for pathogenic infections in the small intestine by examining whether HIEs from different regions of the small intestine from different patients are susceptible to human rotavirus (HRV) infection. Little is known about HRVs, as they generally replicate poorly in transformed cell lines, and host range restriction prevents their replication in many animal models, whereas many animal rotaviruses (ARVs) exhibit a broader host range and replicate in mice. Using HRVs, including the Rotarix RV1 vaccine strain, and ARVs, we evaluated host susceptibility, virus production, and cellular responses of HIEs. HRVs infect at higher rates and grow to higher titers than do ARVs. HRVs infect differentiated enterocytes and enteroendocrine cells, and viroplasms and lipid droplets are induced. Heterogeneity in replication was seen in HIEs from different patients. HRV infection and RV enterotoxin treatment of HIEs caused physiological lumenal expansion detected by time-lapse microscopy, recapitulating one of the hallmarks of rotavirus-induced diarrhea. These results demonstrate that HIEs are a novel pathophysiological model that will allow the study of HRV biology, including host restriction, cell type restriction, and virus-induced fluid secretion. IMPORTANCE Our research establishes HIEs as nontransformed cell culture models to understand human intestinal physiology and pathophysiology and the epithelial response, including host restriction of gastrointestinal infections such as HRV infection. HRVs remain a major worldwide cause of diarrhea-associated morbidity and mortality in children ≤5 years of age. Current in vitro models of rotavirus infection rely primarily on the use of animal rotaviruses because HRV growth is limited in most transformed cell lines and animal models. We demonstrate that HIEs are novel, cellularly diverse, and physiologically relevant epithelial cell cultures that recapitulate in vivo properties of HRV infection. HIEs will allow the study of HRV biology, including human host-pathogen and live, attenuated vaccine interactions; host and cell type restriction; virus-induced fluid secretion; cell-cell communication within the epithelium; and the epithelial response to infection in cultures from genetically diverse individuals. Finally, drug therapies to prevent/treat diarrheal disease can be tested in these physiologically active cultures.


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.


Cellular and molecular gastroenterology and hepatology | 2016

Enterohemorrhagic Escherichia coli Reduces Mucus and Intermicrovillar Bridges in Human Stem Cell-Derived Colonoids

Julie In; Jennifer Foulke-Abel; Nicholas C. Zachos; Anne-Marie Hansen; James B. Kaper; Harris D. Bernstein; Marc K. Halushka; Sarah E. Blutt; Mary K. Estes; Mark Donowitz; Olga Kovbasnjuk

Background & Aims Enterohemorrhagic Escherichia coli (EHEC) causes over 70,000 episodes of foodborne diarrhea annually in the United States. The early sequence of events that precede life-threatening hemorrhagic colitis and hemolytic uremic syndrome is not fully understood due to the initial asymptomatic phase of the disease and the lack of a suitable animal model. We determined the initial molecular events in the interaction between EHEC and human colonic epithelium. Methods Human colonoids derived from adult proximal colonic stem cells were developed into monolayers to study EHEC-epithelial interactions. Monolayer confluency and differentiation were monitored by transepithelial electrical resistance measurements. The monolayers were apically infected with EHEC, and the progression of epithelial damage over time was assessed using biochemical and imaging approaches. Results Human colonoid cultures recapitulate the differential protein expression patterns characteristic of the crypt and surface colonocytes. Mucus-producing differentiated colonoid monolayers are preferentially colonized by EHEC. Upon colonization, EHEC forms characteristic attaching and effacing lesions on the apical surface of colonoid monolayers. Mucin 2, a main component of colonic mucus, and protocadherin 24 (PCDH24), a microvillar resident protein, are targeted by EHEC at early stages of infection. The EHEC-secreted serine protease EspP initiates brush border damage through PCDH24 reduction. Conclusions Human colonoid monolayers are a relevant pathophysiologic model that allow the study of early molecular events during enteric infections. Colonoid monolayers provide access to both apical and basolateral surfaces, thus providing an advantage over three-dimensional cultures to study host–pathogen interactions in a controllable and tractable manner. EHEC reduces colonic mucus and affects the brush border cytoskeleton in the absence of commensal bacteria.


Gastroenterology | 2016

Human Enteroids as a Model of Upper Small Intestinal Ion Transport Physiology and Pathophysiology.

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

BACKGROUND & AIMS Human intestinal crypt-derived enteroids are a model of intestinal ion transport that require validation by comparison with cell culture and animal models. We used human small intestinal enteroids to study neutral Na(+) absorption and stimulated fluid and anion secretion under basal and regulated conditions in undifferentiated and differentiated cultures to show their functional relevance to ion transport physiology and pathophysiology. METHODS Human intestinal tissue specimens were obtained from an endoscopic biopsy or surgical resections performed at Johns Hopkins Hospital. Crypts were isolated, enteroids were propagated in culture, induced to undergo differentiation, and transduced with lentiviral vectors. Crypt markers, surface cell enzymes, and membrane ion transporters were characterized using quantitative reverse-transcription polymerase chain reaction, immunoblot, or immunofluorescence analyses. We used multiphoton and time-lapse confocal microscopy to monitor intracellular pH and luminal dilatation in enteroids under basal and regulated conditions. RESULTS Enteroids differentiated upon withdrawal of WNT3A, yielding decreased crypt markers and increased villus-like characteristics. Na(+)/H(+) exchanger 3 activity was similar in undifferentiated and differentiated enteroids, and was affected by known inhibitors, second messengers, and bacterial enterotoxins. Forskolin-induced swelling was completely dependent on cystic fibrosis transmembrane conductance regulator and partially dependent on Na(+)/H(+) exchanger 3 and Na(+)/K(+)/2Cl(-) cotransporter 1 inhibition in undifferentiated and differentiated enteroids. Increases in cyclic adenosine monophosphate with forskolin caused enteroid intracellular acidification in HCO3(-)-free buffer. Cyclic adenosine monophosphate-induced enteroid intracellular pH acidification as part of duodenal HCO3(-) secretion appears to require cystic fibrosis transmembrane conductance regulator and electrogenic Na(+)/HCO3(-) cotransporter 1. CONCLUSIONS Undifferentiated or crypt-like, and differentiated or villus-like, human enteroids represent distinct points along the crypt-villus axis; they can be used to characterize electrolyte transport processes along the vertical axis of the small intestine. The duodenal enteroid model showed that electrogenic Na(+)/HCO3(-) cotransporter 1 might be a target in the intestinal mucosa for treatment of secretory diarrheas.


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.


Stem Cell Research & Therapy | 2013

Human enteroids: preclinical models of non-inflammatory diarrhea

Olga Kovbasnjuk; Nicholas C. Zachos; Julie In; Jennifer Foulke-Abel; Khalil Ettayebi; Joseph M. Hyser; James R. Broughman; Xi Lei Zeng; Sabine Middendorp; Hugo R. de Jonge; Mary K. Estes; Mark Donowitz

Researchers need an available and easy-to-use model of the human intestine to better understand human intestinal physiology and pathophysiology of diseases, and to offer an enhanced platform for developing drug therapy. Our work employs human enteroids derived from each of the major intestinal sections to advance understanding of several diarrheal diseases, including those caused by cholera, rotavirus and enterohemorrhagic Escherichia coli. An enteroid bank is being established to facilitate comparison of segmental, developmental, and regulatory differences in transport proteins that can influence therapy efficacy. Basic characterization of major ion transport protein expression, localization and function in the human enteroid model sets the stage to study the effects of enteric infection at the transport level, as well as to monitor potential responses to pharmacological intervention.


PLOS ONE | 2013

Serine Protease EspP from Enterohemorrhagic Escherichia Coli Is Sufficient to Induce Shiga Toxin Macropinocytosis in Intestinal Epithelium

Julie In; Valeriy Lukyanenko; Jennifer Foulke-Abel; Ann L. Hubbard; Michael Delannoy; Anne-Marie Hansen; James B. Kaper; Nadia Boisen; James P. Nataro; Chengru Zhu; Edgar C. Boedeker; Jorge A. Girón; Olga Kovbasnjuk

Life-threatening intestinal and systemic effects of the Shiga toxins produced by enterohemorrhagic Escherichia coli (EHEC) require toxin uptake and transcytosis across intestinal epithelial cells. We have recently demonstrated that EHEC infection of intestinal epithelial cells stimulates toxin macropinocytosis, an actin-dependent endocytic pathway. Host actin rearrangement necessary for EHEC attachment to enterocytes is mediated by the type 3 secretion system which functions as a molecular syringe to translocate bacterial effector proteins directly into host cells. Actin-dependent EHEC attachment also requires the outer membrane protein intimin, a major EHEC adhesin. Here, we investigate the role of type 3 secretion in actin turnover occurring during toxin macropinocytosis. Toxin macropinocytosis is independent of EHEC type 3 secretion and intimin attachment. EHEC soluble factors are sufficient to stimulate macropinocytosis and deliver toxin into enterocytes in vitro and in vivo; intact bacteria are not required. Intimin-negative enteroaggregative Escherichia coli (EAEC) O104:H4 robustly stimulate Shiga toxin macropinocytosis into intestinal epithelial cells. The apical macropinosomes formed in intestinal epithelial cells move through the cells and release their cargo at these cells’ basolateral sides. Further analysis of EHEC secreted proteins shows that a serine protease EspP alone is able to stimulate host actin remodeling and toxin macropinocytosis. The observation that soluble factors, possibly serine proteases including EspP, from each of two genetically distinct toxin-producing strains, can stimulate Shiga toxin macropinocytosis and transcellular transcytosis alters current ideas concerning mechanisms whereby Shiga toxin interacts with human enterocytes. Mechanisms important for this macropinocytic pathway could suggest new potential therapeutic targets for Shiga toxin-induced disease.


Toxins | 2018

Enterohemorrhagic E. Coli (EHEC)—Secreted serine protease EspP stimulates electrogenic ion transport in human colonoid monolayers

Chung-Ming Tse; Julie In; Jianyi Yin; Mark Donowitz; Michele Doucet; Jennifer Foulke-Abel; Fernando Ruiz-Perez; James P. Nataro; Nicholas C. Zachos; James B. Kaper; Olga Kovbasnjuk

One of the characteristic manifestations of Shiga-toxin-producing Escherichia coli (E. coli) infection in humans, including EHEC and Enteroaggregative E. coli O104:H4, is watery diarrhea. However, neither Shiga toxin nor numerous components of the type-3 secretion system have been found to independently elicit fluid secretion. We used the adult stem-cell-derived human colonoid monolayers (HCM) to test whether EHEC-secreted extracellular serine protease P (EspP), a member of the serine protease family broadly expressed by diarrheagenic E. coli can act as an enterotoxin. We applied the Ussing chamber/voltage clamp technique to determine whether EspP stimulates electrogenic ion transport indicated by a change in short-circuit current (Isc). EspP stimulates Isc in HCM. The EspP-stimulated Isc does not require protease activity, is not cystic fibrosis transmembrane conductance regulator (CFTR)-mediated, but is partially Ca2+-dependent. EspP neutralization with a specific antibody reduces its potency in stimulating Isc. Serine Protease A, secreted by Enteroaggregative E. coli, also stimulates Isc in HCM, but this current is CFTR-dependent. In conclusion, EspP stimulates colonic CFTR-independent active ion transport and may be involved in the pathophysiology of EHEC diarrhea. Serine protease toxins from E. coli pathogens appear to serve as enterotoxins, potentially significantly contributing to watery diarrhea.


Gastroenterology | 2013

887 Intraluminal Release of Galectin 4 in EHEC Infection Provides a Novel Host Defense Mechanism

Julie In; Sabine André; Hans-Joachim Gabius; Olga Kovbasnjuk

G A A b st ra ct s no significant difference in pancreatic MPO (unit mg of protein) in wild type 2981+734; CB(-/-)3042+767;T7(-/-) 3070+292 (P.0.05) treated with L-arginine, but it was significantly higher compared to saline treated animals(149+12). We also did not find a significant difference in lung MPO in unit /mg of protein in (Wild type 7766+1852; CB(-/)6234+602 and T7(-/-)7185+1236 treated with L-arginine HCL(P .0.05) but significantly higher than saline treated(2555+285) animals. These findings correlate with our finding in cerulein model of acute pancreatitis. Conclusion: Regardless of the model of acute pancreatitis trypsinogen activation significantly decreases acinar cell necrosis, but local and systemic inflammation is independent of trypsin activation.

Collaboration


Dive into the Julie In's collaboration.

Top Co-Authors

Avatar

Olga Kovbasnjuk

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Mark Donowitz

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

Nicholas C. Zachos

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Mary K. Estes

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Sarah E. Blutt

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jianyi Yin

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge