Network


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

Hotspot


Dive into the research topics where Jared Honeycutt is active.

Publication


Featured researches published by Jared Honeycutt.


Nature Cell Biology | 2014

Cellular origin of bladder neoplasia and tissue dynamics of its progression to invasive carcinoma

Kunyoo Shin; Agnes Lim; Justin I. Odegaard; Jared Honeycutt; Sally Kawano; Michael H. Hsieh; Philip A. Beachy

Understanding how malignancies arise within normal tissues requires identification of the cancer cell of origin and knowledge of the cellular and tissue dynamics of tumour progression. Here we examine bladder cancer in a chemical carcinogenesis model that mimics muscle-invasive human bladder cancer. With no prior bias regarding genetic pathways or cell types, we prospectively mark or ablate cells to show that muscle-invasive bladder carcinomas arise exclusively from Sonic hedgehog (Shh)-expressing stem cells in basal urothelium. These carcinomas arise clonally from a single cell whose progeny aggressively colonize a major portion of the urothelium to generate a lesion with histological features identical to human carcinoma in situ. Shh-expressing basal cells within this precursor lesion become tumour-initiating cells, although Shh expression is lost in subsequent carcinomas. We thus find that invasive carcinoma is initiated from basal urothelial stem cells but that tumour cell phenotype can diverge significantly from that of the cancer cell of origin.


Trends in Parasitology | 2014

Controversies and challenges in research on urogenital schistosomiasis-associated bladder cancer

Jared Honeycutt; Olfat Hammam; Chi-Ling Fu; Michael H. Hsieh

Urogenital schistosomiasis, infection with Schistosoma haematobium, is linked to increased risk for the development of bladder cancer, but the importance of various mechanisms responsible for this association remains unclear, in part, owing to lack of sufficient and appropriate animal models. New advances in the study of this parasite, bladder regenerative processes, and human schistosomal bladder cancers may shed new light on the complex biological processes that connect S. haematobium infection to bladder carcinogenesis.


The Journal of Infectious Diseases | 2015

New Research Tools for Urogenital Schistosomiasis

Gabriel Rinaldi; Neil D. Young; Jared Honeycutt; Paul J. Brindley; Robin B. Gasser; Michael H. Hsieh

Approximately 200,000,000 people have schistosomiasis (schistosome infection). Among the schistosomes, Schistosoma haematobium is responsible for the most infections, which are present in 110 million people globally, mostly in sub-Saharan Africa. This pathogen causes an astonishing breadth of sequelae: hematuria, anemia, dysuria, stunting, uremia, bladder cancer, urosepsis, and human immunodeficiency virus coinfection. Refined estimates of the impact of schistosomiasis on quality of life suggest that it rivals malaria. Despite S. haematobiums importance, relevant research has lagged. Here, we review advances that will deepen knowledge of S. haematobium. Three sets of breakthroughs will accelerate discoveries in the pathogenesis of urogenital schistosomiasis (UGS): (1) comparative genomics, (2) the development of functional genomic tools, and (3) the use of animal models to explore S. haematobium-host interactions. Comparative genomics for S. haematobium is feasible, given the sequencing of multiple schistosome genomes. Features of the S. haematobium genome that are conserved among platyhelminth species and others that are unique to S. haematobium may provide novel diagnostic and drug targets for UGS. Although there are technical hurdles, the integrated use of these approaches can elucidate host-pathogen interactions during this infection and can inform the development of techniques for investigating schistosomes in their human and snail hosts and the development of therapeutics and vaccines for the control of UGS.


Experimental Parasitology | 2015

Schistosoma haematobium egg-induced bladder urothelial abnormalities dependent on p53 are modulated by host sex

Jared Honeycutt; Olfat Hammam; Michael H. Hsieh

INTRODUCTION AND OBJECTIVE The bladder urothelium changes dramatically during Schistosoma haematobium infection (urogenital schistosomiasis). These alterations include hyperplasia, ulceration, dysplasia, squamous metaplasia and frank carcinogenesis. Defining the pathways underpinning these urothelial responses will contribute to a deeper understanding of how S. haematobium egg expulsion, hematuria, and bladder cancer develop in humans. The tumor suppressor gene p53 is of particular interest, given its role in many cancers, including bladder cancer generally and schistosomal bladder cancer specifically. METHODS Transgenic mice featuring tamoxifen-inducible Cre recombinase activity in cells expressing the urothelial-specific gene uroplakin-3a (Upk3a-GCE mice) were crossed with either TdTomato-floxed-EGFP reporter or p53-floxed mice. Mice were administered tamoxifen or vehicle control to induce excision of floxed genes. TdTomato-EGFP reporter mice were sacrificed and their bladders harvested, sectioned, and imaged by fluorescence microscopy. p53-floxed mice underwent bladder wall injection with S. haematobium eggs or vehicle controls. Three months later, mice were sacrificed and their bladders subjected to histological analysis (H&E staining). RESULTS We first confirmed the phenotypic fidelity of Upk3a-GCE mice by crossing them with TdTomato-floxed-EGFP reporter mice and administering tamoxifen to their progeny. As expected, these progeny switched from TdTomato to EGFP expression in their bladder urothelium. Having confirmed the phenotype of Upk3a-GCE mice, we next crossed them to p53-floxed mice. The resulting progeny were given tamoxifen or vehicle control to render them urothelial p53-haploinsufficient or -intact, respectively. Then, we injected S. haematobium eggs or control vehicle into the bladder walls of these mice. Male p53-intact, egg-injected mice exhibited similar histological changes as their p53-haploinsufficient counterparts, including urothelial hyperplasia and ulceration. In contrast, female p53-intact, egg-injected mice featured no urothelial ulceration, whereas their p53-haploinsufficient counterparts often had significant ulceration. CONCLUSIONS Urothelial p53 signaling indeed seems to affect urothelial homeostasis during S. haematobium infection, albeit in a sex-specific manner. Ongoing work seeks to determine whether p53 mediates associated alterations in urothelial cell cycle status and frank carcinogenesis in the setting of urogenital schistosomiasis.


PLOS Neglected Tropical Diseases | 2015

Alterations in DNA methylation may be the key to early detection and treatment of schistosomal bladder cancer

Simon Conti; Jared Honeycutt; Justin I. Odegaard; Mark L. Gonzalgo; Michael H. Hsieh

Bladder cancers arise from transformed urothelial cells that line the bladder. These cancers are urothelial or squamous cell carcinomas or, more rarely, additional histologic variants such as adenocarcinoma. The most important bladder cancer risk factors worldwide are arguably smoking and urogenital schistosomiasis. The parasitic Schistosoma haematobium worm causes urogenital schistosomiasis in approximately 112 million people, primarily in sub-Saharan Africa and the Middle East [1]. During infection, S. haematobium worms lay highly inflammatory eggs in the bladder wall. This inflammation is thought to promote carcinogenesis through unclear mechanisms. People with chronic urogenital schistosomiasis exhibit increased risk and earlier onset of bladder cancer (up to two decades earlier), with a predominance of squamous cell carcinoma [2]. Consequently, S. haematobium has been categorized as a Group I carcinogen (“carcinogenic to humans”) by the International Agency on Research on Cancer of the World Health Organization [3].


PLOS Neglected Tropical Diseases | 2014

A new mouse model for female genital schistosomiasis.

Monica L. Richardson; Chi-Ling Fu; Luke Pennington; Jared Honeycutt; Justin L. Odegaard; Yi-Ju Hsieh; Olfat Hammam; Simon Conti; Michael H. Hsieh

Background Over 112 million people worldwide are infected with Schistosoma haematobium, one of the most prevalent schistosome species affecting humans. Female genital schistosomiasis (FGS) occurs when S. haematobium eggs are deposited into the female reproductive tract by adult worms, which can lead to pelvic pain, vaginal bleeding, genital disfigurement and infertility. Recent evidence suggests co-infection with S. haematobium increases the risks of contracting sexually transmitted diseases such as HIV. The associated mechanisms remain unclear due to the lack of a tractable animal model. We sought to create a mouse model conducive to the study of immune modulation and genitourinary changes that occur with FGS. Methods To model FGS in mice, we injected S. haematobium eggs into the posterior vaginal walls of 30 female BALB/c mice. A control group of 20 female BALB/c mice were injected with uninfected LVG hamster tissue extract. Histology, flow cytometry and serum cytokine levels were assessed at 2, 4, 6, and 8 weeks post egg injection. Voiding studies were performed at 1 week post egg injection. Results Vaginal wall injection with S. haematobium eggs resulted in synchronous vaginal granuloma development within 2 weeks post-egg injection that persisted for at least 6 additional weeks. Flow cytometric analysis of vaginal granulomata revealed infiltration by CD4+ T cells with variable expression of the HIV co-receptors CXCR4 and CCR5. Granulomata also contained CD11b+F4/80+ cells (macrophages and eosinophils) as well as CXCR4+MerTK+ macrophages. Strikingly, vaginal wall-injected mice featured significant urinary frequency despite the posterior vagina being anatomically distant from the bladder. This may represent a previously unrecognized overactive bladder response to deposition of schistosome eggs in the vagina. Conclusion We have established a new mouse model that could potentially enable novel studies of genital schistosomiasis in females. Ongoing studies will further explore the mechanisms by which HIV target cells may be drawn into FGS-associated vaginal granulomata.


Cell Host & Microbe | 2017

Pseudogenization of the Secreted Effector Gene sseI Confers Rapid Systemic Dissemination of S. Typhimurium ST313 within Migratory Dendritic Cells

Sarah E. Carden; Gregory T. Walker; Jared Honeycutt; Kyler A. Lugo; Trung Pham; Amanda Jacobson; Donna M. Bouley; Juliana Idoyaga; Renée M. Tsolis; Denise M. Monack


Cell Host & Microbe | 2018

A Gut Commensal-Produced Metabolite Mediates Colonization Resistance to Salmonella Infection

Amanda Jacobson; Lilian H. Lam; Manohary Rajendram; Fiona B. Tamburini; Jared Honeycutt; Trung Pham; Will Van Treuren; Kali Pruss; Stephen Russell Stabler; Kyler A. Lugo; Donna M. Bouley; Jose G. Vilches-Moure; Mark Smith; Justin L. Sonnenburg; Ami S. Bhatt; Kerwyn Casey Huang; Denise M. Monack


The Journal of Urology | 2015

MP36-09 SCHISTOSOMA HAEMATOBIUM EGG-INDUCED BLADDER UROTHELIAL ABNORMALITIES ARE MODULATED BY P53 IN A GENDER-DEPENDENT FASHION

Jared Honeycutt; Olfat Hammam; Michael H. Hsieh


The Journal of Urology | 2014

MP21-03 EARLY DNA METHYLATION EVENTS IN BLADDER CARCINOGENESIS AND THEIR IMPLICATIONS FOR INFLAMMATION-INDUCED BLADDER CANCER

Simon Conti; Jared Honeycutt; Michael H. Hsieh

Collaboration


Dive into the Jared Honeycutt's collaboration.

Top Co-Authors

Avatar

Michael H. Hsieh

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Olfat Hammam

Theodor Bilharz Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge