Network


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

Hotspot


Dive into the research topics where Melissa A. Farrow is active.

Publication


Featured researches published by Melissa A. Farrow.


PLOS Pathogens | 2012

Clostridium difficile Toxin B Causes Epithelial Cell Necrosis through an Autoprocessing-Independent Mechanism

Nicole M. Chumbler; Melissa A. Farrow; Lynne A. Lapierre; Jeffrey L. Franklin; David B. Haslam; James R. Goldenring; D. Borden Lacy

Clostridium difficile is the most common cause of antibiotic-associated nosocomial infection in the United States. C. difficile secretes two homologous toxins, TcdA and TcdB, which are responsible for the symptoms of C. difficile associated disease. The mechanism of toxin action includes an autoprocessing event where a cysteine protease domain (CPD) releases a glucosyltransferase domain (GTD) into the cytosol. The GTD acts to modify and inactivate Rho-family GTPases. The presumed importance of autoprocessing in toxicity, and the apparent specificity of the CPD active site make it, potentially, an attractive target for small molecule drug discovery. In the course of exploring this potential, we have discovered that both wild-type TcdB and TcdB mutants with impaired autoprocessing or glucosyltransferase activities are able to induce rapid, necrotic cell death in HeLa and Caco-2 epithelial cell lines. The concentrations required to induce this phenotype correlate with pathology in a porcine colonic explant model of epithelial damage. We conclude that autoprocessing and GTD release is not required for epithelial cell necrosis and that targeting the autoprocessing activity of TcdB for the development of novel therapeutics will not prevent the colonic tissue damage that occurs in C. difficile – associated disease.


Nature | 2015

Cloning and variation of ground state intestinal stem cells

Xia Wang; Yusuke Yamamoto; Lane Wilson; Ting Zhang; Brooke E. Howitt; Melissa A. Farrow; Florian Kern; Gang Ning; Yue Hong; Chiea Chuen Khor; Benoit Chevalier; Denis Bertrand; Lingyan Wu; Niranjan Nagarajan; Francisco A. Sylvester; Jeffrey S. Hyams; Thomas J. Devers; Roderick T. Bronson; D. Borden Lacy; Khek Yu Ho; Christopher P. Crum; Frank McKeon; Wa Xian

Stem cells of the gastrointestinal tract, pancreas, liver and other columnar epithelia collectively resist cloning in their elemental states. Here we demonstrate the cloning and propagation of highly clonogenic, ‘ground state’ stem cells of the human intestine and colon. We show that derived stem-cell pedigrees sustain limited copy number and sequence variation despite extensive serial passaging and display exquisitely precise, cell-autonomous commitment to epithelial differentiation consistent with their origins along the intestinal tract. This developmentally patterned and epigenetically maintained commitment of stem cells is likely to enforce the functional specificity of the adult intestinal tract. Using clonally derived colonic epithelia, we show that toxins A or B of the enteric pathogen Clostridium difficile recapitulate the salient features of pseudomembranous colitis. The stability of the epigenetic commitment programs of these stem cells, coupled with their unlimited replicative expansion and maintained clonogenicity, suggests certain advantages for their use in disease modelling and regenerative medicine.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Clostridium difficile toxin B-induced necrosis is mediated by the host epithelial cell NADPH oxidase complex

Melissa A. Farrow; Nicole M. Chumbler; Lynne A. Lapierre; Jeffrey L. Franklin; Stacey A. Rutherford; James R. Goldenring; D. Borden Lacy

Significance The toxin-producing bacterium Clostridium difficile is the leading cause of antibiotic-associated infection in hospitals worldwide. An estimated 500,000 cases of C. difficile infection (CDI) occur annually in the US, with a cost approaching 3 billion dollars. The principle virulence factors in C. difficile pathogenesis are TcdA and TcdB, two large homologous toxins capable of entering host cells to cause fluid secretion, inflammation, and necrosis of the colonic mucosa. Given that numerous investigators are currently targeting these toxins for the development of novel CDI therapies, understanding the mechanism of toxicity is a significant priority. Clostridium difficile infection (CDI) is a leading cause of health care-associated diarrhea and has increased in incidence and severity over the last decade. Pathogenesis is mediated by two toxins, TcdA and TcdB, which cause fluid secretion, inflammation, and necrosis of the colonic mucosa. TcdB is a potent cytotoxin capable of inducing enzyme-independent necrosis in both cells and tissue. In this study, we show that TcdB-induced cell death depends on assembly of the host epithelial cell NADPH oxidase (NOX) complex and the production of reactive oxygen species (ROS). Treating cells with siRNAs directed against key components of the NOX complex, chemical inhibitors of NOX function, or molecules that scavenge superoxide or ROS confers protection against toxin challenge. To test the hypothesis that chemical inhibition of TcdB-induced cytotoxicity can protect against TcdB-induced tissue damage, we treated colonic explants with diphenyleneiodonium (DPI), a flavoenzyme inhibitor, or N-acetylcysteine (NAC), an antioxidant. TcdB-induced ROS production in colonic tissue was inhibited with DPI, and both DPI and NAC conferred protection against TcdB-induced tissue damage. The efficacy of DPI and NAC provides proof of concept that chemical attenuation of ROS could serve as a viable strategy for protecting the colonic mucosa of patients with CDI.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Identification of an epithelial cell receptor responsible for Clostridium difficile TcdB-induced cytotoxicity

Michelle E. LaFrance; Melissa A. Farrow; Ramyavardhanee Chandrasekaran; Jinsong Sheng; Donald H. Rubin; D. Borden Lacy

Significance Clostridium difficile is a toxin-producing bacterium that is a frequent cause of hospital-acquired and antibiotic-associated diarrhea. The incidence, severity, and costs associated with C. difficile infection (CDI) are increasing, making C. difficile an important public health concern. As a toxin-mediated disease, there is significant interest in understanding the receptors that mediate the cellular entry and function of these toxins. The targeted disruption of toxin-receptor interactions could provide novel therapeutic strategies that can either augment or replace the need for antibiotic therapies in the treatment of CDI. Clostridium difficile is the leading cause of hospital-acquired diarrhea in the United States. The two main virulence factors of C. difficile are the large toxins, TcdA and TcdB, which enter colonic epithelial cells and cause fluid secretion, inflammation, and cell death. Using a gene-trap insertional mutagenesis screen, we identified poliovirus receptor-like 3 (PVRL3) as a cellular factor necessary for TcdB-mediated cytotoxicity. Disruption of PVRL3 expression by gene-trap mutagenesis, shRNA, or CRISPR/Cas9 mutagenesis resulted in resistance of cells to TcdB. Complementation of the gene-trap or CRISPR mutants with PVRL3 resulted in restoration of TcdB-mediated cell death. Purified PVRL3 ectodomain bound to TcdB by pull-down. Pretreatment of cells with a monoclonal antibody against PVRL3 or prebinding TcdB to PVRL3 ectodomain also inhibited cytotoxicity in cell culture. The receptor is highly expressed on the surface epithelium of the human colon and was observed to colocalize with TcdB in both an explant model and in tissue from a patient with pseudomembranous colitis. These data suggest PVRL3 is a physiologically relevant binding partner that can serve as a target for the prevention of TcdB-induced cytotoxicity in C. difficile infection.


Journal of Biological Chemistry | 2012

Structural determinants of Clostridium difficile toxin A glucosyltransferase activity.

Rory N. Pruitt; Nicole M. Chumbler; Stacey A. Rutherford; Melissa A. Farrow; David B. Friedman; Ben Spiller; D. Borden Lacy

Background: C. difficile TcdA and TcdB glucosylate small GTPases. Results: Structural and functional studies reveal comparable activities with Rho substrates, enhanced activities following autoprocessing, and TcdA-specific modification of Rap2A. Conclusion: TcdA is a potent enzyme and modifies a broader array of GTPase substrates than TcdB. Significance: These findings highlight the importance of autoprocessing for activity and reveal differences in target specificity between the toxins. The principle virulence factors in Clostridium difficile pathogenesis are TcdA and TcdB, homologous glucosyltransferases capable of inactivating small GTPases within the host cell. We present crystal structures of the TcdA glucosyltransferase domain in the presence and absence of the co-substrate UDP-glucose. Although the enzymatic core is similar to that of TcdB, the proposed GTPase-binding surface differs significantly. We show that TcdA is comparable with TcdB in its modification of Rho family substrates and that, unlike TcdB, TcdA is also capable of modifying Rap family GTPases both in vitro and in cells. The glucosyltransferase activities of both toxins are reduced in the context of the holotoxin but can be restored with autoproteolytic activation and glucosyltransferase domain release. These studies highlight the importance of cellular activation in determining the array of substrates available to the toxins once delivered into the cell.


Nature microbiology | 2016

Crystal structure of Clostridium difficile toxin A

Nicole M. Chumbler; Stacey A. Rutherford; Zhifen Zhang; Melissa A. Farrow; John P. Lisher; Erik Farquhar; David P. Giedroc; Benjamin W. Spiller; Roman A. Melnyk; D. Borden Lacy

Clostridium difficile infection is the leading cause of hospital-acquired diarrhoea and pseudomembranous colitis. Disease is mediated by the actions of two toxins, TcdA and TcdB, which cause the diarrhoea, as well as inflammation and necrosis within the colon1,2. The toxins are large (308 and 270 kDa, respectively), homologous (47% amino acid identity) glucosyltransferases that target small GTPases within the host3,4. The multidomain toxins enter cells by receptor-mediated endocytosis and, upon exposure to the low pH of the endosome, insert into and deliver two enzymatic domains across the membrane. Eukaryotic inositol-hexakisphosphate (InsP6) binds an autoprocessing domain to activate a proteolysis event that releases the N-terminal glucosyltransferase domain into the cytosol. Here, we report the crystal structure of a 1,832-amino-acid fragment of TcdA (TcdA1832), which reveals a requirement for zinc in the mechanism of toxin autoprocessing and an extended delivery domain that serves as a scaffold for the hydrophobic α-helices involved in pH-dependent pore formation. A surface loop of the delivery domain whose sequence is strictly conserved among all large clostridial toxins is shown to be functionally important, and is highlighted for future efforts in the development of vaccines and novel therapeutics.


Journal of Biological Chemistry | 2011

NFAT and IRF Proteins Regulate Transcription of the Anti-HIV Gene, APOBEC3G

Melissa A. Farrow; Eun Young Kim; Steven M. Wolinsky; Ann M. Sheehy

The human cytidine deaminase APOBEC3G (A3G) is an innate restriction factor that inhibits human immunodeficiency virus, type 1 (HIV-1) replication. Regulation of A3G gene expression plays an important role in this suppression. Currently, an understanding of the mechanism of this gene regulation is largely unknown. Here, we have identified and characterized a TATA-less core promoter with an NFAT/IRF-4 composite binding site that confers cell type-specific transcriptional regulation. We found that A3G expression is critically dependent on NFATc1/NFATc2 and IRF-4. When either NFATc1 or NFATc2 and IRF-4 were co-expressed, A3G promoter activity was observed in cells that normally lack A3G expression and expression was not detected in the presence of the individual factors. This induced A3G expression allowed normally permissive CEMss cells to adopt a nonpermissive state, able to resist an HIV-1Δvif challenge. This represents the first reporting of manipulating the restrictive state of a cell type via gene regulation. Identification of NFAT and IRF family members as critical regulators of A3G expression offers important insight into the transcriptional control mechanisms that regulate innate immune responses and identifies specific targets for therapeutic intervention aimed at effectively boosting our natural immunity, in the form of a host defensive factor, against HIV-1.


Future Microbiology | 2008

Vif and Apobec3G in the innate immune response to HIV: a tale of two proteins

Melissa A. Farrow; Ann M. Sheehy

It is now 26 years after the first published report on HIV, and the global epidemic continues unabated, with estimates of over 33 million people currently infected, worldwide. Development of targeted therapies aimed at perturbing the HIV life cycle can be achieved only with a detailed comprehension of the dynamics of virus-host interactions within the cell. One such critical virus-host interaction is the recently elucidated interplay between the viral Vif protein and the innate immune defense molecule Apobec3G. Apobec3G potently suppresses HIV replication, but Vif can alleviate this inhibition, rescuing viral infectivity. Early work describing the characterization of Vif and the cloning and identification of Apobec3G as an antiviral are discussed. Recent advances detailing the mechanisms of the Vif-Apobec3G regulatory circuit and our nascent understanding of Apobec3G endogenous function are also presented. Collectively, these studies have shed light on potential novel therapeutic strategies aimed at exploiting Apobec3G antiviral function to abrogate HIV replication.


Infection and Immunity | 2016

Clostridium difficile Toxins TcdA and TcdB Cause Colonic Tissue Damage by Distinct Mechanisms

Nicole M. Chumbler; Melissa A. Farrow; Lynne A. Lapierre; Jeffrey L. Franklin; D. Borden Lacy

ABSTRACT As the major cause of antibiotic-associated diarrhea, Clostridium difficile is a serious problem in health care facilities worldwide. C. difficile produces two large toxins, TcdA and TcdB, which are the primary virulence factors in disease. The respective functions of these toxins have been difficult to discern, in part because the cytotoxicity profiles for these toxins differ with concentration and cell type. The goal of this study was to develop a cell culture model that would allow a side-by-side mechanistic comparison of the toxins. Conditionally immortalized, young adult mouse colonic (YAMC) epithelial cells demonstrate an exquisite sensitivity to both toxins with phenotypes that agree with observations in tissue explants. TcdA intoxication results in an apoptotic cell death that is dependent on the glucosyltransferase activity of the toxin. In contrast, TcdB has a bimodal mechanism; it induces apoptosis in a glucosyltransferase-dependent manner at lower concentrations and glucosyltransferase-independent necrotic death at higher concentrations. The direct comparison of the responses to TcdA and TcdB in cells and colonic explants provides the opportunity to unify a large body of observations made by many independent investigators.


Journal of Proteome Research | 2017

Integrated, High-Throughput, Multiomics Platform Enables Data-Driven Construction of Cellular Responses and Reveals Global Drug Mechanisms of Action

Jeremy L. Norris; Melissa A. Farrow; Danielle B. Gutierrez; Lauren D. Palmer; Nicole Muszynski; Stacy D. Sherrod; James C. Pino; Jamie L. Allen; Jeffrey M. Spraggins; Alex L.R. Lubbock; Ashley T Jordan; William J. Burns; James C Poland; Carrie E. Romer; M. Lisa Manier; Yuan-Wei Nei; Boone M. Prentice; Kristie L. Rose; Salisha Hill; Raf Van de Plas; Tina Tsui; Nathaniel M. Braman; M Ray Keller; Stacey A. Rutherford; Nichole A Lobdell; Carlos F. Lopez; D. Borden Lacy; John A. McLean; John P. Wikswo; Eric P. Skaar

An understanding of how cells respond to perturbation is essential for biological applications; however, most approaches for profiling cellular response are limited in scope to pre-established targets. Global analysis of molecular mechanism will advance our understanding of the complex networks constituting cellular perturbation and lead to advancements in areas, such as infectious disease pathogenesis, developmental biology, pathophysiology, pharmacology, and toxicology. We have developed a high-throughput multiomics platform for comprehensive, de novo characterization of cellular mechanisms of action. Platform validation using cisplatin as a test compound demonstrates quantification of over 10 000 unique, significant molecular changes in less than 30 days. These data provide excellent coverage of known cisplatin-induced molecular changes and previously unrecognized insights into cisplatin resistance. This proof-of-principle study demonstrates the value of this platform as a resource to understand complex cellular responses in a high-throughput manner.

Collaboration


Dive into the Melissa A. Farrow's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stacey A. Rutherford

University of Texas Health Science Center at Tyler

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brooke E. Howitt

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Carlos F. Lopez

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Christopher P. Crum

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge