Network


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

Hotspot


Dive into the research topics where Francis M. Hughes is active.

Publication


Featured researches published by Francis M. Hughes.


Fish & Shellfish Immunology | 2010

Apoptosis as a host defense mechanism in Crassostrea virginica and its modulation by Perkinsus marinus.

Francis M. Hughes; Brent Foster; Snimar Grewal; Inna M. Sokolova

Dermo disease caused by the obligatory intracellular protozoan Perkinsus marinus causes extensive oyster mortalities leading to tremendous losses in the oyster industry and damage to estuarine ecosystems. To better understand the mechanisms of the parasites evasion of the host immune defense system, we have investigated the molecular mechanisms of P.marinus-induced inhibition of apoptosis in oyster cells as a potential parasites survival strategy. We found that P. marinus modulates apoptosis of oyster immune cells (hemocytes) in a way that may help the parasite to establish infection. We found an increase in apoptosis in the initial stages of infection in vitro and in vivo, consistent with a host response to this intracellular parasite. During infection with highly virulent strains of P. marinus, this was followed by suppression and a return of apoptosis to basal levels 8-24 h post-infection, strongly indicating the parasite-induced inhibition of the immune response. In contrast, during infections with intermediate or low virulence strains of P. marinus, a transient suppression of apoptosis 4-8 h post-infection was followed by sustained elevation of hemocyte apoptosis at later stages, indicating that hemocytes were able to overcome the parasite-induced suppression and successfully combat the infection. Studies of the mechanisms of P. marinus-induced apoptosis indicated that the early post-infection stimulation of apoptosis is caspase-independent. However, this process can be driven (although to a lesser degree) by the killed parasite, suggesting that oyster hemocytes respond to cell surface molecules of P. marinus. Overall, this study provides novel insights into pathogen-induced modulation of apoptosis and its role in parasite virulence and establishment of infections.


American Journal of Physiology-renal Physiology | 2014

Inflammasomes are important mediators of cyclophosphamide-induced bladder inflammation

Francis M. Hughes; Nivardo Vivar; James Kennis; Jeffery D. Pratt-Thomas; Danielle W. Lowe; Brooke E. Shaner; Paul J. Nietert; Laura Spruill; J. Todd Purves

Bladder inflammation (cystitis) underlies numerous bladder pathologies and is elicited by a plethora of agents such as urinary tract infections, bladder outlet obstruction, chemotherapies, and catheters. Pattern recognition receptors [Toll-like receptors (TLRs) and Nod-like receptors (NLRs)] that recognize pathogen- and/or damage-associated molecular patterns (PAMPs and/or DAMPs, respectively) are key components of the innate immune system that coordinates the production (TLRs) and maturation (NLRs) of proinflammatory IL-1β. Despite multiple studies of TLRs in the bladder, none have investigated NLRs beyond one small survey. We now demonstrate that NLRP3 and NLRC4, and their binding partners apoptosis-associated speck-like protein containing a COOH-terminal caspase recruitment domain (ASC) and NLR family apoptosis inhibitory protein (NAIP), are expressed in the bladder and localized predominantly to the urothelia. Activated NLRs form inflammasomes that activate caspase-1. Placement of a NLRP3- or NLRC4-activating PAMP or NLRP3-activating DAMPs into the lumen of the bladder stimulated caspase-1 activity. To investigate inflammasomes in vivo, we induced cystitis with cyclophosphamide (CP, 150 mg/kg ip) in the presence or absence of the inflammasome inhibitor glyburide. Glyburide completely blocked CP-induced activation of caspase-1 and the production of IL-1β at 4 h. At 24 h, glyburide reduced two markers of inflammation by 30-50% and reversed much of the inflammatory morphology. Furthermore, glyburide reversed changes in bladder physiology (cystometry) induced by CP. In conclusion, NLRs/inflammasomes are present in the bladder urothelia and respond to DAMPs and PAMPs, whereas NLRP3 inhibition blocks bladder dysfunction in the CP model. The coordinated response of NLRs and TLRs in the urothelia represents a first-line innate defense that may provide an important target for pharmacological intervention.


Journal of Medicinal Chemistry | 2010

Identification and Functional Characterization of a Stable, Centrally Active Derivative of the Neurotensin (8—13) Fragment as a Potential First-in-Class Analgesic

Francis M. Hughes; Brooke E. Shaner; Lisa A. May; Lyndsay Zotian; Justin O. Brower; R. Jeremy Woods; Michael Cash; Dustin Morrow; Fabienne Massa; Jean Mazella; Thomas A. Dix

The neurotensin hexapapetide fragment NT(8-13) is a potent analgesic when administered directly to the central nervous system but does not cross the blood-brain barrier. A total of 43 novel derivatives of NT(8-13) were evaluated, with one, ABS212 (1), being most active in four rat models of pain when administered peripherally. Compound 1 binds to human neurotensin receptors 1 and 2 with IC(50) of 10.6 and 54.2 nM, respectively, and tolerance to the compound in a rat pain model did not develop after 12 days of daily administration. When it was administered peripherally, serum levels and neurotensin receptor binding potency of 1 peaked within 5 min and returned to baseline within 90-120 min; however, analgesic activity remained near maximum for >240 min. This could be due to its metabolism into an active fragment; however, all 4- and 5-mer hydrolysis products were inactive. This pharmacokinetic/pharmacodynamic dichotomy is discussed. Compound 1 is a candidate for development as a first-in-class analgesic.


BJUI | 2015

A novel urodynamic model for lower urinary tract assessment in awake rats

Marc P. Schneider; Francis M. Hughes; Anne K. Engmann; J. Todd Purves; Hansjörg Kasper; Marco Tedaldi; Laura Spruill; Miriam Gullo; Martin E. Schwab; Thomas M. Kessler

To develop a urodynamic model incorporating external urethral sphincter (EUS) electromyography (EMG) in awake rats.


The Journal of Urology | 2016

The NLRP3 Inflammasome Mediates Inflammation Produced by Bladder Outlet Obstruction

Francis M. Hughes; Hayden M. Hill; Case M. Wood; Andrew T. Edmondson; Aliya Dumas; Wen-Chi Foo; James M. Oelsen; Goran Rac; J. Todd Purves

PURPOSE While bladder outlet obstruction is well established to elicit an inflammatory reaction in the bladder that leads to overactive bladder and fibrosis, little is known about the mechanism by which this is initiated. NLRs (NOD-like receptors) and the structures that they form (inflammasomes) have been identified as sensors of cellular damage, including pressure induced damage, and triggers of inflammation. Recently we identified these structures in the urothelium. In this study we assessed the role of the NLRP3 (NACHT, LRR and PYD domains-containing protein 3) inflammasome in bladder dysfunction resulting from bladder outlet obstruction. MATERIALS AND METHODS Bladder outlet obstruction was created in female rats by inserting a 1 mm outer diameter transurethral catheter, tying a silk ligature around the urethra and removing the catheter. Untreated and sham operated rats served as controls. Rats with bladder outlet obstruction were given vehicle (10% ethanol) or 10 mg/kg glyburide (a NLRP3 inhibitor) orally daily for 12 days. Inflammasome activity, bladder hypertrophy, inflammation and bladder function (urodynamics) were assessed. RESULTS Bladder outlet obstruction increased urothelial inflammasome activity, bladder hypertrophy and inflammation, and decreased voided volume. Glyburide blocked inflammasome activation, reduced hypertrophy and prevented inflammation. The decrease in voided volume was also attenuated by glyburide mechanistically as an increase in detrusor contraction duration and voiding period. CONCLUSION Results suggest the importance of the NLRP3 inflammasome in the induction of inflammation and bladder dysfunction secondary to bladder outlet obstruction. Arresting these processes with NLRP3 inhibitors may prove useful to treat the symptoms that they produce.


Neurourology and Urodynamics | 2015

S-Nitrosoglutathione Protects the Spinal Bladder: Novel Therapeutic Approach to Post-Spinal Cord Injury Bladder Remodeling

Anandakumar Shunmugavel; Mushfiquddin Khan; Francis M. Hughes; J. Todd Purves; Avtar K. Singh; Inderjit Singh

Bladder and renal dysfunction are secondary events of the inflammatory processes induced by spinal cord injury (SCI). S‐Nitrosoglutathione (GSNO), an endogenous nitrosylating agent is pleiotropic and has anti‐inflammatory property. Hence, GSNO ameliorates inflammatory sequelae observed in bladder and renal tissues after SCI. Thus, we postulate that GSNO will improve the recovery of micturition dysfunction by quenching the bladder tissue inflammation associated with SCI.


The Open Medicinal Chemistry Journal | 2013

Development of a Peptide-derived orally-active kappa-opioid receptor agonist targeting peripheral pain.

Francis M. Hughes; Brooke E. Shaner; Justin O. Brower; R. Jeremy Woods; Thomas A. Dix

Kappa-opioid agonists are particularly efficacious in the treatment of peripheral pain but suffer from central nervous system (CNS)-mediated effects that limit their development. One promising kappa-agonist is the peptidic compound CR665. Although not orally available, CR665 given i.v. exhibits high peripheral to CNS selectivity and benefits patients with visceral and neuropathic pain. In this study we have generated a series of derivatives of CR665 and screened them for oral activity in the acetic acid-induced rat writhing assay for peripheral pain. Five compounds were further screened for specificity of activation of kappa receptors as well as agonism and antagonism at mu and delta receptors, which can lead to off-target effects. All active derivatives engaged the kappa receptor with EC50s in the low nM range while agonist selectivity for kappa over mu or delta was >11,000-200,000-fold. No antagonist activity was detected. One compound was chosen for further analysis (Compound 9). An oral dose response of 9 in rats yielded an EC50 of 4.7 mg/kg, approaching a druggable level for an oral analgesic. To assess the peripheral selectivity of this compound an i.v. dose response in rats was assessed in the writhing assay and hotplate assay (an assay of CNS-mediated pain). The EC50 in the writhing assay was 0.032 mg/kg while no activity was detectable in the hotplate assay at doses as high as 30 mg/kg, indicating a peripheral selectivity of >900-fold. We propose that compound 9 is a candidate for development as an orally-available peripherally-restricted kappa agonist.


Journal of clinical & cellular immunology | 2016

The NACHT, LRR and PYD Domains-Containing protein 3 (NLRP3) Inflammasome Mediates Inflammation and Voiding Dysfunction in a Lipopolysaccharide-Induced Rat Model of Cystitis

Francis M. Hughes; James Kennis; Melissa N Youssef; Danielle W. Lowe; Brooke E. Shaner; J. Todd Purves

OBJECTIVE NOD-like receptors (NLRs) sense sterile and non-sterile signals and form inflammasomes which trigger an inflammatory response through the activation of caspase-1 and release of IL-1β. Recently we have shown the presence of several NLRs in the bladder urothelia and demonstrated the importance of NLRP3 in bladder outlet obstruction and cyclophosphamide-induced cystitis, both models of sterile inflammation. In this study we explore a role for NLRP3 in mediating the response to LPS, a key antigen of uropathogenic bacteria. METHOD In order to bypass the protective glycosaminoglycan layer lining the urothelium, LPS was directly injected into the bladder wall of Sprague-Dawley rats. Glyburide (a NLRP3 inhibitor) or vehicle was administered orally prior to and after injection. Rats were analyzed 24 h later. Inflammasome activity (caspase-1 activity, IL-1β release) and inflammation (Evans Blue extravasation, bladder weight) were assessed, as was physiological bladder function (urodynamics). RESULTS Injection of LPS stimulated inflammasome activation (caspase-1 activity) and the release of IL-1β into the urine which was prevented by glyburide. Likewise, LPS increased inflammation, (bladder weight and the extravasation of Evans blue dye), and this was reversed by glyburide. Functionally, animals injected with saline alone demonstrated decreased voiding volume as measured by urodynamics. In the presence of LPS, additional urinary dysfunction was evident with decreased voiding pressures and threshold pressures. The decrease in voiding pressure was blocked by glyburide but the decrease in threshold pressure was not, suggesting that LPS has significant effects mediated by inflammasome-dependent and -independent mechanisms. CONCLUSION Overall, the results demonstrate the potential importance of inflammasomes in bacterial cystitis as well as the ability of the bladder wall injection technique to isolate the in vivo effects of specific inflammasome ligands to the physiological changes associated with cystitis.


International Urology and Nephrology | 2015

The potential repertoire of the innate immune system in the bladder: expression of pattern recognition receptors in the rat bladder and a rat urothelial cell line (MYP3 cells)

Francis M. Hughes; David P. Turner; J. Todd Purves

AbstractPurpose The urothelium is a frontline sensor of the lower urinary tract, sampling the bladder lumen and stimulating an immune response to infectious and noxious agents. Pattern recognition receptors (PRRs) recognize such agents and coordinate the innate response, often by forming inflammasomes that activate caspase-1 and the release of interleukin-1. We have shown the presence of one PRR (NLRP3) in the urothelia and its central role in the inflammatory response to cyclophosphamide. The purpose of this study was to (1) assess the likely range of the PPR response by assessing the repertoire present in the rat bladder and (2) determine the utility of the MYP3 rat urothelia cell line for in vitro studies by assessing its PPR repertoire and functional responsiveness.MethodsImmunohistochemistry was performed for seven PPRs (NLRP1, NLRP3, NLRP6, NLRP7, NLRP12, NLRC4 and AIM2) on bladder sections and MYP3 cells. For functionality, MYP3 cells were challenged with the quintessential NLRP3 activator ATP and assessed for caspase-1 activation.ResultsAll PPRs examined were expressed in the bladder and localized to the urothelial layer with several also in the detrusor (none in the interstitia). MYP3 cells also expressed all PRRs with a variable intracellular location. ATP-stimulated caspase-1 activity in MYP3 cells in a dose-dependent manner was reduced by knockdown of NLRP3 expression.ConclusionThe results suggest that the bladder possesses the capacity to initiate an innate immune response to a wide array of uropathological agents and the MYP3 cells will provide an excellent investigational tool for this field.


American Journal of Physiology-renal Physiology | 2017

Bladder fibrosis during outlet obstruction is triggered through the NLRP3 inflammasome and the production of IL-1β.

Francis M. Hughes; Stephanie J. Sexton; Huixia Jin; Vihasa Govada; J. Todd Purves

Bladder outlet obstruction (BOO) triggers inflammation in the bladder through the NLRP3 inflammasome. BOO also activates fibrosis, which is largely responsible for the decompensation of the bladder in the chronic state. Because fibrosis can be driven by inflammation, we have explored a role for NLRP3 (and IL-1β produced by NLRP3) in the activation and progression of BOO-induced fibrosis. Female rats were divided into five groups: 1) control, 2) sham, 3) BOO + vehicle, 4) BOO + the NLRP3 inhibitor glyburide, or 5) BOO + the IL-1β receptor antagonist anakinra. Fibrosis was assessed by Massons trichrome stain, collagen secretion via Sirius Red, and protein localization by immunofluorescence. BOO increased collagen production in the bladder, which was blocked by glyburide and anakinra, clearly implicating the NLRP3/IL-1β pathway in fibrosis. The collagen was primarily found in the lamina propria and the smooth muscle, while IL-1 receptor 1 and prolyl 4-hydroylase (an enzyme involved in the intracellular modification of collagen) both localized to the urothelium and the smooth muscle. Lysyl oxidase, the enzyme involved in the final extracellular assembly of mature collagen fibrils, was found to some extent in the lamina propria where its expression was greatly enhanced during BOO. In vitro studies demonstrated isolated urothelial cells from BOO rats secreted substantially more collagen than controls, and collagen expression in control cultures could be directly stimulated by IL-1β. In summary, NLRP3-derived-IL-1β triggers fibrosis during BOO, most likely through an autocrine loop in which IL-1β acts on urothelia to drive collagen production.

Collaboration


Dive into the Francis M. Hughes's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian M. Inouye

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brooke E. Shaner

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Laura Spruill

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Danielle W. Lowe

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

James Kennis

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge