Network


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

Hotspot


Dive into the research topics where William S. Lawrence is active.

Publication


Featured researches published by William S. Lawrence.


Circulation Research | 2012

Role of RyR2 Phosphorylation at S2814 during Heart Failure Progression

Jonathan L. Respress; Ralph J. van Oort; Na Li; Natale Rolim; Sayali S. Dixit; Angela deAlmeida; Niels Voigt; William S. Lawrence; Darlene G. Skapura; Kristine Skårdal; Ulrik Wisløff; Thomas Wieland; Xun Ai; Steven M. Pogwizd; Dobromir Dobrev; Xander H.T. Wehrens

Rationale: Increased activity of Ca2+/calmodulin-dependent protein kinase II (CaMKII) is thought to promote heart failure (HF) progression. However, the importance of CaMKII phosphorylation of ryanodine receptors (RyR2) in HF development and associated diastolic sarcoplasmic reticulum Ca2+ leak is unclear. Objective: Determine the role of CaMKII phosphorylation of RyR2 in patients and mice with nonischemic and ischemic forms of HF. Methods and Results: Phosphorylation of the primary CaMKII site S2814 on RyR2 was increased in patients with nonischemic, but not with ischemic, HF. Knock-in mice with an inactivated S2814 phosphorylation site were relatively protected from HF development after transverse aortic constriction compared with wild-type littermates. After transverse aortic constriction, S2814A mice did not exhibit pulmonary congestion and had reduced levels of atrial natriuretic factor. Cardiomyocytes from S2814A mice exhibited significantly lower sarcoplasmic reticulum Ca2+ leak and improved sarcoplasmic reticulum Ca2+ loading compared with wild-type mice after transverse aortic constriction. Interestingly, these protective effects on cardiac contractility were not observed in S2814A mice after experimental myocardial infarction. Conclusions: Our results suggest that increased CaMKII phosphorylation of RyR2 plays a role in the development of pathological sarcoplasmic reticulum Ca2+ leak and HF development in nonischemic forms of HF such as transverse aortic constriction in mice.


The Open Microbiology Journal | 2010

Protection Afforded by Fluoroquinolones in Animal Models of Respiratory Infections with Bacillus anthracis, Yersinia pestis, and Francisella tularensis

Johnny W. Peterson; Scott T. Moen; Daniel P. Healy; Jennifer Pawlik; Joanna Taormina; Jason Hardcastle; John M. Thomas; William S. Lawrence; Cindy Ponce; Bagram M. Chatuev; Bryan T. Gnade; Sheri M. Foltz; Stacy L. Agar; Jian Sha; Gary R. Klimpel; Michelle L. Kirtley; Tonyia Eaves-Pyles; Ashok K. Chopra

Successful treatment of inhalation anthrax, pneumonic plague and tularemia can be achieved with fluoroquinolone antibiotics, such as ciprofloxacin and levofloxacin, and initiation of treatment is most effective when administered as soon as possible following exposure. Bacillus anthracis Ames, Yersinia pestis CO92, and Francisella tularensis SCHU S4 have equivalent susceptibility in vitro to ciprofloxacin and levofloxacin (minimal inhibitory concentration is 0.03 μg/ml); however, limited information is available regarding in vivo susceptibility of these infectious agents to the fluoroquinolone antibiotics in small animal models. Mice, guinea pig, and rabbit models have been developed to evaluate the protective efficacy of antibiotic therapy against these life-threatening infections. Our results indicated that doses of ciprofloxacin and levofloxacin required to protect mice against inhalation anthrax were approximately 18-fold higher than the doses of levofloxacin required to protect against pneumonic plague and tularemia. Further, the critical period following aerosol exposure of mice to either B. anthracis spores or Y. pestis was 24 h, while mice challenged with F. tularensis could be effectively protected when treatment was delayed for as long as 72 h postchallenge. In addition, it was apparent that prolonged antibiotic treatment was important in the effective treatment of inhalation anthrax in mice, but short-term treatment of mice with pneumonic plague or tularemia infections were usually successful. These results provide effective antibiotic dosages in mice, guinea pigs, and rabbits and lay the foundation for the development and evaluation of combinational treatment modalities.


Hypertension | 2014

TWIK-2 channel deficiency leads to pulmonary hypertension through a rho-kinase-mediated process

Lavannya M. Pandit; Eric E. Lloyd; Julia Reynolds; William S. Lawrence; Corey Reynolds; Xander H.T. Wehrens; Robert M. Bryan

TWIK-2 (KCNK6) is a member of the 2-pore domain (K2P) family of potassium channels, which are highly expressed in the vascular system. We tested the hypothesis that TWIK-2 deficiency leads to pulmonary hypertension. TWIK-2 knockout mice and their wildtype littermates at 8 weeks of age had similar mean right ventricular systolic pressures (24±3 and 21±3 mm Hg, respectively.) Significantly, by 20 weeks of age, the mean right ventricular systolic pressures in TWIK-2 knockout mice increased to 35±3 mm Hg (P⩽0.036), whereas mean right ventricular systolic pressures in wildtype littermates remained at 22±3 mm Hg. Elevated mean right ventricular systolic pressures in the TWIK-2 knockout mice was accompanied by pulmonary vascular remodeling as determined by a 25% increase in the cross-sectional area of the vessels occupied by the vessel wall. Additionally, secondary branches of the pulmonary artery from 20-week-old TWIK-2 knockout mice showed an enhanced contractile response to U46619 (10–6 moles/L), a thromboxane A2 mimetic, which was completely abolished with the Rho-kinase inhibitor, Y27632 (10–6 and 10–5 moles/L). Treatment of TWIK-2 knockout mice with the Rho-kinase inhibitor, fasudil, in the drinking water for 12 weeks, abolished the development of pulmonary hypertension and attenuated the vessel remodeling. We concluded that mice deficient in the TWIK-2 channel develop pulmonary hypertension between 8 and 20 weeks of age through a mechanism involving Rho-kinase. Our results suggest that downregulation of TWIK-2 in the pulmonary vasculature may be an underlying mechanism in the development of pulmonary hypertension.


Toxins | 2011

Hemodynamic Effects of Anthrax Toxins in the Rabbit Model and the Cardiac Pathology Induced by Lethal Toxin

William S. Lawrence; Jeffrey R. Marshall; Diana Zavala; Lori E. Weaver; Wallace B. Baze; Scott T. Moen; Elbert B. Whorton; Randy L. Gourley; Johnny W. Peterson

Anthrax lethal toxin (LeTx) and edema toxin (EdTx) have been shown to alter hemodynamics in the rodent model, while LeTx primarily is reported to induce extensive tissue pathology. However, the rodent model has limitations when used for comparison to higher organisms such as humans. The rabbit model, on the other hand, has gained recognition as a useful model for studying anthrax infection and its pathophysiological effects. In this study, we assessed the hemodynamic effects of lethal toxin (LeTx) and edema toxin (EdTx) in the rabbit model using physiologically relevant amounts of the toxins. Moreover, we further examine the pathological effects of LeTx on cardiac tissue. We intravenously injected Dutch-belted rabbits with either low-dose and high-dose recombinant LeTx or a single dose of EdTx. The animals’ heart rate and mean arterial pressure were continuously monitored via telemetry until either 48 or 72 h post-challenge. Additional animals challenged with LeTx were used for cardiac troponin I (cTnI) quantitation, cardiac histopathology, and echocardiography. LeTx depressed heart rate at the lower dose and mean arterial pressure (MAP) at the higher dose. EdTx, on the other hand, temporarily intensified heart rate while lowering MAP. Both doses of LeTx caused cardiac pathology with the higher dose having a more profound effect. Lastly, left-ventricular dilation due to LeTx was not apparent at the given time-points. Our study demonstrates the hemodynamic effects of anthrax toxins, as well as the pathological effects of LeTx on the heart in the rabbit model, and it provides further evidence for the toxins’ direct impact on the heart.


Frontiers in Immunology | 2017

A bivalent anthrax-plague vaccine that can protect against two Tier-1 bioterror pathogens, Bacillus anthracis and Yersinia pestis

Pan Tao; Marthandan Mahalingam; Jingen Zhu; Mahtab Moayeri; Michelle L. Kirtley; Eric C. Fitts; Jourdan A. Andersson; William S. Lawrence; Stephen H. Leppla; Ashok K. Chopra; Venigalla B. Rao

Bioterrorism remains as one of the biggest challenges to global security and public health. Since the deadly anthrax attacks of 2001 in the United States, Bacillus anthracis and Yersinia pestis, the causative agents of anthrax and plague, respectively, gained notoriety and were listed by the CDC as Tier-1 biothreat agents. Currently, there is no Food and Drug Administration-approved vaccine against either of these threats for mass vaccination to protect general public, let alone a bivalent vaccine. Here, we report the development of a single recombinant vaccine, a triple antigen consisting of all three target antigens, F1 and V from Y. pestis and PA from B. anthracis, in a structurally stable context. Properly folded and soluble, the triple antigen retained the functional and immunogenicity properties of all three antigens. Remarkably, two doses of this immunogen adjuvanted with Alhydrogel® elicited robust antibody responses in mice, rats, and rabbits and conferred complete protection against inhalational anthrax and pneumonic plague. No significant antigenic interference was observed. Furthermore, we report, for the first time, complete protection of animals against simultaneous challenge with Y. pestis and the lethal toxin of B. anthracis, demonstrating that a single biodefense vaccine can protect against a bioterror attack with weaponized B. anthracis and/or Y. pestis. This bivalent anthrax–plague vaccine is, therefore, a strong candidate for stockpiling, after demonstration of its safety and immunogenicity in human clinical trials, as part of national preparedness against two of the deadliest bioterror threats.


Clinical and Vaccine Immunology | 2016

A Replication-Defective Human Type 5 Adenovirus-Based Trivalent Vaccine Confers Complete Protection against Plague in Mice and Nonhuman Primates

Jian Sha; Michelle L. Kirtley; Curtis Klages; Tatiana E. Erova; Maxim V. Telepnev; Duraisamy Ponnusamy; Eric C. Fitts; Wallace B. Baze; Satheesh K. Sivasubramani; William S. Lawrence; Igor Patrikeev; Jennifer E. Peel; Jourdan A. Andersson; Elena V. Kozlova; Bethany L. Tiner; Johnny W. Peterson; David McWilliams; Snehal Patel; Eric Rothe; Vladimir L. Motin; Ashok K. Chopra

ABSTRACT Currently, no plague vaccine exists in the United States for human use. The capsular antigen (Caf1 or F1) and two type 3 secretion system (T3SS) components, the low-calcium-response V antigen (LcrV) and the needle protein YscF, represent protective antigens of Yersinia pestis. We used a replication-defective human type 5 adenovirus (Ad5) vector and constructed recombinant monovalent and trivalent vaccines (rAd5-LcrV and rAd5-YFV) that expressed either the codon-optimized lcrV or the fusion gene designated YFV (consisting of ycsF, caf1, and lcrV). Immunization of mice with the trivalent rAd5-YFV vaccine by either the intramuscular (i.m.) or the intranasal (i.n.) route provided protection superior to that with the monovalent rAd5-LcrV vaccine against bubonic and pneumonic plague when animals were challenged with Y. pestis CO92. Preexisting adenoviral immunity did not diminish the protective response, and the protection was always higher when mice were administered one i.n. dose of the trivalent vaccine (priming) followed by a single i.m. booster dose of the purified YFV antigen. Immunization of cynomolgus macaques with the trivalent rAd5-YFV vaccine by the prime-boost strategy provided 100% protection against a stringent aerosol challenge dose of CO92 to animals that had preexisting adenoviral immunity. The vaccinated and challenged macaques had no signs of disease, and the invading pathogen rapidly cleared with no histopathological lesions. This is the first report showing the efficacy of an adenovirus-vectored trivalent vaccine against pneumonic plague in mouse and nonhuman primate (NHP) models.


BMC Research Notes | 2011

Telemetric left ventricular monitoring using wireless telemetry in the rabbit model

Mallory K Tate; William S. Lawrence; Randy L. Gourley; Diana Zavala; Lori E. Weaver; Scott T. Moen; Johnny W. Peterson

BackgroundHeart failure is a critical condition that affects many people and often results from left ventricular dysfunction. Numerous studies investigating this condition have been performed using various model systems. To do so, investigators must be able to accurately measure myocardial performance in order to determine the degree of left ventricular function. In this model development study, we employ a wireless telemetry system purchased from Data Sciences International to continuously assess left ventricular function in the rabbit model.FindingsWe surgically implanted pressure-sensitive catheters fitted to wireless radio-transmitters into the left ventricle of Dutch-belted rabbits. Following recovery of the animals, we continuously recorded indices of cardiac contractility and ventricular relaxation at baseline for a given time period. The telemetry system allowed us to continuously record baseline left ventricular parameters for the entire recording period. During this time, the animals were unrestrained and fully conscious. The values we recorded are similar to those obtained using other reported methods.ConclusionsThe wireless telemetry system can continuously measure left ventricular pressure, cardiac contractility, and cardiac relaxation in the rabbit model. These results, which were obtained just as baseline levels, substantiate the need for further validation in this model system of left ventricular assessment.


npj Vaccines | 2017

Protection against inhalation anthrax by immunization with Salmonella enterica serovar Typhi Ty21a stably producing protective antigen of Bacillus anthracis

B. Kim Lee Sim; Minglin Li; Manuel Osorio; Yun Wu; Tint T. Wai; Johnny W. Peterson; Eric R. James; Sumana Chakravarty; Lixin Gao; Rui Xu; Natasha Kc; Richard E. Stafford; William S. Lawrence; Linsey A. Yeager; Jennifer E. Peel; Satheesh K. Sivasubramani; Ashok K. Chopra; Svetlana Filippova; Stephen L. Hoffman

The national blueprint for biodefense concluded that the United States is underprepared for biological threats. The licensed anthrax vaccine absorbed vaccine, BioThrax, requires administration of at least 3–5 intramuscular doses. The anthrax vaccine absorbed vaccine consists of complex cell-free culture filtrates of a toxigenic Bacillus anthracis strain and causes tenderness at the injection site and significant adverse events. We integrated a codon-optimized, protective antigen gene of B. anthracis (plus extracellular secretion machinery), into the chromosome of the licensed, oral, live-attenuated typhoid fever vaccineTy21a to form Ty21a-PA-01 and demonstrated excellent expression of the gene encoding protective antigen. We produced the vaccine in a 10-L fermenter; foam-dried and vialed it, and characterized the dried product. The vaccine retained ~50% viability for 20 months at ambient temperature. Sera from animals immunized by the intraperitoneal route had high levels of anti-protective antigen antibodies by enzyme-linked immunosorbent assay and anthrax lethal toxin-neutralizing activity. Immunized mice were fully protected against intranasal challenge with ~5 LD50 of B. anthracis Sterne spores, and 70% (7/10) of vaccinated rabbits were protected against aerosol challenge with 200 LD50 of B. anthracis Ames spores. There was a significant correlation between protection and antibody levels determined by enzyme-linked immunosorbent assay and toxin-neutralizing activity. These data provide the foundation for achievement of our ultimate goal, which is to develop an oral anthrax vaccine that is stable at ambient temperatures and induces the rapid onset of durable, high-level protection after a 1-week immunization regimen.Counterterrorism: creating a safe, stable, easily stored anthrax vaccineA vaccine candidate for anthrax infection shows promise for improving preparedness for a biological attack. Bacillus anthracis, the bacterium responsible for anthrax is a top-tier bioterrorism agent due to its high lethality and spore stability. The current FDA-approved anthrax vaccine and other vaccine candidates in development lack ease of preparation, have short shelf lives and adverse effects. B. Kim Lee Sim of Protein Potential LLC and her collaborators combined key B. anthracis genetic material into an existing typhoid vaccine. The vaccine vector possesses high stability, a strong safety record, and offers long-term protection after oral administration, which Sim’s group hopes to preserve in their candidate anthrax vaccine. The team showed that their hybrid vaccine conferred excellent protection in rabbits and a short vaccination regimen, and suggest further studies into its suitability for human vaccine studies.


The Journal of Infectious Diseases | 2018

Experimental Infection of Syrian Hamsters With Aerosolized Nipah Virus

Olivier Escaffre; Terence E. Hill; Tetsuro Ikegami; Terry L. Juelich; Jennifer K. Smith; Lihong Zhang; David E Perez; Colm Atkins; Arnold Park; William S. Lawrence; Satheesh K. Sivasubramani; Jennifer E. Peel; Johnny W. Peterson; Benhur Lee; Alexander N. Freiberg

Background Nipah virus (NiV) is a paramyxovirus (genus Henipavirus) that can cause severe respiratory illness and encephalitis in humans. Transmission occurs through consumption of NiV-contaminated foods, and contact with NiV-infected animals or human body fluids. However, it is unclear whether aerosols derived from aforesaid sources or others also contribute to transmission, and current knowledge on NiV-induced pathogenicity after small-particle aerosol exposure is still limited. Methods Infectivity, pathogenicity, and real-time dissemination of aerosolized NiV in Syrian hamsters was evaluated using NiV-Malaysia (NiV-M) and/or its recombinant expressing firefly luciferase (rNiV-FlucNP). Results Both viruses had an equivalent pathogenicity in hamsters, which developed respiratory and neurological symptoms of disease, similar to using intranasal route, with no direct correlations to the dose. We showed that virus replication was predominantly initiated in the lower respiratory tract and, although delayed, also intensely in the oronasal cavity and possibly the brain, with gradual increase of signal in these regions until at least day 5-6 postinfection. Conclusion Hamsters infected with small-particle aerosolized NiV undergo similar clinical manifestations of the disease as previously described using liquid inoculum, and exhibit histopathological lesions consistent with NiV patient reports. NiV droplets could therefore play a role in transmission by close contact.


Comparative Medicine | 2009

The physiologic responses of Dutch belted rabbits infected with inhalational anthrax.

William S. Lawrence; Jason Hardcastle; Douglas Brining; Lori E. Weaver; Cindy Ponce; Elbert B. Whorton; Johnny W. Peterson

Collaboration


Dive into the William S. Lawrence's collaboration.

Top Co-Authors

Avatar

Johnny W. Peterson

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Ashok K. Chopra

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Scott T. Moen

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Cindy Ponce

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Jennifer E. Peel

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Jian Sha

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Lori E. Weaver

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Michelle L. Kirtley

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Satheesh K. Sivasubramani

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Wallace B. Baze

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge