Network


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

Hotspot


Dive into the research topics where Amanda H. Loftin is active.

Publication


Featured researches published by Amanda H. Loftin.


Journal of Orthopaedic Research | 2015

The role of chairman and research director in influencing scholarly productivity and research funding in academic orthopaedic surgery

Alexandra I. Stavrakis; Ankur D. Patel; Zachary D. C. Burke; Amanda H. Loftin; Erik M. Dworsky; Mauricio Silva; Nicholas M. Bernthal

The purpose of this study was to determine what orthopaedic surgery department leadership characteristics are most closely correlated with securing NIH funding and increasing scholarly productivity. Scopus database was used to identify number of publications/h‐index for 4,328 faculty, department chairs (DC), and research directors (RD), listed on departmental websites from 138 academic orthopaedic departments in the United States. NIH funding data was obtained for the 2013 fiscal year. While all programs had a DC, only 46% had a RD. Of


Frontiers of Medicine in China | 2015

Current Animal Models of Postoperative Spine Infection and Potential Future Advances

Alexandra I. Stavrakis; Amanda H. Loftin; Elizabeth L. Lord; Yan Hu; J. E. Manegold; Erik M. Dworsky; Anthony A. Scaduto; Nicholas M. Bernthal

54,925,833 in NIH funding allocated to orthopaedic surgery faculty in 2013, 3% of faculty and 31% of departments were funded. 16% of funded institutions had a funded DC whereas 65% had a funded RD. Department productivity and funding were highly correlated to leadership productivity and funding(p< 0.05). Mean funding was


Journal of Orthopaedic Research | 2017

Novel in vivo mouse model of implant related spine infection

Eric E M D Dworsky; Vishal Hegde; Amanda H. Loftin; Sherif S R Richman; Yan Y H Hu; Elizabeth L. Lord; Kevin P. Francis; Lloyd S. Miller; Jeffrey C. Wang; Anthony A. Scaduto; Nicholas M. Bernthal

1,700,000 for departments with a NIH‐funded RD,


Antimicrobial Agents and Chemotherapy | 2014

Combination Prophylactic Therapy with Rifampin Increases Efficacy against an Experimental Staphylococcus epidermidis Subcutaneous Implant-Related Infection

Alexandra I. Stavrakis; Jared A. Niska; Jonathan H. Shahbazian; Amanda H. Loftin; Romela Irene Ramos; Fabrizio Billi; Kevin P. Francis; Michael Otto; Nicholas M. Bernthal; Daniel Z. Uslan; Lloyd S. Miller

104,000 for departments with an unfunded RD, and


PLOS ONE | 2017

Combinatory antibiotic therapy increases rate of bacterial kill but not final outcome in a novel mouse model of Staphylococcus aureus spinal implant infection

Yan Hu; Vishal Hegde; Daniel Johansen; Amanda H. Loftin; Erik M. Dworsky; Stephen D. Zoller; Howard Y. Park; Christopher D. Hamad; George E. Nelson; Kevin P. Francis; Anthony A. Scaduto; Nicholas M. Bernthal

72,000 for departments with no RD. These findings suggest that orthopaedic department academic success is directly associated with scholarly productivity and funding of both DC and RD. The findings further highlight the correlation between a funded RD and a well‐funded department. This does not hold for an unfunded RD.


The Scientific World Journal | 2013

Understanding Infection: A Primer on Animal Models of Periprosthetic Joint Infection

Alexandra I. Stavrakis; Jared A. Niska; Amanda H. Loftin; Fabrizio Billi; Nicholas M. Bernthal

Implant related infection following spine surgery is a devastating complication for patients and can potentially lead to significant neurological compromise, disability, morbidity, and even mortality. This paper provides an overview of the existing animal models of postoperative spine infection and highlights the strengths and weaknesses of each model. In addition, there is discussion regarding potential modifications to these animal models to better evaluate preventative and treatment strategies for this challenging complication. Current models are effective in simulating surgical procedures but fail to evaluate infection longitudinally using multiple techniques. Potential future modifications to these models include using advanced imaging technologies to evaluate infection, use of bioluminescent bacterial species, and testing of novel treatment strategies against multiple bacterial strains. There is potential to establish a postoperative spine infection model using smaller animals, such as mice, as these would be a more cost-effective screening tool for potential therapeutic interventions.


Archive | 2016

Questions to Ask Your Medical Oncology Colleagues

Noah Federman; Amanda H. Loftin; Nicholas M. Bernthal

Post‐operative spine infections are a challenge, as hardware must often be retained to prevent destabilization of the spine, and bacteria form biofilm on implants, rendering them inaccessible to antibiotic therapy, and immune cells. A model of posterior‐approach spinal surgery was created in which a stainless steel k‐wire was transfixed into the L4 spinous process of 12‐week‐old C57BL/six mice. Mice were then randomized to receive either one of three concentrations (1 × 102, 1 × 103, and 1 × 104 colony forming units (CFU)) of a bioluminescent strain of Staphylococcus aureus or normal saline at surgery. The mice were then longitudinally imaged for bacterial bioluminescence to quantify infection. The 1 × 102 CFU group had a decrease in signal down to control levels by POD 25, while the 1 × 103 and 1 × 104 CFU groups maintained a 10‐fold higher signal through POD 35. Bacteria were then harvested from the pin and surrounding tissue for confirmatory CFU counts. All mice in the 1 × 104 CFU group experienced wound breakdown, while no mice in the other groups had this complication. Once an optimal bacterial concentration was determined, mice expressing enhanced green fluorescent protein in their myeloid cells (Lys‐EGFP) were utilized to contemporaneously quantify bacterial burden, and immune response. Neutrophil fluorescence peaked for both groups on POD 3, and then declined. The infected group continued to have a response above the control group through POD 35. This study, establishes a noninvasive in vivo mouse model of spine implant infection that can quantify bacterial burden and host inflammation longitudinally in real time without requiring animal sacrifice.


Global Dermatology | 2016

Academic characteristics for successful national institutes of health funding in dermatology

Ramtin Yaghoubnejad; Roy Akarakian; Amanda H. Loftin; Aram A. Namavar

ABSTRACT The incidence of infections related to cardiac devices (such as permanent pacemakers) has been increasing out of proportion to implantation rates. As management of device infections typically requires explantation of the device, optimal prophylactic strategies are needed. Cefazolin and vancomycin are widely used as single agents for surgical prophylaxis against cardiac device-related infections. However, combination antibiotic prophylaxis may further reduce infectious complications. To model a localized subcutaneous implant-related infection, a bioluminescent strain of Staphylococcus epidermidis was inoculated onto a medical-procedure-grade titanium disc, which was placed into a subcutaneous pocket in the backs of mice. In vivo bioluminescence imaging, quantification of ex vivo CFU from the capsules and implants, variable-pressure scanning electron microscopy (VP-SEM), and neutrophil enhanced green fluorescent protein (EGFP) fluorescence in LysEGFP mice were employed to monitor the infection. This model was used to evaluate the efficacies of low- and high-dose cefazolin (50 and 200 mg/kg of body weight) and vancomycin (10 and 110 mg/kg) intravenous prophylaxis with or without rifampin (25 mg/kg). High-dose cefazolin and high-dose vancomycin treatment resulted in almost complete bacterial clearance, whereas both low-dose cefazolin and low-dose vancomycin reduced the in vivo and ex vivo bacterial burden only moderately. The addition of rifampin to low-dose cefazolin and vancomycin was highly effective in further reducing the CFU harvested from the implants. However, vancomycin-rifampin was more effective than cefazolin-rifampin in further reducing the CFU harvested from the surrounding tissue capsules. Future studies in humans will be required to determine whether the addition of rifampin has improved efficacy in preventing device-related infections in clinical practice.


Journal of Bone and Joint Surgery, American Volume | 2016

In Vivo Efficacy of a “Smart” Antimicrobial Implant Coating

Alexandra I. Stavrakis; Suwei Zhu; Vishal Hegde; Amanda H. Loftin; Alyssa G. Ashbaugh; Jared A. Niska; Lloyd S. Miller; Tatiana Segura; Nicholas M. Bernthal

Background Management of spine implant infections (SII) are challenging. Explantation of infected spinal hardware can destabilize the spine, but retention can lead to cord compromise and biofilm formation, complicating management. While vancomycin monotherapy is commonly used, in vitro studies have shown reduced efficacy against biofilm compared to combination therapy with rifampin. Using an established in vivo mouse model of SII, we aim to evaluate whether combination therapy has increased efficacy compared to both vancomycin alone and infected controls. Methods An L-shaped, Kirschner-wire was transfixed into the L4 spinous process of 12-week-old C57BL/6 mice, and inoculated with bioluminescent Staphylococcus aureus. Mice were randomized into a vancomycin group, a combination group with vancomycin plus rifampin, or a control group receiving saline. Treatment began on post-operative day (POD) 7 and continued through POD 14. In vivo imaging was performed to monitor bioluminescence for 35 days. Colony-forming units (CFUs) were cultured on POD 35. Results Bioluminescence peaked around POD 7 for all groups. The combination group had a 10-fold decrease in signal by POD 10. The vancomycin and control groups reached similar levels on POD 17 and 21, respectively. On POD 25 the combination group dropped below baseline, but rebounded to the same level as the other groups, demonstrating a biofilm-associated infection by POD 35. Quantification of CFUs on POD 35 confirmed an ongoing infection in all three groups. Conclusions Although both therapies were initially effective, they were not able to eliminate implant biofilm bacteria, resulting in a rebound infection after antibiotic cessation. This model shows, for the first time, why histologic-based, static assessments of antimicrobials can be misleading, and the importance of longitudinal tracking of infection. Future studies can use this model to test combinations of antibiotic therapies to see if they are more effective in eliminating biofilm prior to human trials.


Journal of Bone and Joint Surgery, American Volume | 2017

Single-Dose, Preoperative Vitamin-D Supplementation Decreases Infection in a Mouse Model of Periprosthetic Joint Infection

Vishal Hegde; Erik M. Dworsky; Alexandra I. Stavrakis; Amanda H. Loftin; Stephen D. Zoller; Howard Y. Park; Sherif S R Richman; Daniel Johansen; Yan Hu; Julie A. Taylor; Christopher D. Hamad; Rene F. Chun; Weixian Xi; John S. Adams; Nicholas M. Bernthal

Periprosthetic joint infections are devastating complications for patients and for our health system. With growing demand for arthroplasty, the incidence of these infections is projected to increase exponentially. This paper is a review of existing animal models to study periprosthetic infection aimed at providing scientists with a succinct presentation of strengths and weaknesses of available in vivo systems. These systems represent the tools available to investigate novel antimicrobial therapies and reduce the clinical and economic impact of implant infections.

Collaboration


Dive into the Amanda H. Loftin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vishal Hegde

University of California

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

Jared A. Niska

University of California

View shared research outputs
Top Co-Authors

Avatar

Lloyd S. Miller

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge