Network


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

Hotspot


Dive into the research topics where Benjamin T. Corona is active.

Publication


Featured researches published by Benjamin T. Corona.


Biomaterials | 2013

The promotion of a functional fibrosis in skeletal muscle with volumetric muscle loss injury following the transplantation of muscle-ECM

Benjamin T. Corona; Xiaowu Wu; Catherine L. Ward; Jennifer S. McDaniel; Christopher R. Rathbone; Thomas J. Walters

Tissue engineering strategies that primarily use biological extracellular matrices (ECMs) with or without the inclusion of a stem or progenitor cell source are under development for the treatment of trauma resulting in the loss of a large volume of skeletal muscle (i.e., volumetric muscle loss; VML). The explicit goal is to restore functional capacity to the injured tissue by promoting generation of muscle fibers. In the current study, a syngeneic muscle-derived ECM (mECM) was transplanted in a rat tibialis anterior (TA) muscle VML model. Instead of muscle fiber generation a large fibrotic mass was produced by mECM transplantation out to six months post-injury. Surprisingly, recovery of one-third of the original functional deficit was still achieved by two months post-injury following mECM transplantation. These counterintuitive findings may be due, at least in part, to the ability of mECM to attenuate muscle damage in the remaining muscle as compared to non-repaired muscle. These findings point to a novel role of biological ECMs for the treatment of VML, wherein the remaining muscle mass is protected from prolonged overload injury.


American Journal of Physiology-cell Physiology | 2013

Autologous minced muscle grafts: a tissue engineering therapy for the volumetric loss of skeletal muscle

Benjamin T. Corona; Koyal Garg; Catherine L. Ward; Jennifer S. McDaniel; Thomas J. Walters; Christopher R. Rathbone

Volumetric muscle loss (VML) results in a large void deficient in the requisite materials for regeneration for which there is no definitive clinical standard of care. Autologous minced muscle grafts (MG), which contain the essential components for muscle regeneration, may embody an ideal tissue engineering therapy for VML. The purpose of this study was to determine if orthotopic transplantation of MG acutely after VML in the tibialis anterior muscle of male Lewis rats promotes functional tissue regeneration. Herein we report that over the first 16 wk postinjury, MG transplantation 1) promotes remarkable regeneration of innervated muscle fibers within the defect area (i.e., de novo muscle fiber regeneration); 2) reduced evidence of chronic injury in the remaining muscle mass compared with nonrepaired muscles following VML (i.e., transplantation attenuated chronically upregulated transforming growth factor-β1 gene expression and the presence of centrally located nuclei in 30% of fibers observed in nonrepaired muscles); and 3) significantly improves net torque production (i.e., ∼55% of the functional deficit in nonrepaired muscles was restored). Additionally, voluntary wheel running was shown to reduce the heightened accumulation of extracellular matrix deposition observed within the regenerated tissue of MG-repaired sedentary rats 8 wk postinjury (collagen 1% area: sedentary vs. runner, ∼41 vs. 30%), which may have been the result of an augmented inflammatory response [i.e., M1 (CCR7) and M2 (CD163) macrophage expression was significantly greater in runner than sedentary MG-repaired muscles 2 wk postinjury]. These findings support further exploration of autologous minced MGs for the treatment of VML.


Biomaterials | 2015

An acellular biologic scaffold does not regenerate appreciable de novo muscle tissue in rat models of volumetric muscle loss injury.

Amit Aurora; Janet L. Roe; Benjamin T. Corona; Thomas J. Walters

Extracellular matrix (ECM) derived scaffolds continue to be investigated for the treatment of volumetric muscle loss (VML) injuries. Clinically, ECM scaffolds have been used for lower extremity VML repair; in particular, MatriStem™, a porcine urinary bladder matrix (UBM), has shown improved functional outcomes and vascularization, but limited myogenesis. However, efficacy of the scaffold for the repair of traumatic muscle injuries has not been examined systematically. In this study, we demonstrate that the porcine UBM scaffold when used to repair a rodent gastrocnemius musculotendinous junction (MTJ) and tibialis anterior (TA) VML injury does not support muscle tissue regeneration. In the MTJ model, the scaffold was completely resorbed without tissue remodeling, suggesting that the scaffold may not be suitable for the clinical repair of muscle-tendon injuries. In the TA VML injury, the scaffold remodeled into a fibrotic tissue and showed functional improvement, but not due to muscle fiber regeneration. The inclusion of physical rehabilitation also did not improve functional response or tissue remodeling. We conclude that the porcine UBM scaffold when used to treat VML injuries may hasten the functional recovery through the mechanism of scaffold mediated functional fibrosis. Thus for appreciable muscle regeneration, repair strategies that incorporate myogenic cells, vasculogenic accelerant and a myoconductive scaffold need to be developed.


BioResearch Open Access | 2012

A Standardized Rat Model of Volumetric Muscle Loss Injury for the Development of Tissue Engineering Therapies

Xiaowu Wu; Benjamin T. Corona; Xiaoyu Chen; Thomas J. Walters

Abstract Soft tissue injuries involving volumetric muscle loss (VML) are defined as the traumatic or surgical loss of skeletal muscle with resultant functional impairment and represent a challenging clinical problem for both military and civilian medicine. In response, a variety of tissue engineering and regenerative medicine treatments are under preclinical development. A wide variety of animal models are being used, all with critical limitations. The objective of this study was to develop a model of VML that was reproducible and technically uncomplicated to provide a standardized platform for the development of tissue engineering and regenerative medicine solutions to VML repair. A rat model of VML involving excision of ∼20% of the muscles mass from the superficial portion of the middle third of the tibialis anterior (TA) muscle was developed and was functionally characterized. The contralateral TA muscle served as the uninjured control. Additionally, uninjured age-matched control rats were also tested to determine the effect of VML on the contralateral limb. TA muscles were assessed at 2 and 4 months postinjury. VML muscles weighed 22.7% and 19.5% less than contralateral muscles at 2 and 4 months postinjury, respectively. These differences were accompanied by a reduction in peak isometric tetanic force (Po) of 28.4% and 32.5% at 2 and 4 months. Importantly, Po corrected for differences in body weight and muscle wet weights were similar between contralateral and age-matched control muscles, indicating that VML did not have a significant impact on the contralateral limb. Lastly, repair of the injury with a biological scaffold resulted in rapid vascularization and integration with the wound. The technical simplicity, reliability, and clinical relevance of the VML model developed in this study make it ideal as a standard model for the development of tissue engineering solutions for VML.


Journal of Orthopaedic Research | 2015

Volumetric muscle loss: persistent functional deficits beyond frank loss of tissue.

Koyal Garg; Catherine L. Ward; Brady J. Hurtgen; Jason M. Wilken; Daniel J. Stinner; Joseph C. Wenke; Johnny G. Owens; Benjamin T. Corona

Open fracture is a common occurrence in civilian and military populations. Though great strides have been made in limb salvage efforts, persistent muscle strength deficits can contribute to a diminished limb function after the bone has healed. Over the past decade, a growing effort to establish therapies directed at de novo muscle regeneration has produced several therapeutic approaches. As this effort progresses and as therapies reach clinical testing, many questions remain regarding the pathophysiology of the volumetric loss of skeletal muscle. The current study demonstrates, in a rat “open fracture” model, that the volumetric loss of skeletal muscle results in persistent functional deficits that are dependent on muscle length and joint angle. Moreover, the injured muscle has an increased stiffness during passive stretch and a reduced functional excursion. A case study of a patient with an open type III tibia fracture resulting in volumetric muscle loss in the anterior and posterior compartment is also presented. Eighteen months after injury and tibia healing, persistent functional deficits are apparent with many of the same qualities demonstrated in the animal model. Muscle architectural adaptations likely underlie the altered intrinsic functional characteristics of the remaining musculature. Published 2014. This article is a U.S. Government work and is in the public domain in the USA. J Orthop Res 33:40–46, 2015.


Frontiers in Pharmacology | 2015

Therapeutic strategies for preventing skeletal muscle fibrosis after injury

Koyal Garg; Benjamin T. Corona; Thomas J. Walters

Skeletal muscle repair after injury includes a complex and well-coordinated regenerative response. However, fibrosis often manifests, leading to aberrant regeneration and incomplete functional recovery. Research efforts have focused on the use of anti-fibrotic agents aimed at reducing the fibrotic response and improving functional recovery. While there are a number of mediators involved in the development of post-injury fibrosis, TGF-β1 is the primary pro-fibrogenic growth factor and several agents that inactivate TGF-β1 signaling cascade have emerged as promising anti-fibrotic therapies. A number of these agents are FDA approved for other conditions, clearing the way for rapid translation into clinical treatment. In this article, we provide an overview of muscles host response to injury with special emphasis on the cellular and non-cellular mediators involved in the development of fibrosis. This article also reviews the findings of several pre-clinical studies that have utilized anti-fibrotic agents to improve muscle healing following most common forms of muscle injuries. Although some studies have shown positive results with anti-fibrotic treatment, others have indicated adverse outcomes. Some concerns and questions regarding the clinical potential of these anti-fibrotic agents have also been presented.


Journal of Rehabilitation Research and Development | 2015

Volumetric muscle loss leads to permanent disability following extremity trauma.

Benjamin T. Corona; Jessica C. Rivera; Johnny G. Owens; Joseph C. Wenke; Christopher R. Rathbone

Extremity injuries comprise the majority of battlefield injuries and contribute the most to long-term disability of servicemembers. The purpose of this study was to better define the contribution of muscle deficits and volumetric muscle loss (VML) to the designation of long-term disability in order to better understand their effect on outcomes for limb-salvage patients. Medically retired servicemembers who sustained a combat-related type III open tibia fracture (Orthopedic cohort) were reviewed for results of their medical evaluation leading to discharge from military service. A cohort of battlefield-injured servicemembers (including those with nonorthopedic injuries) who were medically retired because of various injuries (General cohort) was also examined. Muscle conditions accounted for 65% of the disability of patients in the Orthopedic cohort. Among the General cohort, 92% of the muscle conditions were identified as VML. VML is a condition that contributes significantly to long-term disability, and the development of therapies addressing VML has the potential to fill a significant void in orthopedic care.


Biomaterials | 2016

Challenges to acellular biological scaffold mediated skeletal muscle tissue regeneration.

Benjamin T. Corona; Sarah M. Greising

Volumetric muscle loss (VML) injuries present a complex and heterogeneous clinical problem that results in a chronic loss of muscle tissue and strength. The primary limitation to muscle tissue regeneration after VML injury is the frank loss of all native muscle constituents in the defect, especially satellite cells and the basal lamina. Recent advancements in regenerative medicine have set forth encouraging and emerging translational and therapeutic options for these devastating injuries including the surgical implantation of acellular biological scaffolds. While these biomaterials can modulate the wound environment, the existing data do not support their capacity to promote appreciable muscle fiber regeneration that can contribute to skeletal muscle tissue functional improvements. An apparent restriction of endogenous satellite cell (i.e., pax7(+)) migration to acellular biological scaffolds likely underlies this deficiency. This work critically evaluates the role of an acellular biological scaffold in orchestrating skeletal muscle tissue regeneration, specifically when used as a regenerative medicine approach for VML injury.


Biomaterials | 2013

The Role of Endothelial Cells in Myofiber Differentiation and the Vascularization and Innervation of Bioengineered Muscle Tissue in vivo

Tracy Criswell; Benjamin T. Corona; Zhan Wang; Yu Zhou; Yong Xu; George J. Christ; Shay Soker

Musculoskeletal disorders are a major cause of disability and effective treatments are currently lacking. Tissue engineering affords the possibility of new therapies utilizing cells and biomaterials for the recovery of muscle volume and function. A major consideration in skeletal muscle engineering is the integration of a functional vasculature within the regenerating tissue. In this study we employed fluorescent cell labels to track the location and differentiation of co-cultured cells in vivo and in vitro. We first utilized a co-culture of fluorescently labeled endothelial cells (ECs) and muscle progenitor cells (MPCs) to investigate the ability of ECs to enhance muscle tissue formation and vascularization in an in vivo model of bioengineered muscle. Scaffolds that had been seeded with both MPCs and ECs showed significantly greater vascularization, tissue formation and enhanced innervation as compared to scaffolds seeded with MPCs alone. Subsequently, we performed in vitro experiments using a 3-cell type system (ECs, MPCs, and pericytes (PCs)) to demonstrate the utility of fluorescent cell labeling for monitoring cell growth and differentiation. The growth and differentiation of individual cell types was determined using live cell fluorescent microscopy demonstrating the utility of fluorescent labels to monitor tissue organization in real time.


BioResearch Open Access | 2015

An Autologous Muscle Tissue Expansion Approach for the Treatment of Volumetric Muscle Loss

Catherine L. Ward; Lisa L. Ji; Benjamin T. Corona

Abstract Volumetric muscle loss (VML) is a hallmark of orthopedic trauma with no current standard of care. As a potential therapy for some VML indications, autologous minced muscle grafts (1 mm3 pieces of muscle) are effective in promoting remarkable de novo fiber regeneration. But they require ample donor muscle tissue and therefore may be limited in their application for large clinical VML. Here, we tested the hypothesis that autologous minced grafts may be volume expanded in a collagen hydrogel, allowing for the use of lesser autologous muscle while maintaining regenerative and functional efficacy. The results of the study indicate that 50% (but not 75%) less minced graft tissue suspended in a collagen hydrogel promoted a functional improvement similar to that of a 100% minced graft repair. However, approximately half of the number of fibers regenerated de novo with 50% graft repair. Moreover, the fibers that regenerated had a smaller cross-sectional area. These findings support the concept of using autologous minced grafts for the regeneration of muscle tissue after VML, but indicate the need to identify optimal carrier materials for expansion.

Collaboration


Dive into the Benjamin T. Corona's collaboration.

Top Co-Authors

Avatar

Catherine L. Ward

Wake Forest Institute for Regenerative Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Koyal Garg

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

George J. Christ

Wake Forest Institute for Regenerative Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alain Watts

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Beth E. Pollot

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Brady J. Hurtgen

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Carlos A. Aguilar

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

James J. Yoo

Wake Forest Institute for Regenerative Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge