Network


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

Hotspot


Dive into the research topics where Anusuya Das is active.

Publication


Featured researches published by Anusuya Das.


World journal of orthopedics | 2012

Osteonecrosis of the femoral head: An update in year 2012

Anjan P. Kaushik; Anusuya Das; Quanjun Cui

Osteonecrosis is a phenomenon involving disruption to the vascular supply to the femoral head, resulting in articular surface collapse and eventual osteoarthritis. Although alcoholism, steroid use, and hip trauma remain the most common causes, several other etiologies for osteonecrosis have been identified. Basic science research utilizing animal models and stem cell applications continue to further elucidate the pathophysiology of osteonecrosis and promise novel treatment options in the future. Clinical studies evaluating modern joint-sparing procedures have demonstrated significant improvements in outcomes, but hip arthroplasty is still the most common procedure performed in these affected younger adults. Further advances in joint-preserving procedures are required and will be widely studied in the coming decade.


Biomaterials | 2013

The promotion of mandibular defect healing by the targeting of S1P receptors and the recruitment of alternatively activated macrophages.

Anusuya Das; Claire E. Segar; Brian B. Hughley; Daniel T. Bowers; Edward A. Botchwey

Endogenous signals originating at the site of injury are involved in the paracrine recruitment, proliferation, and differentiation of circulating progenitor and diverse inflammatory cell types. Here, we investigate a strategy to exploit endogenous cell recruitment mechanisms to regenerate injured bone by local targeting and activation of sphingosine-1-phosphate (S1P) receptors. A mandibular defect model was selected for evaluating regeneration of bone following trauma or congenital disease. The particular challenges of mandibular reconstruction are inherent in the complex anatomy and function of the bone given that the area is highly vascularized and in close proximity to muscle. Nanofibers composed of poly(DL-lactide-co-glycolide) (PLAGA) and polycaprolactone (PCL) were used to delivery FTY720, a targeted agonist of S1P receptors 1 and 3. In vitro culture of bone progenitor cells on drug-loaded constructs significantly enhanced SDF1α mediated chemotaxis of bone marrow mononuclear cells. In vivo results show that local delivery of FTY720 from composite nanofibers enhanced blood vessel ingrowth and increased recruitment of M2 alternatively activated macrophages, leading to significant osseous tissue ingrowth into critical sized defects after 12 weeks of treatment. These results demonstrate that local activation of S1P receptors is a regenerative cue resulting in recruitment of wound healing or anti-inflammatory macrophages and bone healing. Use of such small molecule therapy can provide an alternative to biological factors for the clinical treatment of critical size craniofacial defects.


Journal of Biomedical Materials Research Part A | 2014

Delivery of S1P receptor-targeted drugs via biodegradable polymer scaffolds enhances bone regeneration in a critical size cranial defect

Anusuya Das; Shaun Tanner; Daniel A. Barker; David Green; Edward A. Botchwey

Biodegradable polymer scaffolds can be used to deliver soluble factors to enhance osseous remodeling in bone defects. To this end, we designed a poly(lactic-co-glycolic acid) (PLAGA) microsphere scaffold to sustain the release of FTY720, a selective agonist for sphingosine 1-phosphate (S1P) receptors. The microsphere scaffolds were created from fast degrading 50:50 PLAGA and/or from slow-degrading 85:15 PLAGA. Temporal and spatial regulation of bone remodeling depended on the use of appropriate scaffolds for drug delivery. The release profiles from the scaffolds were used to design an optimal delivery system to treat critical size cranial defects in a rodent model. The ability of local FTY720 delivery to maximize bone regeneration was evaluated with micro-computed tomography (microCT) and histology. Following 4 weeks of defect healing, FTY720 delivery from 85:15 PLAGA scaffolds resulted in a significant increase in bone volumes in the defect region compared to the controls. A 85:15 microsphere scaffolds maintain their structural integrity over a longer period of time, and cause an initial burst release of FTY720 due to surface localization of the drug. This encourages cellular in-growth and an increase in new bone formation.


Craniomaxillofacial Trauma and Reconstruction | 2014

Current Concepts of Bone Tissue Engineering for Craniofacial Bone Defect Repair

Brian A. Fishero; Nikita Kohli; Anusuya Das; J. Jared Christophel; Quanjun Cui

Craniofacial fractures and bony defects are common causes of morbidity and contribute to increasing health care costs. Successful regeneration of bone requires the concomitant processes of osteogenesis and neovascularization. Current methods of repair and reconstruction include rigid fixation, grafting, and free tissue transfer. However, these methods carry innate complications, including plate extrusion, nonunion, graft/flap failure, and donor site morbidity. Recent research efforts have focused on using stem cells and synthetic scaffolds to heal critical-sized bone defects similar to those sustained from traumatic injury or ablative oncologic surgery. Growth factors can be used to augment both osteogenesis and neovascularization across these defects. Many different growth factor delivery techniques and scaffold compositions have been explored yet none have emerged as the universally accepted standard. In this review, we will discuss the recent literature regarding the use of stem cells, growth factors, and synthetic scaffolds as alternative methods of craniofacial fracture repair.


PLOS ONE | 2014

Delivery of Bioactive Lipids from Composite Microgel-Microsphere Injectable Scaffolds Enhances Stem Cell Recruitment and Skeletal Repair

Anusuya Das; Daniel A. Barker; Tiffany Wang; Cheryl M. Lau; Yong Lin; Edward A. Botchwey

In this study, a microgel composed of chitosan and inorganic phosphates was used to deliver poly(lactic-co-glycolic acid) (PLAGA) microspheres loaded with sphingolipid growth factor FTY720 to critical size cranial defects in Sprague Dawley rats. We show that sustained release of FTY720 from injected microspheres used alone or in combination with recombinant human bone morphogenic protein-2 (rhBMP2) improves defect vascularization and bone formation in the presence and absence of rhBMP2 as evaluated by quantitative microCT and histological measurements. Moreover, sustained delivery of FTY720 from PLAGA and local targeting of sphingosine 1-phosphate (S1P) receptors reduces CD45+ inflammatory cell infiltration, promotes endogenous recruitment of CD29+CD90+ bone progenitor cells and enhances the efficacy of rhBMP2 from chitosan microgels. Companion in vitro studies suggest that selective activation of sphingosine receptor subtype-3 (S1P3) via FTY720 treatment induces smad-1 phosphorylation in bone-marrow stromal cells. Additionally, FTY720 enhances stromal cell-derived factor-1 (SDF-1) mediated chemotaxis of CD90+CD11B-CD45- bone progenitor cells in vitro after stimulation with rhBMP2. We believe that use of such small molecule delivery formulations to recruit endogenous bone progenitors may be an attractive alternative to exogenous cell-based therapy.


ACS Nano | 2014

Spatiotemporal Oxygen Sensing Using Dual Emissive Boron Dye–Polylactide Nanofibers

Daniel T. Bowers; Michael L. Tanes; Anusuya Das; Yong Lin; Nicole A. Keane; Rebekah A. Neal; Molly E. Ogle; Kenneth L. Brayman; Cassandra L. Fraser; Edward A. Botchwey

Oxygenation in tissue scaffolds continues to be a limiting factor in regenerative medicine despite efforts to induce neovascularization or to use oxygen-generating materials. Unfortunately, many established methods to measure oxygen concentration, such as using electrodes, require mechanical disturbance of the tissue structure. To address the need for scaffold-based oxygen concentration monitoring, a single-component, self-referenced oxygen sensor was made into nanofibers. Electrospinning process parameters were tuned to produce a biomaterial scaffold with specific morphological features. The ratio of an oxygen sensitive phosphorescence signal to an oxygen insensitive fluorescence signal was calculated at each image pixel to determine an oxygenation value. A single component boron dye–polymer conjugate was chosen for additional investigation due to improved resistance to degradation in aqueous media compared to a boron dye polymer blend. Standardization curves show that in fully supplemented media, the fibers are responsive to dissolved oxygen concentrations less than 15 ppm. Spatial (millimeters) and temporal (minutes) ratiometric gradients were observed in vitro radiating outward from the center of a dense adherent cell grouping on scaffolds. Sensor activation in ischemia and cell transplant models in vivo show oxygenation decreases on the scale of minutes. The nanofiber construct offers a robust approach to biomaterial scaffold oxygen sensing.


Stem Cells | 2017

Sphingosine‐1‐phosphate Receptor‐3 Supports Hematopoietic Stem and Progenitor Cell Residence within the Bone Marrow Niche

Molly E. Ogle; Claire E. Olingy; Anthony O. Awojoodu; Anusuya Das; Rafael A. Ortiz; Hoi Yin Cheung; Edward A. Botchwey

Hematopoietic stem and progenitor cells (HSPCs) egress from bone marrow (BM) during homeostasis and at increased rates during stress; however, the mechanisms regulating their trafficking remain incompletely understood. Here we describe a novel role for lipid receptor, sphingosine‐1‐phosphate receptor 3 (S1PR3), in HSPC residence within the BM niche. HSPCs expressed increased levels of S1PR3 compared to differentiated BM cells. Pharmacological antagonism or knockout (KO) of S1PR3 mobilized HSPCs into blood circulation, suggesting that S1PR3 influences niche localization. S1PR3 antagonism suppressed BM and plasma SDF‐1, enabling HSPCs to migrate toward S1P‐rich plasma. Mobilization synergized with AMD3100‐mediated antagonism of CXCR4, which tethers HSPCs in the niche, and recovered homing deficits of AMD3100‐treated grafts. S1PR3 antagonism combined with AMD3100 improved re‐engraftment and survival in lethally irradiated recipients. Our studies indicate that S1PR3 and CXCR4 signaling cooperate to maintain HSPCs within the niche under homeostasis. These results highlight an important role for S1PR3 in HSPC niche occupancy and trafficking that can be harnessed for both rapid clinical stem cell mobilization and re‐engraftment strategies, as well as the opportunity to design novel therapeutics for control of recruitment, homing, and localization through bioactive lipid signaling. Stem Cells 2017;35:1040–1052


Biomaterials | 2015

Bioactive lipid coating of bone allografts directs engraftment and fate determination of bone marrow-derived cells in rat GFP chimeras

Anusuya Das; Claire E. Segar; Yihsuan Chu; Tiffany Wang; Yong Lin; Chunxi Yang; Xeujun Du; Roy C. Ogle; Quanjun Cui; Edward A. Botchwey

Bone grafting procedures are performed to treat wounds incurred during wartime trauma, accidents, and tumor resections. Endogenous mechanisms of repair are often insufficient to ensure integration between host and donor bone and subsequent restoration of function. We investigated the role that bone marrow-derived cells play in bone regeneration and sought to increase their contributions by functionalizing bone allografts with bioactive lipid coatings. Polymer-coated allografts were used to locally deliver the immunomodulatory small molecule FTY720 in tibial defects created in rat bone marrow chimeras containing genetically-labeled bone marrow for monitoring cell origin and fate. Donor bone marrow contributed significantly to both myeloid and osteogenic cells in remodeling tissue surrounding allografts. FTY720 coatings altered the phenotype of immune cells two weeks post-injury, which was associated with increased vascularization and bone formation surrounding allografts. Consequently, degradable polymer coating strategies that deliver small molecule growth factors such as FTY720 represent a novel therapeutic strategy for harnessing endogenous bone marrow-derived progenitors and enhancing healing in load-bearing bone defects.


Drug Delivery and Translational Research | 2016

Erratum to: Enhanced osseous integration of human trabecular allografts following surface modification with bioactive lipids

Tiffany Wang; Jack R. Krieger; Cynthia Huang; Anusuya Das; Molly Dickinson; Michael P. Francis; Roy C. Ogle; Edward A. Botchwey

In this study, we used extracellular matrix (ECM) gels and human bone allograft as matrix vehicles to deliver the sphingolipid growth factor FTY720 to rodent models of tibial fracture and a critical-sized cranial defect. We show that FTY720 released from injectable ECM gels may accelerate callous formation and resolution and bone volume in a mouse tibial fracture model. We then show that FTY720 binds directly to human trabecular allograft bone and releases over 1 week in vitro. Rat critical-sized cranial defects treated with FTY720-coated grafts show increases in vascularization and bone deposition, with histological and micro-computed topography (microCT) evidence of enhanced bone formation within the graft and defect void. Immunohistochemical analysis suggests that osteogenesis within FTY720-coated grafts is associated with reduced CD68(+) macrophage infiltration and recruitment of CD29(+) bone progenitor cells. Matrix binding of FTY720 thus represents a promising and robust bone regeneration strategy with potential clinical translatability.


Plastic and Reconstructive Surgery | 2015

Deferoxamine mitigates radiation-induced tissue injury in a rat irradiated TRAM flap model.

Alexander F. Mericli; Anusuya Das; Ryan Best; Pamela Rodeheaver; George T. Rodeheaver; Kant Y. Lin

Background: Radiation therapy results in permanent damage to the microvasculature, leading to dermal damage and inelasticity in normal tissues. Deferoxamine is a U.S. Food and Drug Administration–approved iron-chelating medication that has also been shown to increase angiogenesis. The authors hypothesize that the application of deferoxamine will result in increased vascularity and improved tissue elasticity in a rat irradiated transverse rectus abdominis musculocutaneous flap model. Methods: Fifteen rats underwent a transverse rectus abdominis myocutaneous flap and were randomized to three groups: control, radiation therapy, and radiation therapy plus deferoxamine. The flaps in the radiation therapy and radiation therapy plus deferoxamine groups were irradiated with 35 Gy in a single dose. Four weeks after irradiation, rats in the radiation therapy plus deferoxamine group were treated with deferoxamine. Flaps were imaged with micro–computed tomographic angiography. Flap creep and stress relaxation were assessed using a tensiometer. Hematoxylin and eosin, picrosirius red, and Verhoeff-van Gieson staining was performed. Results: Irradiated flaps demonstrated gross stigmata of cutaneous radiation injury within 4 weeks. Histologically, the epidermis in the radiation therapy flaps was found to be thicker than in the radiation therapy plus deferoxamine and control flaps (p < 0.001). Micro–computed tomographic angiography demonstrated a statistically significant (p < 0.05) increase in vascularity in the radiation therapy plus deferoxamine flaps compared with radiation therapy alone. The creep curve was indicative of increased elasticity in the radiation therapy plus deferoxamine flaps compared with radiation therapy flaps. Conclusion: Deferoxamine appears to mitigate radiation-induced hypovascularity and improve tissue elasticity in a rat model of soft-tissue reconstruction.

Collaboration


Dive into the Anusuya Das's collaboration.

Top Co-Authors

Avatar

Edward A. Botchwey

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Quanjun Cui

University of Virginia

View shared research outputs
Top Co-Authors

Avatar

Tiffany Wang

Georgia Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kant Y. Lin

University of Virginia Health System

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yong Lin

University of Virginia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge