Network


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

Hotspot


Dive into the research topics where Jacqueline M. Bliley is active.

Publication


Featured researches published by Jacqueline M. Bliley.


Plastic and Reconstructive Surgery | 2013

Prevalence of endogenous CD34+ adipose stem cells predicts human fat graft retention in a xenograft model.

Brian J. Philips; Tara L. Grahovac; Jolene E. Valentin; Christopher W. Chung; Jacqueline M. Bliley; Melanie E. Pfeifer; Sohini B. Roy; Stephanie E. Dreifuss; Arta Kelmendi-Doko; Russell E. Kling; Sudheer Ravuri; Kacey G. Marra; Vera S. Donnenberg; Albert D. Donnenberg; J. Peter Rubin

Background: Fat grafting is a promising technique for soft-tissue augmentation, although graft retention is highly unpredictable and factors that affect graft survival have not been well defined. Because of their capacity for differentiation and growth factor release, adipose-derived stem cells may have a key role in graft healing. The authors’ objective was to determine whether biological properties of adipose-derived stem cells present within human fat would correlate with in vivo outcomes of graft volume retention. Methods: Lipoaspirate from eight human subjects was processed using a standardized centrifugation technique and then injected subcutaneously into the flanks of 6-week-old athymic nude mice. Graft masses and volumes were measured, and histologic evaluation, including CD31+ staining for vessels, was performed 8 weeks after transplantation. Stromal vascular fraction isolated at the time of harvest from each subject was analyzed for surface markers by multiparameter flow cytometry, and also assessed for proliferation, differentiation capacity, and normoxic/hypoxic vascular endothelial growth factor secretion. Results: Wide variation in percentage of CD34+ progenitors within the stromal vascular fraction was noted among subjects and averaged 21.3 ± 15 percent (mean ± SD). Proliferation rates and adipogenic potential among stromal vascular fraction cells demonstrated moderate interpatient variability. In mouse xenograft studies, retention volumes ranged from approximately 36 to 68 percent after 8 weeks, with an overall average of 52 ± 11 percent. A strong correlation (r = 0.78, slope = 0.76, p < 0.05) existed between stromal vascular fraction percentage of CD34+ progenitors and high graft retention. Conclusion: Inherent biological differences in adipose tissue exist between patients. In particular, concentration of CD34+ progenitor cells within the stromal vascular fraction may be one of the factors used to predict human fat graft retention.


Journal of Tissue Engineering and Regenerative Medicine | 2017

Delivery of chondroitinase ABC and glial cell line-derived neurotrophic factor from silk fibroin conduits enhances peripheral nerve regeneration.

Wesley N. Sivak; James D. White; Jacqueline M. Bliley; Lee W. Tien; Han Tsung Liao; David L. Kaplan; Kacey G. Marra

Nerve conduits are a proven strategy for guiding axon regrowth following injury. This study compares degradable silk–trehalose films containing chondroitinase ABC (ChABC) and/or glial cell line‐derived neurotrophic factor (GDNF) loaded within a silk fibroin‐based nerve conduit in a rat sciatic nerve defect model. Four groups of silk conduits were prepared, with the following silk–trehalose films inserted into the conduit: (a) empty; (b) 1 µg GDNF; (3) 2 U ChABC; and (4) 1 µg GDNF/2 U ChABC. Drug release studies demonstrated 20% recovery of GDNF and ChABC at 6 weeks and 24 h, respectively. Six conduits of each type were implanted into 15 mm sciatic nerve defects in Lewis rats; conduits were explanted for histological analysis at 6 weeks. Tissues stained with Schwann cell S‐100 antibody demonstrated an increased density of cells in both GDNF‐ and ChABC‐treated groups compared to empty control conduits (p < 0.05). Conduits loaded with GDNF and ChABC also demonstrated higher levels of neuron‐specific PGP 9.5 protein when compared to controls (p < 0.05). In this study we demonstrated a method to enhance Schwann cell migration and proliferation and also foster axonal regeneration when repairing peripheral nerve gap defects. Silk fibroin‐based nerve conduits possess favourable mechanical and degradative properties and are further enhanced when loaded with ChABC and GDNF. Copyright


Plastic and Reconstructive Surgery | 2017

Single Implantable FK506 Disk Prevents Rejection in Vascularized Composite Allotransplantation

Jignesh V. Unadkat; Jonas T. Schnider; Firuz Feturi; Wakako Tsuji; Jacqueline M. Bliley; Raman Venkataramanan; Mario G. Solari; Kacey G. Marra; Vijay S. Gorantla; Alexander M. Spiess

Background: In vascularized composite allotransplantation, medication nonadherence leads to increased acute rejections. Improving medication adherence would improve overall allograft survival. Regionally delivered immunosuppression, targeted to sites of allorecognition, may reduce or eliminate the need for daily systemic immunosuppression. Methods: The authors developed biodegradable FK disks containing FK506-loaded double-walled microspheres and tested their efficacy at preventing rejection in a Brown-Norway–to-Lewis rat hindlimb transplantation model. In some experimental group animals, one FK disk was implanted subcutaneously either in native nontransplanted leg or in a transplanted allograft. Regular blood FK506 levels were measured. The endpoint was 180-day allograft survival or grade 3 rejection. At the endpoint, tissue FK506 levels were measured and mixed lymphocytic reaction was performed. Results: A single FK disk maintained systemic blood FK506 levels between 5 and 15 ng/ml for 146 ± 11.1 days. After that, the levels declined to less than 5 ng/ml through the endpoint. There was significantly increased FK506 concentration in groin lymph nodes draining the implanted FK disk. Compared with other groups, animals with an FK disk in the transplanted allograft had 100 percent allograft survival to more than 180 days despite subtherapeutic levels below 5 ng/ml. In these animals, significant T-cell hyporesponsiveness was seen in groin lymph nodes draining the FK disk compared with robust splenic T-cell proliferation. Conclusions: Sustained regional immunosuppression (with a single FK506 disk) maintained the allograft by means of a high regional concentration of FK506. Notably, this was achieved at subtherapeutic blood concentrations of FK506, without any further systemic FK506 administration.


Burns | 2016

Administration of adipose-derived stem cells enhances vascularity, induces collagen deposition, and dermal adipogenesis in burn wounds

Jacqueline M. Bliley; Anne E. Argenta; Latha Satish; Meghan M. McLaughlin; Aaron Dees; Casey Tompkins-Rhoades; Kacey G. Marra; J. Peter Rubin

Current treatment options for severe burn wounds are often insufficient in reconstructing skin and soft tissue defects. Adipose-derived stem cells (ASCs), a readily available source of multipotent stem cells, represent a promising therapy for the treatment of full-thickness burn wounds. Full-thickness burn wounds were created on the paraspinal region of athymic mice. A one-time, sub-eschar injection of 6.8×10(6) ASCs in PBS or PBS alone was administered at 24-h postoperatively. Time to healing was quantified using Image J analysis. At days 4, 7, 14, and 21, mice were sacrificed and tissues were excised for molecular and histological analysis. ASCs were able to survive in burn wounds as determined by the presence of PKH labeling and human PPARγ expression within the wounds. CD-31 staining demonstrated increased vascularity in ASC-treated wounds at POD 4 (p<0.05). Molecular studies showed enhanced adipogenesis, as well as type III and type I collagen deposition in the ASC treated group (p<0.05). An increase in the mRNA expression ratio of type III to type I collagen was also observed following ASC treatment (p<0.05). By enhancing vascularity, collagen deposition, and adipogenesis, ASCs show promise as an adjunctive therapy for the current treatment of full thickness burn wounds.


Plastic and Reconstructive Surgery | 2015

Imaging the Stromal Vascular Fraction during Soft-Tissue Reconstruction.

Jacqueline M. Bliley; Satish L; McLaughlin Mm; Russell E. Kling; Day; Grahovac Tl; Kokai Le; Zhang W; Kacey G. Marra; Rubin Jp

Background: Although fat grafting is an increasingly popular practice, suboptimal volume retention remains an obstacle. Graft enrichment with the stromal vascular fraction has gained attention as a method of increasing graft retention. However, few studies have assessed the fate and impact of transplanted stromal vascular fraction on fat grafts. In vivo imaging techniques can be used to help determine the influence stromal vascular fraction has on transplanted fat. Methods: Stromal vascular fraction was labeled with 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindotricarbocyanine iodide (DiR), a near-infrared dye, and tracked in vivo. Proliferation and differentiation of labeled cells were assessed to confirm that labeling did not adversely affect cellular function. Different doses of labeled stromal vascular fraction were tracked within fat grafts over time using the in vivo imaging system. Results: No significant differences in differentiation and proliferation were observed in labeled versus unlabeled cells (p > 0.05). A pilot study confirmed that stromal vascular fraction fluorescence was localized to fat grafts and different cell doses could be distinguished. A larger-scale in vivo study revealed that stromal vascular fraction fluorescence was statistically significant (p < 0.05) between different cell dose groups and this significance was maintained in higher doses (3 × 106 and 2 × 106 cells/ml of fat graft) for up to 41 days in vivo. Conclusions: DiR labeling allowed the authors to differentiate between cell doses and confirm localization. This article supports the use of DiR labeling in conjunction with in vivo imaging as a tool for imaging stromal vascular fraction within fat grafts. CLINICAL QUESTION/LEVEL OF EVIDENCE: Therapeutic, V.


Methods of Molecular Biology | 2014

Polymeric biomaterials for nerve regeneration: fabrication and implantation of a biodegradable nerve guide.

Wesley N. Sivak; Jacqueline M. Bliley; Kacey G. Marra

Optimizing the quantity, quality, and speed of axon regeneration is important in maximizing functional outcomes following peripheral nerve injury. When severed, injured nerves must be able to regenerate and reconnect to the structures they previously controlled within 12-18 months before sensation and motion are permanently lost. Nerve sprouts from the proximal stump will spontaneously migrate toward the distal stump in the event of a nerve transection. However, surgical intervention remains necessary to repair transection injuries. Regeneration becomes particularly troublesome with large gaps, where autologous nerve grafts or nerve guides are used to repair transected nerves. Nerve conduits function as therapeutic adjuncts, guiding axonal regeneration across gap defects. Despite the availability of several FDA-approved nerve conduits, functional outcomes following their use remain less than optimal. Much work has been focused on developing nerve conduits to improve peripheral nerve repair outcomes. This chapter describes fabrication of a poly(caprolactone) nerve guide and demonstrates its use in a rat sciatic nerve model.


Plastic and reconstructive surgery. Global open | 2015

Long-term Patency of Primary Arterial Repair and the Modified Cold Intolerance Symptom Severity Questionnaire.

Bernd Lannau; Jacqueline M. Bliley; Isaac B. James; Sheri Wang; Wesley N. Sivak; Kang Kim; John Fowler; Alexander M. Spiess

Background: The goal of this study was to assess the long-term arterial patency of repaired arteries in the upper extremity and any morbidity resulting from the subsequent occlusion of these vessels. Concurrently, a new questionnaire, the modified Cold Intolerance Symptom Severity (mod CISS) questionnaire, was developed to allow for better assessment of cold intolerance. Methods: Thirteen patients who had undergone repair of the radial (4 patients), ulnar (6 patients), brachial (1 patient), digital (1), and an undefined lower arm artery (1) were examined using questionnaires, physical examination, and high-resolution ultrasound. Results: Outcome measures that were statistically significantly worse in the group of patients who presented with nerve injuries included cold intolerance symptoms, Disabilities of the Arm, Shoulder, and Hand score, Michigan Hand Questionnaire, and grip strength (middle setting on dynamometer). The results from the mod CISS correlated with high statistical significance with the results of the CISS score for the injured hand. Of note, wrist extension was significantly better with patent arteries. Conclusions: Sixty-seven percent of arterial repairs remained patent at 6 years (mean) follow-up. The presence of nerve injury has a higher impact on the outcome metrics assessed in this study than arterial patency. Our modification of the CISS score enhances its utility as a survey of cold intolerance.


Muscle & Nerve | 2018

Delivery of adipose-derived stem cells in poloxamer hydrogel improves peripheral nerve regeneration: Poloxamer, ASCs, Nerve Repair

Kassandra Allbright; Jacqueline M. Bliley; Emmanuelle Havis; Deokyeol Kim; Gabriella DiBernardo; Damian Grybowski; Matthias Waldner; Isaac B. James; Wesley N. Sivak; J. Peter Rubin; Kacey G. Marra

Introduction: Peripheral nerve damage is associated with high long‐term morbidity. Because of beneficial secretome, immunomodulatory effects, and ease of clinical translation, transplantation with adipose‐derived stem cells (ASC) represents a promising therapeutic modality. Methods: Effect of ASC delivery in poloxamer hydrogel was assessed in a rat sciatic nerve model of critical‐sized (1.5 cm) peripheral nerve injury. Nerve/muscle unit regeneration was assessed via immunostaining explanted nerve, quantitative polymerase chain reaction (qPCR), and histological analysis of reinnervating gastrocnemius muscle. Results: On the basis of viability data, 10% poloxamer hydrogel was selected for in vivo study. Six weeks after transection and repair, the group treated with poloxamer delivered ASCs demonstrated longest axonal regrowth. The qPCR results indicated that the inclusion of ASCs appeared to result in expression of factors that aid in reinnervating muscle tissue. Discussion: Delivery of ASCs in poloxamer addresses multiple facets of the complexity of nerve/muscle unit regeneration, representing a promising avenue for further study. Muscle Nerve 58: 251–260, 2018


Stem Cell Biology and Tissue Engineering in Dental Sciences | 2015

Polymeric Biomaterials as Tissue Scaffolds

Jacqueline M. Bliley; Kacey G. Marra

Optimizing the quantity, quality and speed of axon regeneration is important in maximizing functional outcomes following peripheral nerve injury. When severed, injured nerves must be able to regenerate and reconnect to the structures they previously controlled within 12-18 months before sensation and motion are permanently lost. Nerve sprouts from the proximal stump will spontaneously migrate toward the distal stump in the event of a nerve transection. However, surgical intervention remains necessary to repair transection injuries. Regeneration becomes particularly troublesome with large gaps, where autologous nerve grafts or nerve guides are used to repair transected nerves. Nerve conduits function as therapeutic adjuncts, guiding axonal regeneration across gap defects. Despite the availability of several FDA-approved nerve conduits, functional outcomes following their use remain less than optimal. Much work has been focused on developing nerve conduits to improve peripheral nerve repair outcomes. This chapter describes fabrication of a poly(caprolactone) nerve guide and demonstrates its use in a rat sciatic nerve model.


Frontiers in Immunology | 2018

Characteristics and Immunomodulating Functions of Adipose-Derived and Bone Marrow-Derived Mesenchymal Stem Cells Across Defined Human Leukocyte Antigen Barriers

Matthias Waldner; Wensheng Zhang; Isaac B. James; Kassandra Allbright; Emmanuelle Havis; Jacqueline M. Bliley; Aurora Almadori; Riccardo Schweizer; Jan A. Plock; Kia M. Washington; Vijay S. Gorantla; Mario G. Solari; Kacey G. Marra; J. Peter Rubin

Background Vascularized composite allotransplantation opens new possibilities in reconstructive transplantation such as hand or face transplants. Lifelong immunosuppression and its side-effects are the main drawbacks of this procedure. Mesenchymal stem cells (MSCs) have clinically useful immunomodulatory effects and may be able to reduce the burden of chronic immunosuppression. Herein, we assess and compare characteristics and immunomodulatory capacities of bone marrow- and adipose tissue-derived MSCs isolated from the same human individual across defined human leukocyte antigen (HLA) barriers. Materials and methods Samples of omental (o.) adipose tissue, subcutaneous (s.c.) adipose tissue, and bone marrow aspirate from 10 human organ donors were retrieved and MSCs isolated. Cells were characterized by flow cytometry and differentiated in three lineages: adipogenic, osteogenic, and chondrogenic. In mixed lymphocyte reactions, the ability of adipose-derived mesenchymal stem cells (ASCs) and bone marrow-derived mesenchymal stem cells (BMSCs) to suppress the immune response was assessed and compared within individual donors. HLA mismatched or mitogen stimulations were analyzed in co-culture with different MSC concentrations. Supernatants were analyzed for cytokine contents. Results All cell types, s.c.ASC, o.ASC, and BMSC demonstrated individual differentiation potential and cell surface markers. Immunomodulating effects were dependent on dose and cell passage. Proliferation of responder cells was most effectively suppressed by s.c.ASCs and combination with BMSC resulted in highly efficient immunomodulation. Immunomodulation was not cell contact-dependent and cells demonstrated a specific cytokine secretion. Conclusion When human ASCs and BMSCs are isolated from the same individual, both show effective immunomodulation across defined HLA barriers in vitro. We demonstrate a synergistic effect when cells from the same biologic system were combined. This cell contact-independent function underlines the potential of clinical systemic application of MSCs.

Collaboration


Dive into the Jacqueline M. Bliley's collaboration.

Top Co-Authors

Avatar

Kacey G. Marra

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

J. Peter Rubin

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Isaac B. James

University of Pittsburgh

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
Top Co-Authors

Avatar

Emmanuelle Havis

Centre national de la recherche scientifique

View shared research outputs
Researchain Logo
Decentralizing Knowledge