Network


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

Hotspot


Dive into the research topics where Leena Pradhan-Nabzdyk is active.

Publication


Featured researches published by Leena Pradhan-Nabzdyk.


American Journal of Pathology | 2015

Substance P Promotes Wound Healing in Diabetes by Modulating Inflammation and Macrophage Phenotype

Ermelindo C. Leal; Eugénia Carvalho; Ana Tellechea; Antonios Kafanas; Francesco Tecilazich; Cathal J. Kearney; Sarada Kuchibhotla; Michael E. Auster; Efi Kokkotou; David J. Mooney; Frank W. LoGerfo; Leena Pradhan-Nabzdyk; Aristidis Veves

Diabetic foot ulceration is a major complication of diabetes. Substance P (SP) is involved in wound healing, but its effect in diabetic skin wounds is unclear. We examined the effect of exogenous SP delivery on diabetic mouse and rabbit wounds. We also studied the impact of deficiency in SP or its receptor, neurokinin-1 receptor, on wound healing in mouse models. SP treatment improved wound healing in mice and rabbits, whereas the absence of SP or its receptor impaired wound progression in mice. Moreover, SP bioavailability in diabetic skin was reduced as SP gene expression was decreased, whereas the gene expression and protein levels of the enzyme that degrades SP, neutral endopeptidase, were increased. Diabetes and SP deficiency were associated with absence of an acute inflammatory response important for wound healing progression and instead revealed a persistent inflammation throughout the healing process. SP treatment induced an acute inflammatory response, which enabled the progression to the proliferative phase and modulated macrophage activation toward the M2 phenotype that promotes wound healing. In conclusion, SP treatment reverses the chronic proinflammatory state in diabetic skin and promotes healing of diabetic wounds.


PLOS ONE | 2013

Role of Endothelial Progenitor Cells and Inflammatory Cytokines in Healing of Diabetic Foot Ulcers

Francesco Tecilazich; Thanh Theresa Dinh; Leena Pradhan-Nabzdyk; Ermelindo C. Leal; Ana Tellechea; Antonios Kafanas; Charalambos Gnardellis; Mary L. Magargee; Andre Dejam; Vasilis Toxavidis; John Tigges; Eugénia Carvalho; Thomas E. Lyons; Aristidis Veves

Background To evaluate changes in endothelial progenitor cells (EPCs) and cytokines in patients with diabetic foot ulceration (DFU) in association with wound healing. Methods We studied healthy subjects, diabetic patients not at risk of DFU, at risk of DFU and with active DFU. We prospectively followed the DFU patients over a 12-week period. We also investigated similar changes in diabetic rabbit and mouse models of wound healing. Results All EPC phenotypes except the kinase insert domain receptor (KDR)+CD133+ were reduced in the at risk and the DFU groups compared to the controls. There were no major EPC differences between the control and not at risk group, and between the at risk and DFU groups. Serum stromal-cell derived factor-1 (SDF-1) and stem cell factor (SCF) were increased in DFU patients. DFU patients who healed their ulcers had lower CD34+KDR+ count at visits 3 and 4, serum c-reactive protein (CRP) and granulocyte-macrophage colony-stimulating factor (GM-CSF) at visit 1, interleukin-1 (IL-1) at visits 1 and 4. EPCs tended to be higher in both diabetic animal models when compared to their non-diabetic counterparts both before and ten days after wounding. Conclusions Uncomplicated diabetes does not affect EPCs. EPCs are reduced in patients at risk or with DFU while complete wound healing is associated with CD34+KDR+ reduction, suggesting possible increased homing. Low baseline CRP, IL-1α and GM-CSF serum levels were associated with complete wound healing and may potentially serve as prognostic markers of DFU healing. No animal model alone is representative of the human condition, indicating the need for multiple experimental models.


Biomaterials | 2014

Gene silencing in human aortic smooth muscle cells induced by PEI-siRNA complexes released from dip-coated electrospun poly(ethylene terephthalate) grafts.

Christoph S. Nabzdyk; Maggie Chun; Hunter S. Oliver-Allen; Saif G. Pathan; Matthew D. Phaneuf; Jin-Oh You; Leena Pradhan-Nabzdyk; Frank W. LoGerfo

An excessive tissue response to prosthetic arterial graft material leads to intimal hyperplasia (IH), the leading cause of late graft failure. Seroma and abnormal capsule formation may also occur after prosthetic material implantation. The matricellular protein Thrombospondin-2 (TSP-2) has shown to be upregulated in response to biomaterial implantation. This study evaluates the uptake and release of small interfering RNA (siRNA) from unmodified and surface functionalized electrospun PET graft materials. ePET graft materials were synthesized using electrospinning technology. Subsets of the ePET materials were then chemically modified to create surface functional groups. Unmodified and surface-modified ePET grafts were dip-coated in siRNAs alone or siRNAs complexed with transfection reagents polyethyleneimine (PEI) or Lipofectamine RNAiMax. Further, control and TSP-2 siRNA-PEI complex treated ePET samples were placed onto a confluent layer of human aortic smooth muscle cells (AoSMCs). Complexation of all siRNAs with PEI led to a significant increase in adsorption to unmodified ePET. TSP-2 siRNA-PEI released from unmodified-ePET silenced TSP-2 in AoSMC. Regardless of the siRNA-PEI complex evaluated, AoSMC migrated into the ePET. siRNA-PEI complexes delivered to AoSMC from dip-coated ePET can result in gene knockdown. This methodology for siRNA delivery may improve the tissue response to vascular and other prosthetics.


PLOS ONE | 2012

Temporal Network Based Analysis of Cell Specific Vein Graft Transcriptome Defines Key Pathways and Hub Genes in Implantation Injury

Manoj Bhasin; Zhen Huang; Leena Pradhan-Nabzdyk; Junaid Y. Malek; Philip J. LoGerfo; Mauricio A. Contreras; Patrick Guthrie; Eva Csizmadia; Nicholas D. Andersen; Olivier Kocher; Christiane Ferran; Frank W. LoGerfo

Vein graft failure occurs between 1 and 6 months after implantation due to obstructive intimal hyperplasia, related in part to implantation injury. The cell-specific and temporal response of the transcriptome to vein graft implantation injury was determined by transcriptional profiling of laser capture microdissected endothelial cells (EC) and medial smooth muscle cells (SMC) from canine vein grafts, 2 hours (H) to 30 days (D) following surgery. Our results demonstrate a robust genomic response beginning at 2 H, peaking at 12–24 H, declining by 7 D, and resolving by 30 D. Gene ontology and pathway analyses of differentially expressed genes indicated that implantation injury affects inflammatory and immune responses, apoptosis, mitosis, and extracellular matrix reorganization in both cell types. Through backpropagation an integrated network was built, starting with genes differentially expressed at 30 D, followed by adding upstream interactive genes from each prior time-point. This identified significant enrichment of IL-6, IL-8, NF-κB, dendritic cell maturation, glucocorticoid receptor, and Triggering Receptor Expressed on Myeloid Cells (TREM-1) signaling, as well as PPARα activation pathways in graft EC and SMC. Interactive network-based analyses identified IL-6, IL-8, IL-1α, and Insulin Receptor (INSR) as focus hub genes within these pathways. Real-time PCR was used for the validation of two of these genes: IL-6 and IL-8, in addition to Collagen 11A1 (COL11A1), a cornerstone of the backpropagation. In conclusion, these results establish causality relationships clarifying the pathogenesis of vein graft implantation injury, and identifying novel targets for its prevention.


Diabetes | 2016

Mast Cells Regulate Wound Healing in Diabetes

Ana Tellechea; Ermelindo C. Leal; Antonios Kafanas; Michael E. Auster; Sarada Kuchibhotla; Yana Ostrovsky; Francesco Tecilazich; Dimitrios Baltzis; Yongjun Zheng; Eugénia Carvalho; Janice M. Zabolotny; Zuyi Weng; Anastasia I. Petra; Arti B. Patel; Smaro Panagiotidou; Leena Pradhan-Nabzdyk; Theoharis C. Theoharides; Aristidis Veves

Diabetic foot ulceration is a severe complication of diabetes that lacks effective treatment. Mast cells (MCs) contribute to wound healing, but their role in diabetes skin complications is poorly understood. Here we show that the number of degranulated MCs is increased in unwounded forearm and foot skin of patients with diabetes and in unwounded dorsal skin of diabetic mice (P < 0.05). Conversely, postwounding MC degranulation increases in nondiabetic mice, but not in diabetic mice. Pretreatment with the MC degranulation inhibitor disodium cromoglycate rescues diabetes-associated wound-healing impairment in mice and shifts macrophages to the regenerative M2 phenotype (P < 0.05). Nevertheless, nondiabetic and diabetic mice deficient in MCs have delayed wound healing compared with their wild-type (WT) controls, implying that some MC mediator is needed for proper healing. MCs are a major source of vascular endothelial growth factor (VEGF) in mouse skin, but the level of VEGF is reduced in diabetic mouse skin, and its release from human MCs is reduced in hyperglycemic conditions. Topical treatment with the MC trigger substance P does not affect wound healing in MC-deficient mice, but improves it in WT mice. In conclusion, the presence of nondegranulated MCs in unwounded skin is required for proper wound healing, and therapies inhibiting MC degranulation could improve wound healing in diabetes.


The International Journal of Lower Extremity Wounds | 2015

Alginate and DNA Gels Are Suitable Delivery Systems for Diabetic Wound Healing

Ana Tellechea; Eduardo A. Silva; Jianghong Min; Ermelindo C. Leal; Michael E. Auster; Leena Pradhan-Nabzdyk; William M. Shih; David J. Mooney; Aristidis Veves

Diabetic foot ulcers (DFU) represent a severe health problem and an unmet clinical challenge. In this study, we tested the efficacy of novel biomaterials in improving wound healing in mouse models of diabetes mellitus (DM). The biomaterials are composed of alginate- and deoxyribonucleic acid (DNA)-based gels that allow incorporation of effector cells, such as outgrowth endothelial cells (OEC), and provide sustained release of bioactive factors, such as neuropeptides and growth factors, which have been previously validated in experimental models of DM wound healing or hind limb ischemia. We tested these biomaterials in mice and demonstrate that they are biocompatible and can be injected into the wound margins without major adverse effects. In addition, we show that the combination of OEC and the neuropeptide Substance P has a better healing outcome than the delivery of OEC alone, while subtherapeutic doses of vascular endothelial growth factor (VEGF) are required for the transplanted cells to exert their beneficial effects in wound healing. In summary, alginate and DNA scaffolds could serve as potential delivery systems for the next-generation DFU therapies.


Biomaterials | 2015

pH-responsive scaffolds generate a pro-healing response

Jin-Oh You; Marjan Rafat; Dariela Almeda; Natalia Maldonado; Peng Guo; Christoph S. Nabzdyk; Maggie Chun; Frank W. LoGerfo; John W. Hutchinson; Leena Pradhan-Nabzdyk; Debra T. Auguste

A principal challenge in wound healing is a lack of cell recruitment, cell infiltration, and vascularization, which occurs in the absence of temporal and spatial cues. We hypothesized that a scaffold that expands due to local changes in pH may alter oxygen and nutrient transport and the local cell density, leading to enhanced cell deposition and survival. In this study, we present a pH-responsive scaffold that increases oxygen transport, as confirmed by our finite element model analysis, and cell proliferation relative to a non-responsive scaffold. In vivo, responsive scaffolds induce a pro-healing gene expression profile indicative of enhanced angiogenesis, granulation tissue formation, and tissue remodeling. Scaffolds that stretch in response to their environment may be a hallmark for tissue regeneration.


BMC Genomics | 2016

The effects of transfection reagent polyethyleneimine (PEI) and non-targeting control siRNAs on global gene expression in human aortic smooth muscle cells

Nurazhani A. Raof; Deepa Rajamani; Hsun-Chieh Chu; Aniket Gurav; Joel M. Johnson; Frank W. LoGerfo; Manoj Bhasin; Leena Pradhan-Nabzdyk

BackgroundRNA interference (RNAi) is a powerful platform utilized to target transcription of specific genes and downregulate the protein product. To achieve effective silencing, RNAi is usually applied to cells or tissue with a transfection reagent to enhance entry into cells. A commonly used control is the same transfection reagent plus a “noncoding RNAi”. However, this does not control for the genomic response to the transfection reagent alone or in combination with the noncoding RNAi. These control effects while not directly targeting the gene in question may influence expression of other genes that in turn alter expression of the target. The current study was prompted by our work focused on prevention of vascular bypass graft failure and our experience with gene silencing in human aortic smooth muscle cells (HAoSMCs) where we suspected that off target effects through this mechanism might be substantial. We have used Next Generation Sequencing (NGS) technology and bioinformatics analysis to examine the genomic response of HAoSMCs to the transfection reagent alone (polyethyleneimine (PEI)) or in combination with commercially obtained control small interfering RNA (siRNAs) (Dharmacon and Invitrogen).ResultsCompared to untreated cells, global gene expression of HAoSMcs after transfection either with PEI or in combination with control siRNAs displayed significant alterations in gene transcriptome after 24 h. HAoSMCs transfected by PEI alone revealed alterations of 213 genes mainly involved in inflammatory and immune responses. HAoSMCs transfected by PEI complexed with siRNA from either Dharmacon or Invitrogen showed substantial gene variation of 113 and 85 genes respectively. Transfection of cells with only PEI or with PEI and control siRNAs resulted in identification of 20 set of overlapping altered genes. Further, systems biology analysis revealed key master regulators in cells transfected with control siRNAs including the cytokine, Interleukin (IL)-1, transcription factor GATA Binding Protein (GATA)-4 and the methylation enzyme, Enhancer of zeste homolog 2 (EZH-2) a cytokine with an apical role in initiating the inflammatory response.ConclusionsSignificant off-target effects in HAoSMCs transfected with PEI alone or in combination with control siRNAs may lead to misleading conclusions concerning the effectiveness of a targeted siRNA strategy. The lack of structural information about transfection reagents and “non coding” siRNA is a hindrance in the development of siRNA based therapeutics.


Journal of Nanomaterials | 2015

Development of a composite electrospun polyethylene terephthalate-polyglycolic acid material: potential use as a drug-eluting vascular graft

Christoph S. Nabzdyk; Maggie Chun; Saif G. Pathan; David W. Nelson; Jin-Oh You; Matthew D. Phaneuf; Frank W. LoGerfo; Leena Pradhan-Nabzdyk

Intimal hyperplasia (IH), an excessive wound healing response of an injured vessel wall after bypass grafting, typically leads to prosthetic bypass graft failure. In an approach to ameliorate IH, nondegradable poly(ethylene terephthalate) or PET, which has been used in prosthetic vascular grafts for over 60 years, and biodegradable poly(glycolic acid) or PGA were electrospun using different techniques to generate a material that may serve as permanent scaffold and as a drug/biologic delivery device. PET and PGA polymers were electrospun from either a single-blended solution (ePET/ePGA-s) or two separate polymer solutions (ePET/ePGA-d). ePET/ePGA-d material revealed two distinct fibers and was significantly stronger than the single fiber ePET/ePGA-s material. After 21 days of incubation in PBS, ePET-PGA-s showed fiber strand breaks likely due to the degradation of the PGA within the ePET-ePGA-s fiber, while the ePET/ePGA-d material showed intact ePET fibers even after ePGA fiber degradation. The ePET/ePGA- material was able to release red fluorescent dye for at least 14 days. Attachment of human aortic smooth muscle cells (AoSMCs) was similar to both materials. ePET/ePGA-d materials maybe a step towards bypass graft materials that can be custom-designed to promote cellular attachment while serving as a drug delivery platform for IH prevention.


Journal of Translational Medicine | 2017

RNAi therapy to the wall of arteries and veins: anatomical, physiologic, and pharmacological considerations

Christoph S. Nabzdyk; Leena Pradhan-Nabzdyk; Frank W. LoGerfo

BackgroundCardiovascular disease remains a major health care challenge. The knowledge about the underlying mechanisms of the respective vascular disease etiologies has greatly expanded over the last decades. This includes the contribution of microRNAs, endogenous non-coding RNA molecules, known to vastly influence gene expression. In addition, short interference RNA has been established as a mechanism to temporarily affect gene expression. This review discusses challenges relating to the design of a RNA interference therapy strategy for the modulation of vascular disease. Despite advances in medical and surgical therapies, atherosclerosis (ATH), aortic aneurysms (AA) are still associated with high morbidity and mortality. In addition, intimal hyperplasia (IH) remains a leading cause of late vein and prosthetic bypass graft failure. Pathomechanisms of all three entities include activation of endothelial cells (EC) and dedifferentiation of vascular smooth muscle cells (VSMC). RNA interference represents a promising technology that may be utilized to silence genes contributing to ATH, AA or IH. Successful RNAi delivery to the vessel wall faces multiple obstacles. These include the challenge of cell specific, targeted delivery of RNAi, anatomical barriers such as basal membrane, elastic laminae in arterial walls, multiple layers of VSMC, as well as adventitial tissues. Another major decision point is the route of delivery and potential methods of transfection. A plethora of transfection reagents and adjuncts have been described with varying efficacies and side effects. Timing and duration of RNAi therapy as well as target gene choice are further relevant aspects that need to be addressed in a temporo-spatial fashion.ConclusionsWhile multiple preclinical studies reported encouraging results of RNAi delivery to the vascular wall, it remains to be seen if a single target can be sufficient to the achieve clinically desirable changes in the injured vascular wall in humans. It might be necessary to achieve simultaneous and/or sequential silencing of multiple, synergistically acting target genes. Some advances in cell specific RNAi delivery have been made, but a reliable vascular cell specific transfection strategy is still missing. Also, off-target effects of RNAi and unwanted effects of transfection agents on gene expression are challenges to be addressed. Close collaborative efforts between clinicians, geneticists, biologists, and chemical and medical engineers will be needed to provide tailored therapeutics for the various types of vascular diseases.

Collaboration


Dive into the Leena Pradhan-Nabzdyk's collaboration.

Top Co-Authors

Avatar

Frank W. LoGerfo

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ana Tellechea

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Aristidis Veves

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Christoph S. Nabzdyk

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mauricio A. Contreras

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Maggie Chun

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Matthew D. Phaneuf

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Antonios Kafanas

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Francesco Tecilazich

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge