Network


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

Hotspot


Dive into the research topics where Andrew Hertsenberg is active.

Publication


Featured researches published by Andrew Hertsenberg.


Science Translational Medicine | 2014

Human limbal biopsy-derived stromal stem cells prevent corneal scarring.

Sayan Basu; Andrew Hertsenberg; Martha L. Funderburgh; Michael K. Burrow; Mary M. Mann; Yiqin Du; Kira L. Lathrop; Fatima N. Syed-Picard; Sheila M. Adams; David E. Birk; James L. Funderburgh

Human stromal stem cells isolated from limbal biopsies prevented corneal scarring in a murine model of corneal wounding. All Eyes on Limbal Stem Cells Our corneas—transparent, collagen-based structures that allow us to see—are easily damaged by trauma and infection, resulting in scarring and, in many cases, blindness. Although corneal transplant is the clinical norm, adverse immune responses and a shortage of cornea donors are serious limitations. Basu and colleagues devised a personalized cell-based, nonsurgical approach to prevent corneal scarring. They obtained mesenchymal stem cells from the human limbus (the region between the cornea and the sclera) and confirmed that they could be differentiated into keratocytes (corneal cells) in vitro. The human limbal biopsy–derived stromal cells, or LBSCs, were then placed in a fibrin gel and applied to the surface of debridement wounds in mice. The LBSCs were able to regenerate damaged stromal tissue in the animals, resembling native corneal tissue. Because these cells can be obtained directly from the patient and because fibrin-based products are already used in people, this approach could translate soon to treat stromal scarring, a major cause of corneal blindness. Conventional allograft therapy for corneal scarring is widespread and successful, but donor tissue is not universally available, and some grafts fail owing to rejection and complications such as endothelial failure. We investigated direct treatment of corneal scarring using autologous stem cells, a therapy that, if successful, could reduce the need for corneal grafts. Mesenchymal cells were expanded from small superficial, clinically replicable limbal biopsies of human cadaveric corneo-scleral rims. Limbal biopsy–derived stromal cells (LBSCs) expanded rapidly in media containing human serum, were highly clonogenic, and could generate spheres expressing stem cell genes (ABCG2, Nestin, NGFR, Oct4, PAX6, and Sox2). Human LBSCs differentiated into keratocytes expressing characteristic marker genes (ALDH3A1, AQP1, KERA, and PTGDS) and organized a thick lamellar stroma-like tissue containing aligned collagen and keratan sulfate proteoglycans when cultured on aligned nanofiber substrata. When engrafted into mouse corneal wounds, LBSCs prevented formation of light-scattering scar tissue containing fibrotic matrix components. The presence of LBSCs induced regeneration of ablated stroma with tissue exhibiting lamellar structure and collagen organization indistinguishable from that of native tissue. Because the limbus can be easily biopsied from either eye of an affected individual and LBSCs capable of corneal stromal remodeling can be expanded under xeno-free autologous conditions, these cells present a potential for autologous stem cell–based treatment of corneal stromal blindness.


PLOS ONE | 2013

Differentiation of Human Embryonic Stem Cells into Cells with Corneal Keratocyte Phenotype

Audrey A. Chan; Andrew Hertsenberg; Martha L. Funderburgh; Mary M. Mann; Yiqin Du; K. Davoli; Jocelyn Danielle Mich-Basso; Lei Yang; James L. Funderburgh

Corneal transparency depends on a unique extracellular matrix secreted by stromal keratocytes, mesenchymal cells of neural crest lineage. Derivation of keratocytes from human embryonic stem (hES) cells could elucidate the keratocyte developmental pathway and open a potential for cell-based therapy for corneal blindness. This study seeks to identify conditions inducing differentiation of pluripotent hES cells to the keratocyte lineage. Neural differentiation of hES cell line WA01(H1) was induced by co-culture with mouse PA6 fibroblasts. After 6 days of co-culture, hES cells expressing cell-surface NGFR protein (CD271, p75NTR) were isolated by immunoaffinity adsorption, and cultured as a monolayer for one week. Keratocyte phenotype was induced by substratum-independent pellet culture in serum-free medium containing ascorbate. Gene expression, examined by quantitative RT-PCR, found hES cells co-cultured with PA6 cells for 6 days to upregulate expression of neural crest genes including NGFR, SNAI1, NTRK3, SOX9, and MSX1. Isolated NGFR-expressing cells were free of PA6 feeder cells. After expansion as a monolayer, mRNAs typifying adult stromal stem cells were detected, including BMI1, KIT, NES, NOTCH1, and SIX2. When these cells were cultured as substratum-free pellets keratocyte markers AQP1, B3GNT7, PTDGS, and ALDH3A1 were upregulated. mRNA for keratocan (KERA), a cornea-specific proteoglycan, was upregulated more than 10,000 fold. Culture medium from pellets contained high molecular weight keratocan modified with keratan sulfate, a unique molecular component of corneal stroma. These results show hES cells can be induced to differentiate into keratocytes in vitro. Pluripotent stem cells, therefore, may provide a renewable source of material for development of treatment of corneal stromal opacities.


Methods of Molecular Biology | 2015

Generation of Corneal Keratocytes from Human Embryonic Stem Cells

Andrew Hertsenberg; James L. Funderburgh

Human Embryonic Stem Cells (hESC) offer an important resource as a limitless supply of any differentiated cell type of the human body. Keratocytes, cells from the corneal stroma, may have the potential for restoration of vision in cell therapy and biomedical engineering applications, but these specialized cells are not readily expanded in vitro. Here we describe a two-part method to produce keratocytes from the H1 hESC cell line. The hESC cells, maintained and expanded in feeder-free culture medium are first differentiated to neural crest cells using the stromal-derived inducing activity (SDIA) of the PA6 mouse embryonic fibroblast cell line. The resulting neural crest cells are selected by their expression of cell-surface CD271 and subsequently cultured as 3D pellets in a defined differentiation medium to induce a keratocyte phenotype.


PLOS ONE | 2017

Corneal stromal stem cells reduce corneal scarring by mediating neutrophil infiltration after wounding

Andrew Hertsenberg; Golnar Shojaati; Martha L. Funderburgh; Mary M. Mann; Yiqin Du; James L. Funderburgh

Corneal scarring limits vision for millions of individuals worldwide. Corneal transplantation (keratoplasty) is the standard of care for corneal opacity; however, it bears the risk of graft rejection and infection and is not universally available. Stem cell therapy holds promise as an alternative to keratoplasty. Stem cells from human corneal stroma (CSSC) induce regeneration of transparent corneal tissue in a mouse wound-healing model. In this study we investigated the mechanism by which CSSC prevent deposition of fibrotic tissue. Infiltration by CD11b+/Ly6G+ neutrophils and myeloperoxidase expression were increased in corneas 24 hr after corneal wounding but were reduced in CSSC-treated wounds. Secretion of TSG-6, a protein known to regulate neutrophil migration, was up-regulated in CSSC in response to TNFα and as CSSC differentiate to keratocytes. In vivo, wounded mouse corneas treated with CSSC contained human TSG-6. Inhibition of neutrophil infiltration into cornea by CSSC was reversed when TSG-6 expression was knocked down using siRNA. Silencing of TSG-6 expression in CSSC reduced their ability to block scarring and the expression of mRNA for fibrosis-associated proteins collagen III, tenascin C, and smooth muscle actin in wounded corneas. Neutropenic mice exhibited a significant reduction in corneal scarring and fibrotic mRNA expression 2 weeks after wounding. These results support the conclusion that neutrophil infiltration is an essential event in the fibrotic response to corneal damage and that prevention of scarring by CSSC is mediated by secretion of TSG-6 by these cells.


Journal of Tissue Engineering and Regenerative Medicine | 2018

Scaffold‐free tissue engineering of functional corneal stromal tissue

Fatima N. Syed-Picard; Yiqin Du; Andrew Hertsenberg; Rachelle Palchesko; Martha L. Funderburgh; Adam W. Feinberg; James L. Funderburgh

Blinding corneal scarring is predominately treated with allogeneic graft tissue; however, there is a worldwide shortage of donor tissue leaving millions in need of therapy. Human corneal stromal stem cells (CSSC) have been shown produce corneal tissue when cultured on nanofibre scaffolding, but this tissue cannot be readily separated from the scaffold. In this study, scaffold‐free tissue engineering methods were used to generate biomimetic corneal stromal tissue constructs that can be transplanted in vivo without introducing the additional variables associated with exogenous scaffolding. CSSC were cultured on substrates with aligned microgrooves, which directed parallel cell alignment and matrix organization, similar to the organization of native corneal stromal lamella. CSSC produced sufficient matrix to allow manual separation of a tissue sheet from the grooved substrate. These constructs were cellular and collagenous tissue sheets, approximately 4 μm thick and contained extracellular matrix molecules typical of corneal tissue including collagen types I and V and keratocan. Similar to the native corneal stroma, the engineered corneal tissues contained long parallel collagen fibrils with uniform diameter. After being transplanted into mouse corneal stromal pockets, the engineered corneal stromal tissues became transparent, and the human CSSCs continued to express human corneal stromal matrix molecules. Both in vitro and in vivo, these scaffold‐free engineered constructs emulated stromal lamellae of native corneal stromal tissues. Scaffold‐free engineered corneal stromal constructs represent a novel, potentially autologous, cell‐generated, biomaterial with the potential for treating corneal blindness. Copyright


Progress in Molecular Biology and Translational Science | 2015

Stem Cells in the Cornea

Andrew Hertsenberg; James L. Funderburgh

The cornea is the tough, transparent tissue through which light first enters the eye and functions as a barrier to debris and infection as well as two-thirds of the refractive power of the eye. Corneal damage that is not promptly treated will often lead to scarring and vision impairment. Due to the limited options currently available to treat corneal scars, the identification and isolation of stem cells in the cornea has received much attention, as they may have potential for autologous, cell-based approaches to the treatment of damaged corneal tissue.


Investigative Ophthalmology & Visual Science | 2015

Scaffold-free engineering of stromal lamellar tissue

Fatima N. Syed-Picard; Yiqin Du; Rachelle Palchesko; Martha L. Funderburgh; Andrew Hertsenberg; Adam W. Feinberg; James L. Funderburgh


Investigative Ophthalmology & Visual Science | 2015

Stem cells from corneal stroma suppress T-cell activation via cell-cell interactions

James L. Funderburgh; Martha L. Funderburgh; Mary M. Mann; Yiqin Du; Kyle C. McKenna; Andrew Hertsenberg


Investigative Ophthalmology & Visual Science | 2015

Stromal stem cells prevent corneal scarring after trauma by upregulation of TSG-6 expression.

Andrew Hertsenberg; Michael K. Burrow; James L. Funderburgh


Investigative Ophthalmology & Visual Science | 2014

Prevention of Corneal Scarring by Human Corneal Stromal Stem Cells

Andrew Hertsenberg; Sayan Basu; James L. Funderburgh

Collaboration


Dive into the Andrew Hertsenberg's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yiqin Du

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Mary M. Mann

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sayan Basu

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Adam W. Feinberg

Carnegie Mellon University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge