Network


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

Hotspot


Dive into the research topics where Keith Yeager is active.

Publication


Featured researches published by Keith Yeager.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Engineering anatomically shaped human bone grafts

Warren L. Grayson; Mirjam Fröhlich; Keith Yeager; Sarindr Bhumiratana; M. Ete Chan; Christopher Cannizzaro; Leo Q. Wan; X. Sherry Liu; X. Edward Guo; Gordana Vunjak-Novakovic

The ability to engineer anatomically correct pieces of viable and functional human bone would have tremendous potential for bone reconstructions after congenital defects, cancer resections, and trauma. We report that clinically sized, anatomically shaped, viable human bone grafts can be engineered by using human mesenchymal stem cells (hMSCs) and a “biomimetic” scaffold-bioreactor system. We selected the temporomandibular joint (TMJ) condylar bone as our tissue model, because of its clinical importance and the challenges associated with its complex shape. Anatomically shaped scaffolds were generated from fully decellularized trabecular bone by using digitized clinical images, seeded with hMSCs, and cultured with interstitial flow of culture medium. A bioreactor with a chamber in the exact shape of a human TMJ was designed for controllable perfusion throughout the engineered construct. By 5 weeks of cultivation, tissue growth was evidenced by the formation of confluent layers of lamellar bone (by scanning electron microscopy), markedly increased volume of mineralized matrix (by quantitative microcomputer tomography), and the formation of osteoids (histologically). Within bone grafts of this size and complexity cells were fully viable at a physiologic density, likely an important factor of graft function. Moreover, the density and architecture of bone matrix correlated with the intensity and pattern of the interstitial flow, as determined in experimental and modeling studies. This approach has potential to overcome a critical hurdle—in vitro cultivation of viable bone grafts of complex geometries—to provide patient-specific bone grafts for craniofacial and orthopedic reconstructions.


Journal of Tissue Engineering and Regenerative Medicine | 2012

Biomimetic perfusion and electrical stimulation applied in concert improved the assembly of engineered cardiac tissue

Robert Maidhof; Nina Tandon; Eun Jung Lee; Jianwen Luo; Yi Duan; Keith Yeager; Elisa E. Konofagou; Gordana Vunjak-Novakovic

Maintenance of normal myocardial function depends intimately on synchronous tissue contraction, driven by electrical activation and on adequate nutrient perfusion in support thereof. Bioreactors have been used to mimic aspects of these factors in vitro to engineer cardiac tissue but, due to design limitations, previous bioreactor systems have yet to simultaneously support nutrient perfusion, electrical stimulation and unconstrained (i.e. not isometric) tissue contraction. To the best of our knowledge, the bioreactor system described herein is the first to integrate these three key factors in concert. We present the design of our bioreactor and characterize its capability in integrated experimental and mathematical modelling studies. We then cultured cardiac cells obtained from neonatal rats in porous, channelled elastomer scaffolds with the simultaneous application of perfusion and electrical stimulation, with controls excluding either one or both of these two conditions. After 8 days of culture, constructs grown with simultaneous perfusion and electrical stimulation exhibited substantially improved functional properties, as evidenced by a significant increase in contraction amplitude (0.23 ± 0.10% vs 0.14 ± 0.05%, 0.13 ± 0.08% or 0.09 ± 0.02% in control constructs grown without stimulation, without perfusion, or either stimulation or perfusion, respectively). Consistently, these constructs had significantly improved DNA contents, cell distribution throughout the scaffold thickness, cardiac protein expression, cell morphology and overall tissue organization compared to control groups. Thus, the simultaneous application of medium perfusion and electrical conditioning enabled by the use of the novel bioreactor system may accelerate the generation of fully functional, clinically sized cardiac tissue constructs. Copyright


Journal of Biomechanics | 2008

Dynamic loading of deformable porous media can induce active solute transport

Michael B. Albro; Nadeen O. Chahine; Roland Li; Keith Yeager; Clark T. Hung; Gerard A. Ateshian

Active solute transport mediated by molecular motors across porous membranes is a well-recognized mechanism for transport across the cell membrane. In contrast, active transport mediated by mechanical loading of porous media is a non-intuitive mechanism that has only been predicted recently from theory, but not yet observed experimentally. This study uses agarose hydrogel and dextran molecules as a model experimental system to explore this mechanism. Results show that dynamic loading can enhance the uptake of dextran by a factor greater than 15 over passive diffusion, for certain combinations of gel concentration and dextran molecular weight. Upon cessation of loading, the concentration reverts back to that achieved under passive diffusion. Thus, active solute transport in porous media can indeed be mediated by cyclical mechanical loading.


Science Translational Medicine | 2016

Tissue-engineered autologous grafts for facial bone reconstruction.

Sarindr Bhumiratana; Jonathan Bernhard; David M. Alfi; Keith Yeager; Ryan E. Eton; Jonathan F. Bova; Forum Shah; Jeffrey M. Gimble; Mandi J. Lopez; Sidney B. Eisig; Gordana Vunjak-Novakovic

Anatomically shaped living bone formed in a portable bioreactor using autologous cells and bone matrix repaired the facial ramus-condyle unit in pigs. Saving face Restoring your reputation after a social gaffe may be challenging, but perhaps welcomed in comparison to saving face through restoration of actual bone structure. A delicate and precise process, facial bone reconstruction currently uses bone grafts from the same patient. Cell- and biomaterial-based approaches could benefit this field by providing personalized grafts for deformities of all shapes and sizes. Bhumiratana and colleagues therefore designed a maxillofacial reconstructive strategy centered on a combination of stem cells, decellularized bone, and a custom-designed perfusion bioreactor. The bone was first shaped to the defect in the ramus-condyle unit of minipigs, which have similar jaw anatomies and weight-bearing properties as humans. Then, stem cells were cultured on the bone for several weeks. To mimic the manufacturing and transport chain that could be involved in reconstructing human facial bones, then authors shipped the bioreactor with the living bone inside the site of surgery. Paired histological and image analysis showed that the implanted scaffold material integrated with host tissue, formed new bone, and was vascularized extensively, but only if cells were present. Growing such anatomically correct, large-scale bone constructs could vastly improve regenerative medicine options for patients with craniofacial bone deformities. Facial deformities require precise reconstruction of the appearance and function of the original tissue. The current standard of care—the use of bone harvested from another region in the body—has major limitations, including pain and comorbidities associated with surgery. We have engineered one of the most geometrically complex facial bones by using autologous stromal/stem cells, native bovine bone matrix, and a perfusion bioreactor for the growth and transport of living grafts, without bone morphogenetic proteins. The ramus-condyle unit, the most eminent load-bearing bone in the skull, was reconstructed using an image-guided personalized approach in skeletally mature Yucatán minipigs (human-scale preclinical model). We used clinically approved decellularized bovine trabecular bone as a scaffolding material and crafted it into an anatomically correct shape using image-guided micromilling to fit the defect. Autologous adipose-derived stromal/stem cells were seeded into the scaffold and cultured in perfusion for 3 weeks in a specialized bioreactor to form immature bone tissue. Six months after implantation, the engineered grafts maintained their anatomical structure, integrated with native tissues, and generated greater volume of new bone and greater vascular infiltration than either nonseeded anatomical scaffolds or untreated defects. This translational study demonstrates feasibility of facial bone reconstruction using autologous, anatomically shaped, living grafts formed in vitro, and presents a platform for personalized bone tissue engineering.


Nature | 2018

Advanced maturation of human cardiac tissue grown from pluripotent stem cells

Kacey Ronaldson-Bouchard; Stephen P. Ma; Keith Yeager; Timothy Chen; LouJin Song; Dario Sirabella; Kumi Morikawa; Diogo Teles; Masayuki Yazawa; Gordana Vunjak-Novakovic

Cardiac tissues generated from human induced pluripotent stem cells (iPSCs) can serve as platforms for patient-specific studies of physiology and disease1–6. However, the predictive power of these models is presently limited by the immature state of the cells1, 2, 5, 6. Here we show that this fundamental limitation can be overcome if cardiac tissues are formed from early-stage iPSC-derived cardiomyocytes soon after the initiation of spontaneous contractions and are subjected to physical conditioning with increasing intensity over time. After only four weeks of culture, for all iPSC lines studied, such tissues displayed adult-like gene expression profiles, remarkably organized ultrastructure, physiological sarcomere length (2.2 µm) and density of mitochondria (30%), the presence of transverse tubules, oxidative metabolism, a positive force–frequency relationship and functional calcium handling. Electromechanical properties developed more slowly and did not achieve the stage of maturity seen in adult human myocardium. Tissue maturity was necessary for achieving physiological responses to isoproterenol and recapitulating pathological hypertrophy, supporting the utility of this tissue model for studies of cardiac development and disease.A tissue culture system that provides an increasing intensity of electromechanical stimulation over time enables an in vitro model of cardiac tissue derived from human induced pluripotent stem cells to develop many of the characteristics of adult cardiac tissue.


Journal of Biomechanics | 2014

Combined effects of chemical priming and mechanical stimulation on mesenchymal stem cell differentiation on nanofiber scaffolds

Siddarth D. Subramony; Amanda Su; Keith Yeager; Helen H. Lu

Functional tissue engineering of connective tissues such as the anterior cruciate ligament (ACL) remains a significant clinical challenge, largely due to the need for mechanically competent scaffold systems for grafting, as well as a reliable cell source for tissue formation. We have designed an aligned, polylactide-co-glycolide (PLGA) nanofiber-based scaffold with physiologically relevant mechanical properties for ligament regeneration. The objective of this study is to identify optimal tissue engineering strategies for fibroblastic induction of human mesenchymal stem cells (hMSC), testing the hypothesis that basic fibroblast growth factor (bFGF) priming coupled with tensile loading will enhance hMSC-mediated ligament regeneration. It was observed that compared to the unloaded, as well as growth factor-primed but unloaded controls, bFGF stimulation followed by physiologically relevant tensile loading enhanced hMSC proliferation, collagen production and subsequent differentiation into ligament fibroblast-like cells, upregulating the expression of types I and III collagen, as well as tenasin-C and tenomodulin. The results of this study suggest that bFGF priming increases cell proliferation, while mechanical stimulation of the hMSCs on the aligned nanofiber scaffold promotes fibroblastic induction of these cells. In addition to demonstrating the potential of nanofiber scaffolds for hMSC-mediated functional ligament tissue engineering, this study yields new insights into the interactive effects of chemical and mechanical stimuli on stem cell differentiation.


Methods of Molecular Biology | 2014

Bioreactor Cultivation of Anatomically Shaped Human Bone Grafts

Joshua P. Temple; Keith Yeager; Sarindr Bhumiratana; Gordana Vunjak-Novakovic; Warren L. Grayson

In this chapter, we describe a method for engineering bone grafts in vitro with the specific geometry of the temporomandibular joint (TMJ) condyle. The anatomical geometry of the bone grafts was segmented from computed tomography (CT) scans, converted to G-code, and used to machine decellularized trabecular bone scaffolds into the identical shape of the condyle. These scaffolds were seeded with human bone marrow-derived mesenchymal stem cells (MSCs) using spinner flasks and cultivated for up to 5 weeks in vitro using a custom-designed perfusion bioreactor system. The flow patterns through the complex geometry were modeled using the FloWorks module of SolidWorks to optimize bioreactor design. The perfused scaffolds exhibited significantly higher cellular content, better matrix production, and increased bone mineral deposition relative to non-perfused (static) controls after 5 weeks of in vitro cultivation. This technology is broadly applicable for creating patient-specific bone grafts of varying shapes and sizes.


international conference of the ieee engineering in medicine and biology society | 2009

An integrated motion capture system for evaluation of neuromuscular disease patients

Viktor Gamarnik; Shu Pan; Jared Malke; Casey Chiu; Ben Koo; Jacqueline Montes; Keith Yeager; Jonathan Marra; Sally Dunaway; Megan Montgomery; Nancy Strauss; Darryl C. De Vivo; Petra Kaufmann; Barclay Morrison; Elisa E. Konofagou

There currently exist a variety of methods for evaluating movement in patients suffering from neuromuscular diseases (NMD). These tests are primarily performed in the clinical setting and evaluated by highly trained individuals, rather than evaluating patient in their natural environments (i.e., home or school). Currently available automated motion capture modalities offer a highly accurate means of assessing general motion, but are also limited to a highly controlled setting. Recent advances in MEMS technology have introduced the possibility of robust motion capture in uncontrolled environments, while minimizing user interference with self-initiated motion, especially in weaker subjects. The goal of this study is to design and evaluate a MEMS-sensor-based system for motion capture in the NMD patient population. The highly interdisciplinary effort has led to significant progress toward the implementation of a new device, which is accurate, clinically relevant, and highly affordable.


international conference of the ieee engineering in medicine and biology society | 2015

Mimicking biophysical stimuli within bone tumor microenvironment.

Alessandro Marturano-Kruik; Keith Yeager; D. Bach; Aranzazu Villasante; Elisa Cimetta; Gordana Vunjak-Novakovic

In vivo, cells reside in a complex environment regulating their fate and function. Most of this complexity is lacking in standard in vitro models, leading to readouts falling short of predicting the actual in vivo situation. The use of engineering tools, combined with deep biological knowledge, leads to the development and use of bioreactors providing biologically sound niches. Such bioreactors offer new tools for biological research, and are now also entering the field of cancer research. Here we present the development and validation of a modular bioreactor system providing: (i) high throughput analyses, (ii) a range of biological conditions, (iii) high degree of control, and (iv) application of physiological stimuli to the cultured samples. The bioreactor was used to engineer a three-dimensional (3D) tissue model of cancer, where the effects of mechanical stimulation on the tumor phenotype were evaluated. Mechanical stimuli applied to the engineered tumor model activated the mechanotransduction machinery and resulted in measurable changes of mRNA levels towards a more aggressive tumor phenotype.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Human bone perivascular niche-on-a-chip for studying metastatic colonization

Alessandro Marturano-Kruik; Michele M. Nava; Keith Yeager; Alan Chramiec; Luke Hao; Samuel Robinson; Edward Guo; Manuela Teresa Raimondi; Gordana Vunjak-Novakovic

Significance Improved human preclinical models are needed to better predict patients’ responses to anticancer drugs. Increasing the complexity of models may be a successful strategy only if crucial components of a tumor are identified, replicated, and controlled in vitro. We developed a perivascular niche to study metastatic colonization of the bone. Using a microfluidic chip, we exposed the niche to interstitial flow, oxygen gradients, and external forces, and established a dense vascular network. Cancer cells colonizing the bone resisted targeted therapy by entering a slow proliferative state. We expect that microfluidic niche-on-chip models will facilitate the development of drugs targeting persistent tumor cells into the bone and help manage the risk of metastatic relapse. Eight out of 10 breast cancer patients die within 5 years after the primary tumor has spread to the bones. Tumor cells disseminated from the breast roam the vasculature, colonizing perivascular niches around blood capillaries. Slow flows support the niche maintenance by driving the oxygen, nutrients, and signaling factors from the blood into the interstitial tissue, while extracellular matrix, endothelial cells, and mesenchymal stem cells regulate metastatic homing. Here, we show the feasibility of developing a perfused bone perivascular niche-on-a-chip to investigate the progression and drug resistance of breast cancer cells colonizing the bone. The model is a functional human triculture with stable vascular networks within a 3D native bone matrix cultured on a microfluidic chip. Providing the niche-on-a-chip with controlled flow velocities, shear stresses, and oxygen gradients, we established a long-lasting, self-assembled vascular network without supplementation of angiogenic factors. We further show that human bone marrow-derived mesenchymal stem cells, which have undergone phenotypical transition toward perivascular cell lineages, support the formation of capillary-like structures lining the vascular lumen. Finally, breast cancer cells exposed to interstitial flow within the bone perivascular niche-on-a-chip persist in a slow-proliferative state associated with increased drug resistance. We propose that the bone perivascular niche-on-a-chip with interstitial flow promotes the formation of stable vasculature and mediates cancer cell colonization.

Collaboration


Dive into the Keith Yeager's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anjali Saqi

Columbia University Medical Center

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
Researchain Logo
Decentralizing Knowledge