Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters










Publication year range
1.
Bioengineering (Basel) ; 11(4)2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38671729

ABSTRACT

Static cold storage (SCS), the current clinical gold standard for organ preservation, provides surgeons with a limited window of time between procurement and transplantation. In vascularized composite allotransplantation (VCA), this time limitation prevents many viable allografts from being designated to the best-matched recipients. Machine perfusion (MP) systems hold significant promise for extending and improving organ preservation. Most of the prior MP systems for VCA have been built and tested for large animal models. However, small animal models are beneficial for high-throughput biomolecular investigations. This study describes the design and development of a multiparametric bioreactor with a circuit customized to perfuse rat abdominal wall VCAs. To demonstrate its concept and functionality, this bioreactor system was employed in a small-scale demonstrative study in which biomolecular metrics pertaining to graft viability were evaluated non-invasively and in real time. We additionally report a low incidence of cell death from ischemic necrosis as well as minimal interstitial edema in machine perfused grafts. After up to 12 h of continuous perfusion, grafts were shown to survive transplantation and reperfusion, successfully integrating with recipient tissues and vasculature. Our multiparametric bioreactor system for rat abdominal wall VCA provides an advanced framework to test novel techniques to enhance normothermic and sub-normothermic VCA preservations in small animal models.

2.
bioRxiv ; 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38617372

ABSTRACT

Calvarial nerves, along with vasculature, influence skull formation during development and following injury, but it remains unclear how calvarial nerves are spatially distributed during postnatal growth and aging. Studying the spatial distribution of nerves in the skull remains challenging due to a lack of methods to image and quantify 3D structures in intact bone. To visualize calvarial 3D neurovascular architecture, we imaged nerves and endothelial cells with lightsheet microscopy. We employed machine-learning-based segmentation to facilitate high-resolution characterization from post-natal day 0 (P0) to Week 80 (80wk). We found that TUBB3+ nerve density decreased with aging with the frontal bone demonstrating earlier onset age-related nerve loss than the parietal bone. In addition, nerves in the periosteum and dura mater exhibited similar yet distinct temporal patterns of nerve growth and loss. While no difference was observed in TUBB3+ nerves during skeletal maturation (P0 → 12wk), we did observe an increase in the volume of unmyelinated nerves in the dura mater. Regarding calvarial vasculature, larger CD31hiEmcn- vessel density increased with aging, while CD31hiEmcnhi vessel density was reduced. For all nerve markers studied, calvarial nerves maintained a preferential spatial association with CD31hiEmcnhi vessels that decreased with aging. Additionally, we used a model of Apert syndrome that demonstrates early coronal suture fusion to explore the impact of suture-related disease on neurovascular architecture. We identified a mild dysregulation of dural nerves and minor shifts in vessel populations. Collectively, this 3D, spatiotemporal characterization of calvarial nerves throughout the lifespan and provides new insights into age-induced neurovascular architecture.

3.
Curr Dev Nutr ; 8(Suppl 1): 102027, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38476725

ABSTRACT

Populations in low- and middle-income countries (LMIC) typically consume less than the recommended daily amount of protein. Alternative protein (AP) sources could help combat malnutrition, but this requires careful consideration of elements needed to further establish AP products in LMIC. Key considerations include technological, nutritional, safety, social, and economic challenges. This perspective analyzes these considerations in achieving dietary diversity in LMIC, using a combination of traditional and novel protein sources with high nutritional value, namely, soy, mycoprotein, and cultivated meat. Technological approaches to modulate the technofunctionality and bitter off-tastes of plant-sourced proteins facilitate processing and ensure consumer acceptance. Economic considerations for inputs, infrastructure for production, and transportation represent key elements to scale up AP. Dietary diversification is indispensable and LMIC cannot rely on plant proteins alone to provide adequate protein intake sustainably. Investments in infrastructure and innovation are urgently needed to offer diverse sources of protein in LMIC.

4.
Angiogenesis ; 27(1): 105-119, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38032405

ABSTRACT

The healing of calvarial bone defects is a pressing clinical problem that involves the dynamic interplay between angiogenesis and osteogenesis within the osteogenic niche. Although structural and functional vascular remodeling (i.e., angiogenic evolution) in the osteogenic niche is a crucial modulator of oxygenation, inflammatory and bone precursor cells, most clinical and pre-clinical investigations have been limited to characterizing structural changes in the vasculature and bone. Therefore, we developed a new multimodality imaging approach that for the first time enabled the longitudinal (i.e., over four weeks) and dynamic characterization of multiple in vivo functional parameters in the remodeled vasculature and its effects on de novo osteogenesis, in a preclinical calvarial defect model. We employed multi-wavelength intrinsic optical signal (IOS) imaging to assess microvascular remodeling, intravascular oxygenation (SO2), and osteogenesis; laser speckle contrast (LSC) imaging to assess concomitant changes in blood flow and vascular maturity; and micro-computed tomography (µCT) to validate volumetric changes in calvarial bone. We found that angiogenic evolution was tightly coupled with calvarial bone regeneration and corresponded to distinct phases of bone healing, such as injury, hematoma formation, revascularization, and remodeling. The first three phases occurred during the initial two weeks of bone healing and were characterized by significant in vivo changes in vascular morphology, blood flow, oxygenation, and maturity. Overall, angiogenic evolution preceded osteogenesis, which only plateaued toward the end of bone healing (i.e., four weeks). Collectively, these data indicate the crucial role of angiogenic evolution in osteogenesis. We believe that such multimodality imaging approaches have the potential to inform the design of more efficacious tissue-engineering calvarial defect treatments.


Subject(s)
Bone Regeneration , Skull , X-Ray Microtomography , Skull/diagnostic imaging , Skull/blood supply , Skull/injuries , Bone Regeneration/physiology , Osteogenesis/physiology , Wound Healing
5.
J Transl Med ; 21(1): 609, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37684651

ABSTRACT

Vascularized composite allotransplantation can improve quality of life and restore functionality. However, the complex tissue composition of vascularized composite allografts (VCAs) presents unique clinical challenges that increase the likelihood of transplant rejection. Under prolonged static cold storage, highly damage-susceptible tissues such as muscle and nerve undergo irreversible degradation that may render allografts non-functional. Skin-containing VCA elicits an immunogenic response that increases the risk of recipient allograft rejection. The development of quantitative metrics to evaluate VCAs prior to and following transplantation are key to mitigating allograft rejection. Correspondingly, a broad range of bioanalytical methods have emerged to assess the progression of VCA rejection and characterize transplantation outcomes. To consolidate the current range of relevant technologies and expand on potential for development, methods to evaluate ex vivo VCA status are herein reviewed and comparatively assessed. The use of implantable physiological status monitoring biochips, non-invasive bioimpedance monitoring to assess edema, and deep learning algorithms to fuse disparate inputs to stratify VCAs are identified.


Subject(s)
Composite Tissue Allografts , Vascularized Composite Allotransplantation , Quality of Life , Transplantation, Homologous , Algorithms
6.
Adv Healthc Mater ; 12(29): e2301944, 2023 11.
Article in English | MEDLINE | ID: mdl-37565378

ABSTRACT

Porous tissue-engineered 3D-printed scaffolds are a compelling alternative to autografts for the treatment of large periorbital bone defects. Matching the defect-specific geometry has long been considered an optimal strategy to restore pre-injury anatomy. However, studies in large animal models have revealed that biomaterial-induced bone formation largely occurs around the scaffold periphery. Such ectopic bone formation in the periorbital region can affect vision and cause disfigurement. To enhance anatomic reconstruction, geometric mismatches are introduced in the scaffolds used to treat full thickness zygomatic defects created bilaterally in adult Yucatan minipigs. 3D-printed, anatomically-mirrored scaffolds are used in combination with autologous stromal vascular fraction of cells (SVF) for treatment. An advanced image-registration workflow is developed to quantify the post-surgical geometric mismatch and correlate it with the spatial pattern of the regenerating bone. Osteoconductive bone growth on the dorsal and ventral aspect of the defect enhances scaffold integration with the native bone while medio-lateral bone growth leads to failure of the scaffolds to integrate. A strong positive correlation is found between geometric mismatch and orthotopic bone deposition at the defect site. The data suggest that strategic mismatch >20% could improve bone scaffold design to promote enhanced regeneration, osseointegration, and long-term scaffold survivability.


Subject(s)
Printing, Three-Dimensional , Tissue Scaffolds , Swine , Animals , Swine, Miniature , Biocompatible Materials/pharmacology , Bone Regeneration , Osteogenesis
7.
Curr Osteoporos Rep ; 21(5): 503-518, 2023 10.
Article in English | MEDLINE | ID: mdl-37578676

ABSTRACT

PURPOSE OF REVIEW: This review examines the diverse functional relationships that exist between the peripheral nervous system (PNS) and bone, including key advances over the past century that inform our efforts to translate these discoveries for skeletal repair. RECENT FINDINGS: The innervation of the bone during development, homeostasis, and regeneration is highly patterned. Consistent with this, there have been nearly 100 studies over the past century that have used denervation approaches to isolate the effects of the different branches of the PNS on the bone. Overall, a common theme of balance emerges whereby an orchestration of both local and systemic neural functions must align to promote optimal skeletal repair while limiting negative consequences such as pain. An improved understanding of the functional bidirectional pathways linking the PNS and bone has important implications for skeletal development and regeneration. Clinical advances over the next century will necessitate a rigorous identification of the mechanisms underlying these effects that is cautious not to oversimplify the in vivo condition in diverse states of health and disease.


Subject(s)
Bone and Bones , Peripheral Nervous System , Humans
8.
Nat Chem Biol ; 19(9): 1127-1137, 2023 09.
Article in English | MEDLINE | ID: mdl-37024727

ABSTRACT

The interleukin-4 (IL-4) cytokine plays a critical role in modulating immune homeostasis. Although there is great interest in harnessing this cytokine as a therapeutic in natural or engineered formats, the clinical potential of native IL-4 is limited by its instability and pleiotropic actions. Here, we design IL-4 cytokine mimetics (denoted Neo-4) based on a de novo engineered IL-2 mimetic scaffold and demonstrate that these cytokines can recapitulate physiological functions of IL-4 in cellular and animal models. In contrast with natural IL-4, Neo-4 is hyperstable and signals exclusively through the type I IL-4 receptor complex, providing previously inaccessible insights into differential IL-4 signaling through type I versus type II receptors. Because of their hyperstability, our computationally designed mimetics can directly incorporate into sophisticated biomaterials that require heat processing, such as three-dimensional-printed scaffolds. Neo-4 should be broadly useful for interrogating IL-4 biology, and the design workflow will inform targeted cytokine therapeutic development.


Subject(s)
Cytokines , Interleukin-4 , Animals , Signal Transduction
9.
Bioengineering (Basel) ; 10(4)2023 Mar 29.
Article in English | MEDLINE | ID: mdl-37106621

ABSTRACT

Vascularized composite allotransplantation addresses injuries to complex anatomical structures such as the face, hand, and abdominal wall. Prolonged static cold storage of vascularized composite allografts (VCA) incurs damage and imposes transportation limits to their viability and availability. Tissue ischemia, the major clinical indication, is strongly correlated with negative transplantation outcomes. Machine perfusion and normothermia can extend preservation times. This perspective introduces multiplexed multi-electrode bioimpedance spectroscopy (MMBIS), an established bioanalytical method to quantify the interaction of the electrical current with tissue components, capable of measuring tissue edema, as a quantitative, noninvasive, real-time, continuous monitoring technique to provide crucially needed assessment of graft preservation efficacy and viability. MMBIS must be developed, and appropriate models explored to address the highly complex multi-tissue structures and time-temperature changes of VCA. Combined with artificial intelligence (AI), MMBIS can serve to stratify allografts for improvement in transplantation outcomes.

10.
Microvasc Res ; 148: 104518, 2023 07.
Article in English | MEDLINE | ID: mdl-36894024

ABSTRACT

Assessing intravascular blood oxygen saturation (SO2) is crucial for characterizing in vivo microenvironmental changes in preclinical models of injury and disease. However, most conventional optical imaging techniques for mapping in vivo SO2 assume or compute a single value of the optical path-length in tissue. This is especially detrimental when mapping in vivo SO2 in experimental disease or wound healing models that are characterized by vascular and tissue remodeling. Therefore, to circumvent this limitation we developed an in vivo SO2 mapping technique that utilizes hemoglobin-based intrinsic optical signal (IOS) imaging combined with a vascular-centric estimation of optical path-lengths. In vivo arterial and venous SO2 distributions derived with this approach closely matched those reported in the literature, while those derived using the single path-length (i.e. conventional) approach did not. Moreover, in vivo cerebrovascular SO2 strongly correlated (R2 > 0.7) with changes in systemic SO2 measured with a pulse oximeter during hypoxia and hyperoxia paradigms. Finally, in a calvarial bone healing model, in vivo SO2 assessed over four weeks was spatiotemporally correlated with angiogenesis and osteogenesis (R2 > 0.6). During the early stages of bone healing (i.e. day 10), angiogenic vessels surrounding the calvarial defect exhibited mean SO2 that was elevated by10 % (p < 0.05) relative to that observed at a later stage (i.e., day 26), indicative of their role in osteogenesis. These correlations were not evident with the conventional SO2 mapping approach. The feasibility of our wide field-of-view in vivo SO2 mapping approach illustrates its potential for characterizing the microvascular environment in applications ranging from tissue engineering to cancer.


Subject(s)
Hyperoxia , Oxygen Saturation , Humans , Oximetry/methods , Oxygen , Arteries
11.
Bioengineering (Basel) ; 9(11)2022 Nov 15.
Article in English | MEDLINE | ID: mdl-36421094

ABSTRACT

Tissue engineering strategies that combine human pluripotent stem cell-derived myogenic progenitors (hPDMs) with advanced biomaterials provide promising tools for engineering 3D skeletal muscle grafts to model tissue development in vitro and promote muscle regeneration in vivo. We recently demonstrated (i) the potential for obtaining large numbers of hPDMs using a combination of two small molecules without the overexpression of transgenes and (ii) the application of electrospun fibrin microfiber bundles for functional skeletal muscle restoration following volumetric muscle loss. In this study, we aimed to demonstrate that the biophysical cues provided by the fibrin microfiber bundles induce hPDMs to form engineered human skeletal muscle grafts containing multinucleated myotubes that express desmin and myosin heavy chains and that these grafts could promote regeneration following skeletal muscle injuries. We tested a genetic PAX7 reporter line (PAX7::GFP) to sort for more homogenous populations of hPDMs. RNA sequencing and gene set enrichment analyses confirmed that PAX7::GFP-sorted hPDMs exhibited high expression of myogenic genes. We tested engineered human skeletal muscle grafts derived from PAX7::GFP-sorted hPDMs within in vivo skeletal muscle defects by assessing myogenesis, engraftment and immunogenicity using immunohistochemical staining. The PAX7::GFP-sorted groups had moderately high vascular infiltration and more implanted cell association with embryonic myosin heavy chain (eMHC) regions, suggesting they induced pro-regenerative microenvironments. These findings demonstrated the promise for the use of PAX7::GFP-sorted hPDMs on fibrin microfiber bundles and provided some insights for improving the cell-biomaterial system to stimulate more robust in vivo skeletal muscle regeneration.

12.
Am J Physiol Cell Physiol ; 323(5): C1524-C1538, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36189973

ABSTRACT

Vascularization is a crucial step during musculoskeletal tissue regeneration via bioengineered constructs or grafts. Functional vasculature provides oxygen and nutrients to the graft microenvironment, facilitates wound healing, enhances graft integration with host tissue, and ensures the long-term survival of regenerating tissue. Therefore, imaging de novo vascularization (i.e., angiogenesis), changes in microvascular morphology, and the establishment and maintenance of perfusion within the graft site (i.e., vascular microenvironment or VME) can provide essential insights into engraftment, wound healing, as well as inform the design of tissue engineering (TE) constructs. In this review, we focus on state-of-the-art imaging approaches for monitoring the VME in craniofacial TE applications, as well as future advances in this field. We describe how cutting-edge in vivo and ex vivo imaging methods can yield invaluable information regarding VME parameters that can help characterize the effectiveness of different TE constructs and iteratively inform their design for enhanced craniofacial bone regeneration. Finally, we explicate how the integration of novel TE constructs, preclinical model systems, imaging techniques, and systems biology approaches could usher in an era of "image-based tissue engineering."


Subject(s)
Bone and Bones , Tissue Engineering , Humans , Tissue Engineering/methods , Bone Regeneration , Neovascularization, Pathologic , Wound Healing , Tissue Scaffolds , Neovascularization, Physiologic
13.
Biofabrication ; 14(3)2022 06 13.
Article in English | MEDLINE | ID: mdl-35617927

ABSTRACT

Porous Magnesium (Mg) is a promising biodegradable scaffold for treating critical-size bone defects, and as an essential element for human metabolism, Mg has shown sufficient biocompatibility. Its elastic moduli and yield strengths are closer to those of cortical bone than common, inert metallic implants, effectively reducing stress concentrations around host tissue as well as stress shielding. More importantly, Mg can degrade and be absorbed in the human body in a safe and controlled manner, thereby reducing the need for second surgeries to remove implants. The development of porous Mg scaffolds via conventional selective laser melting techniques has been limited due to Mg's low boiling point, high vapor pressures, high reactivity, and non-ideal microstructures in additively manufactured parts. Here we present an exciting alternative to conventional additive techniques: 3D weaving with Mg wires that have controlled chemistries and microstructures. The weaving process offers high throughput manufacturing as well as porous architectures that can be optimized for stiffness and porosity with topology optimization. Once woven, we dip-coat the weaves with polylactic acid to enhance their strength and corrosion resistance. Following fabrication, we characterize their mechanical properties, corrosion behavior, and cell compatibilityin vitro, and we use an intramuscular implantation model to evaluate theirin vivocorrosion behavior and tissue response.


Subject(s)
Magnesium , Prostheses and Implants , Bone and Bones , Elastic Modulus , Humans , Magnesium/chemistry , Porosity , Tissue Scaffolds/chemistry
14.
3D Print Med ; 8(1): 9, 2022 Apr 06.
Article in English | MEDLINE | ID: mdl-35384521

ABSTRACT

Bone tissue engineering strategies aimed at treating critical-sized craniofacial defects often utilize novel biomaterials and scaffolding. Rapid manufacturing of defect-matching geometries using 3D-printing strategies is a promising strategy to treat craniofacial bone loss to improve aesthetic and regenerative outcomes. To validate manufacturing quality, a robust, three-dimensional quality assurance pipeline is needed to provide an objective, quantitative metric of print quality if porous scaffolds are to be translated from laboratory to clinical settings. Previously published methods of assessing scaffold print quality utilized one- and two-dimensional measurements (e.g., strut widths, pore widths, and pore area) or, in some cases, the print quality of a single phantom is assumed to be representative of the quality of all subsequent prints. More robust volume correlation between anatomic shapes has been accomplished; however, it requires manual user correction in challenging cases such as porous objects like bone scaffolds. Here, we designed porous, anatomically-shaped scaffolds with homogenous or heterogenous porous structures. We 3D-printed the designs with acrylonitrile butadiene styrene (ABS) and used cone-beam computed tomography (CBCT) to obtain 3D image reconstructions. We applied the iterative closest point algorithm to superimpose the computational scaffold designs with the CBCT images to obtain a 3D volumetric overlap. In order to avoid false convergences while using an autonomous workflow for volumetric correlation, we developed an independent iterative closest point (I-ICP10) algorithm using MATLAB®, which applied ten initial conditions for the spatial orientation of the CBCT images relative to the original design. Following successful correlation, scaffold quality can be quantified and visualized on a sub-voxel scale for any part of the volume.

15.
Biomaterials ; 282: 121392, 2022 03.
Article in English | MEDLINE | ID: mdl-35134701

ABSTRACT

Critical-sized midfacial bone defects present a unique clinical challenge due to their complex three-dimensional shapes and intimate associations with sensory organs. To address this challenge, a point-of-care treatment strategy for functional, long-term regeneration of 2 cm full-thickness segmental defects in the zygomatic arches of Yucatan minipigs is evaluated. A digital workflow is used to 3D-print anatomically precise, porous, biodegradable scaffolds from clinical-grade poly-ε-caprolactone and decellularized bone composites. The autologous stromal vascular fraction of cells (SVF) is isolated from adipose tissue extracts and infused into the scaffolds that are implanted into the zygomatic ostectomies. Bone regeneration is assessed up to 52 weeks post-operatively in acellular (AC) and SVF groups (BV/DV = 0.64 ± 0.10 and 0.65 ± 0.10 respectively). In both treated groups, bone grows from the adjacent tissues and restores the native anatomy. Significantly higher torque is required to fracture the bone-scaffold interface in the SVF (7.11 ± 2.31 N m) compared to AC groups (2.83 ± 0.23 N m). Three-dimensional microcomputed tomography analysis reveals two distinct regenerative patterns: osteoconduction along the periphery of scaffolds to form dense lamellar bone and small islands of woven bone deposits growing along the struts in the scaffold interior. Overall, this study validates the efficacy of using 3D-printed bioactive scaffolds with autologous SVF to restore geometrically complex midfacial bone defects of clinically relevant sizes while also highlighting remaining challenges to be addressed prior to clinical translation.


Subject(s)
Stromal Vascular Fraction , Tissue Scaffolds , Animals , Bone Regeneration , Osteogenesis , Point-of-Care Systems , Printing, Three-Dimensional , Swine , Swine, Miniature , X-Ray Microtomography
16.
ACS Biomater Sci Eng ; 8(11): 4610-4612, 2022 11 14.
Article in English | MEDLINE | ID: mdl-35157425

ABSTRACT

Tissue engineering has the potential to revolutionize treatments for patients suffering from critical-sized craniofacial bone defects, but it has yet to make a substantial impact in clinical practice. One of the barriers to improving the design of tissue-engineered bone grafts (TEBGs) is the lack of adequate techniques to study how transplanted cells, host cells, and biomaterials interact to facilitate the dynamic healing process. In this perspective, we discuss recent advances in quantitative imaging that may be adapted to provide high spatiotemporal resolution of the 3D tissue microenvironment during cranial bone regeneration. The adoption and application of these imaging technologies will provide a more rigorous framework for evaluating TEBG performance and enable the development of next-generation TEBGs for craniofacial repair.


Subject(s)
Bone Regeneration , Tissue Engineering , Humans , Tissue Engineering/methods , Bone and Bones , Biocompatible Materials
17.
Biomaterials ; 280: 121318, 2022 01.
Article in English | MEDLINE | ID: mdl-34922272

ABSTRACT

Low oxygen (O2) diffusion into large tissue engineered scaffolds hinders the therapeutic efficacy of transplanted cells. To overcome this, we previously studied hollow, hyperbarically-loaded microtanks (µtanks) to serve as O2 reservoirs. To adapt these for bone regeneration, we fabricated biodegradable µtanks from polyvinyl alcohol and poly (lactic-co-glycolic acid) and embedded them to form 3D-printed, porous poly-ε-caprolactone (PCL)-µtank scaffolds. PCL-µtank scaffolds were loaded with pure O2 at 300-500 psi. When placed at atmospheric pressures, the scaffolds released O2 over a period of up to 8 h. We confirmed the inhibitory effects of hypoxia on the osteogenic differentiation of human adipose-derived stem cells (hASCs and we validated that µtank-mediated transient hyperoxia had no toxic impacts on hASCs, possibly due to upregulation of endogenous antioxidant regulator genes. We assessed bone regeneration in vivo by implanting O2-loaded, hASC-seeded, PCL-µtank scaffolds into murine calvarial defects (4 mm diameters × 0.6 mm height) and subcutaneously (4 mm diameter × 8 mm height). In both cases we observed increased deposition of extracellular matrix in the O2 delivery group along with greater osteopontin coverages and higher mineral deposition. This study provides evidence that even short-term O2 delivery from PCL-µtank scaffolds may enhance hASC-mediated bone tissue regeneration.


Subject(s)
Osteogenesis , Tissue Engineering , Animals , Bone Regeneration , Cell Differentiation , Mice , Oxygen/pharmacology , Polyesters/pharmacology , Printing, Three-Dimensional , Tissue Scaffolds
18.
Nat Commun ; 12(1): 6219, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34711819

ABSTRACT

Vascularization is critical for skull development, maintenance, and healing. Yet, there remains a significant knowledge gap in the relationship of blood vessels to cranial skeletal progenitors during these processes. Here, we introduce a quantitative 3D imaging platform to enable the visualization and analysis of high-resolution data sets (>100 GB) throughout the entire murine calvarium. Using this technique, we provide single-cell resolution 3D maps of vessel phenotypes and skeletal progenitors in the frontoparietal cranial bones. Through these high-resolution data sets, we demonstrate that CD31hiEmcnhi vessels are spatially correlated with both Osterix+ and Gli1+ skeletal progenitors during postnatal growth, healing, and stimulated remodeling, and are concentrated at transcortical canals and osteogenic fronts. Interestingly, we find that this relationship is weakened in mice with a conditional knockout of PDGF-BB in TRAP+ osteoclasts, suggesting a potential role for osteoclasts in maintaining the native cranial microvascular environment. Our findings provide a foundational framework for understanding how blood vessels and skeletal progenitors spatially interact in cranial bone, and will enable more targeted studies into the mechanisms of skull disease pathologies and treatments. Additionally, our technique can be readily adapted to study numerous cell types and investigate other elusive phenomena in cranial bone biology.


Subject(s)
Neovascularization, Physiologic , Skull/blood supply , Animals , Becaplermin/genetics , Becaplermin/metabolism , Imaging, Three-Dimensional , Mice , Mice, Inbred C57BL , Microcirculation , Osteoclasts/metabolism , Skull/diagnostic imaging , Skull/metabolism
19.
Int J Radiat Oncol Biol Phys ; 111(2): 528-538, 2021 10 01.
Article in English | MEDLINE | ID: mdl-33989720

ABSTRACT

PURPOSE: Vertebral compression fracture is a common complication of spinal stereotactic body radiation therapy. Development of an in vivo model is crucial to fully understand how focal radiation treatment affects vertebral integrity and biology at various dose fractionation regimens. We present a clinically relevant animal model to analyze the effects of localized, high-dose radiation on vertebral microstructure and mechanical integrity. Using this model, we test the hypothesis that fractionation of radiation dosing can reduce focused radiation therapy's harmful effects on the spine. METHODS AND MATERIALS: The L5 vertebra of New Zealand white rabbits was treated with either a 24-Gy single dose of focused radiation or 3 fractionated 8-Gy doses over 3 consecutive days via the Small Animal Radiation Research Platform. Nonirradiated rabbits were used as controls. Rabbits were euthanized 6 months after irradiation, and their lumbar vertebrae were harvested for radiologic, histologic, and biomechanical testing. RESULTS: Localized single-dose radiation led to decreased vertebral bone volume and trabecular number and a subsequent increase in trabecular spacing and thickness at L5. Hypofractionation of the radiation dose similarly led to reduced trabecular number and increased trabecular spacing and thickness, yet it preserved normalized bone volume. Single-dose irradiated vertebrae displayed lower fracture loads and stiffness compared with those receiving hypofractionated irradiation and with controls. The hypofractionated and control groups exhibited similar fracture load and stiffness. For all vertebral samples, bone volume, trabecular number, and trabecular spacing were correlated with fracture loads and Young's modulus (P < .05). Hypocellularity was observed in the bone marrow of both irradiated groups, but osteogenic features were conserved in only the hypofractionated group. CONCLUSIONS: Single-dose focal irradiation showed greater detrimental effects than hypofractionation on the microarchitectural, cellular, and biomechanical characteristics of irradiated vertebral bodies. Correlation between radiologic measurements and biomechanical properties supported the reliability of this animal model of radiation-induced vertebral compression fracture, a finding that can be applied to future studies of preventative measures.


Subject(s)
Disease Models, Animal , Fractures, Compression/etiology , Lumbar Vertebrae/radiation effects , Radiation Dose Hypofractionation , Radiosurgery/adverse effects , Spinal Fractures/etiology , Animals , Biomechanical Phenomena , Male , Rabbits , Spinal Neoplasms/radiotherapy , Vertebral Body/radiation effects
20.
Front Cell Dev Biol ; 9: 661036, 2021.
Article in English | MEDLINE | ID: mdl-33928087

ABSTRACT

Bioreactor systems are built as controlled environments for biological processes and utilized in the field of tissue engineering to apply mechanical, spatial, and chemical cues to developing tissue grafts. Often the systems are applied to instruct differentiation and maturation of the cells grown inside. Perhaps the most obvious targets for strain and compression-based bioreactors are mechanically active tissues, as it is hypothesized that biomimetic mechanical environments instruct immature cells to form differentiated tissues. One such tissue, skeletal muscle, has been identified as a key candidate for strain application due to the close structure-function relationship of myofibers. Here we detail the multiple uses of a custom-built bioreactor system in combination with electrospun fibrin microfibers for muscle tissue engineering. Outlined below are the methods used in the system to test the mechanical properties of hydrogel-based scaffolds in an aqueous environment, including Young's modulus and poroelasticity. Additionally, we demonstrate the application of tensile strain to sterile cell cultures grown on electrospun scaffolds and perform end-point testing of tissue contractility with the addition of an electrode.

SELECTION OF CITATIONS
SEARCH DETAIL
...