711 research outputs found

    Bioinspired materials and tissue engineering approaches applied to the regeneration of musculoskeletal tissues

    Get PDF
    The musculoskeletal tissues have a prime role in the biomechanical support and metabolic activities of the human body. As musculoskeletal tissues are highly prone to injuries, conditions afflicting these tissues have a great impact on the quality of life of patients worldwide. Tissue engineering approaches hold the promise to develop bioengineered substitutes aiming at the regeneration of failing and injured tissue and organs. To effectively address the tissue-specific structural and biochemical features of musculoskeletal tissues, different biomaterials and techniques have been employed envisioning biomimetic solutions. Herein, the unique composition, structure, and function of the musculoskeletal tissues, namely bone, cartilage, and tendon, as well as state-of-the-art technologies to develop bioinspired strategies for tissue regeneration will be overviewed. Finally, this chapter will also discuss the unmet challenges and future perspectives in the field.FCT Project MagTT PTDC/CTM-CTM/29930/2017 (POCI-01- 0145-FEDER-29930) for A.I.G postdoc grant, the FCT Project PTDC/NAN-MAT/30595/2017 (POCI-01-0145-FEDER-30595) for P.S.B. postdoc grant, and for the assistant researcher contract (RL1) of M.T.R from the project “Accelerating tissue engineering and personalized medicine discoveries by the integration of key enabling nanotechnologies, marine-derived biomaterials and stem cells” supported by Norte Portugal Regional Operational Programme (NORTE 2020), under the PORTUGAL 2020 Partnership Agreement, through the European Regional Development Fund (ERDF). Authors acknowledge the financial support from the European Union Framework Programme for Research and Innovation HORIZON 2020, under the TEAMING Grant agreement No. 739572—The Discoveries CTR and the European Research Council 2017-CoG MagTendon (No. 772817

    High Throughput And Mechano-Active Platforms To Promote Cartilage Regeneration And Repair

    Get PDF
    Traumatic joint injuries initiate acute degenerative changes in articular cartilage that can lead to progressive loss of load-bearing function. As a result, patients often develop post-traumatic osteoarthritis (PTOA), a condition for which there currently exists no biologic interventions. To address this need, tissue engineering aims to mimic the structure and function of healthy, native counterparts. These constructs can be used to not only replace degenerated tissue, but also build in vitro, pre-clinical models of disease. Towards this latter goal, this thesis focuses on the design of a high throughput system to screen new therapeutics in a micro-engineered model of PTOA, and the development of a mechanically-responsive drug delivery system to augment tissue-engineered approaches for cartilage repair. High throughput screening is a powerful tool for drug discovery that can be adapted to include 3D tissue constructs. To facilitate this process for cartilage repair, we built a high throughput mechanical injury platform to create an engineered cartilage model of PTOA. Compressive injury of functionally mature constructs increased cell death and proteoglycan loss, two hallmarks of injury observed in vivo. Comparison of this response to that of native cartilage explants, and evaluation of putative therapeutics, validated this model for subsequent use in small molecule screens. A primary screen of 118 compounds identified a number of ‘hits’ and relevant pathways that may modulate pathologic signaling post-injury. To complement this process of therapeutic discovery, a stimuli-responsive delivery system was designed that used mechanical inputs as the ‘trigger’ mechanism for controlled release. The failure thresholds of these mechanically-activated microcapsules (MAMCs) were influenced by physical properties and composition, as well as matrix mechanical properties in 3D environments. TGF-beta released from the system upon mechano-activation stimulated stem cell chondrogenesis, demonstrating the potential of MAMCs to actively deliver therapeutics within demanding mechanical environments. Taken together, this work advances our capacity to identify and deliver new compounds of clinical relevance to modulate disease progression following traumatic injury using state-of-the-art micro-engineered screening tools and a novel mechanically-activated delivery system. These platforms advance strategies for cartilage repair and regeneration in PTOA and provide new options for the treatment of this debilitating condition

    Tissue Engineering of Tendons

    Get PDF
    Critical size tendon defects demand for tissue samples replacing the missing tissue and guiding an effective healing. Autografts, allografts, or xenografts represent viable options; however, limited availability and donor site morbidity go along with this approach, representing big disadvantages. Tissue engineering of tendon tissue is a further strategy fulfilling this need. Basically, an appropriate scaffold material is developed and tested for its biomechanical suitability as a graft material. In addition, cell seeding might improve biointegration of the tissue engineered construct (TEC). Different cell sources as well as different cultivation procedures can be applied in order to tune the envisioned primary strength of the TEC. In this chapter, in vitro fabrication protocols and mechanical tests as well as animal in vivo experiments will be presented—covering various (bio)materials, cell types, and cultivation procedures

    Scaffolding strategies for tissue engineering and regenerative medicine applications

    Get PDF
    During the past two decades, tissue engineering and the regenerative medicine field have invested in the regeneration and reconstruction of pathologically altered tissues, such as cartilage, bone, skin, heart valves, nerves and tendons, and many others. The 3D structured scaffolds and hydrogels alone or combined with bioactive molecules or genes and cells are able to guide the development of functional engineered tissues, and provide mechanical support during in vivo implantation. Naturally derived and synthetic polymers, bioresorbable inorganic materials, and respective hybrids, and decellularized tissue have been considered as scaffolding biomaterials, owing to their boosted structural, mechanical, and biological properties. A diversity of biomaterials, current treatment strategies, and emergent technologies used for 3D scaffolds and hydrogel processing, and the tissue-specific considerations for scaffolding for Tissue engineering (TE) purposes are herein highlighted and discussed in depth. The newest procedures focusing on the 3D behavior and multi-cellular interactions of native tissues for further use for in vitro model processing are also outlined. Completed and ongoing preclinical research trials for TE applications using scaffolds and hydrogels, challenges, and future prospects of research in the regenerative medicine field are also presented.This research was funded by Norte Portugal Regional Operational Programme (NORTE 2020), under the PORTUGAL 2020 Partnership Agreement, through the European Regional Development Fund (ERDF) (NORTE-01-0145-FEDER-000023) and by the Portuguese Foundation for Science and Technology ((M-ERA-NET/0022/2016), Transitional Rule DL 57/2016 (CTTI-57/18-I3BS(5)), and (IF/01285/2015))

    Engineering Cell–ECM–Material Interactions for Musculoskeletal Regeneration

    Get PDF
    The extracellular microenvironment regulates many of the mechanical and biochemical cues that direct musculoskeletal development and are involved in musculoskeletal disease. The extracellular matrix (ECM) is a main component of this microenvironment. Tissue engineered approaches towards regenerating muscle, cartilage, tendon, and bone target the ECM because it supplies critical signals for regenerating musculoskeletal tissues. Engineered ECM–material scaffolds that mimic key mechanical and biochemical components of the ECM are of particular interest in musculoskeletal tissue engineering. Such materials are biocompatible, can be fabricated to have desirable mechanical and biochemical properties, and can be further chemically or genetically modified to support cell differentiation or halt degenerative disease progression. In this review, we survey how engineered approaches using natural and ECM-derived materials and scaffold systems can harness the unique characteristics of the ECM to support musculoskeletal tissue regeneration, with a focus on skeletal muscle, cartilage, tendon, and bone. We summarize the strengths of current approaches and look towards a future of materials and culture systems with engineered and highly tailored cell–ECM–material interactions to drive musculoskeletal tissue restoration. The works highlighted in this review strongly support the continued exploration of ECM and other engineered materials as tools to control cell fate and make large-scale musculoskeletal regeneration a reality

    Biofabrication Strategies for Musculoskeletal Disorders: Evolution towards Clinical Applications

    Get PDF
    Biofabrication has emerged as an attractive strategy to personalise medical care and provide new treatments for common organ damage or diseases. While it has made impactful headway in e.g., skin grafting, drug testing and cancer research purposes, its application to treat musculoskeletal tissue disorders in a clinical setting remains scarce. Albeit with several in vitro breakthroughs over the past decade, standard musculoskeletal treatments are still limited to palliative care or surgical interventions with limited long-term effects and biological functionality. To better understand this lack of translation, it is important to study connections between basic science challenges and developments with translational hurdles and evolving frameworks for this fully disruptive technology that is biofabrication. This review paper thus looks closely at the processing stage of biofabrication, specifically at the bioinks suitable for musculoskeletal tissue fabrication and their trends of usage. This includes underlying composite bioink strategies to address the shortfalls of sole biomaterials. We also review recent advances made to overcome long-standing challenges in the field of biofabrication, namely bioprinting of low-viscosity bioinks, controlled delivery of growth factors, and the fabrication of spatially graded biological and structural scaffolds to help biofabricate more clinically relevant constructs. We further explore the clinical application of biofabricated musculoskeletal structures, regulatory pathways, and challenges for clinical translation, while identifying the opportunities that currently lie closest to clinical translation. In this article, we consider the next era of biofabrication and the overarching challenges that need to be addressed to reach clinical relevance

    Advanced Gene Therapy Strategies for the Repair of ACL Injuries

    Get PDF
    The anterior cruciate ligament (ACL), the principal ligament for stabilization of the knee, is highly predisposed to injury in the human population. As a result of its poor intrinsic healing capacities, surgical intervention is generally necessary to repair ACL lesions, yet the outcomes are never fully satisfactory in terms of long-lasting, complete, and safe repair. Gene therapy, based on the transfer of therapeutic genetic sequences via a gene vector, is a potent tool to durably and adeptly enhance the processes of ACL repair and has been reported for its workability in various experimental models relevant to ACL injuries in vitro, in situ, and in vivo. As critical hurdles to the effective and safe translation of gene therapy for clinical applications still remain, including physiological barriers and host immune responses, biomaterial-guided gene therapy inspired by drug delivery systems has been further developed to protect and improve the classical procedures of gene transfer in the future treatment of ACL injuries in patients, as critically presented here

    State-of-Art Functional Biomaterials for Tissue Engineering

    Get PDF
    Nanobiotechnology-enabled tissue engineering strategies have emerged as an innovative and promising technique in the field of regenerative medical science. The design and development of multifunctional smart biomaterials compatible to human physiology is crucial to achieve the required biological function with a reduced negative biological response. Several medical bioimplants have been tested to boost life expectancy and better-quality life. The concept of biocompatibility focuses on body acceptance and no harmful effects after implantation, which require shaping the properties of materials synthesis, surface functionalization, and biofunctionality. Such developed bioactive and biodegradable materials have been utilized to achieve the required function at a specific period and sustainability to withstand the surrounding tissues for treating severe injuries and diseases. Thus, exploring new approaches to design multifunctional biocompatible advanced nanostructures to develop next-generation therapies for tissue engineering, this mini-review is an attempt to summarize the advancements in biofunctional smart materials. The review focuses on bio-mimic materials, biomaterials, self-assembly biomaterials, bioprinting functional hydrogels, new polymeric architectures, and hybrid synthetic–natural hydrogels in the field of tissue engineering and regenerative medicine (TERM). This mini-review will serve as a guideline to design future research where the selection of a smart multifunctional biomaterial is crucial to obtain best TERM performance

    American Society for Bone and Mineral Research-Orthopaedic Research Society Joint Task Force Report on Cell-Based Therapies.

    Get PDF
    Cell-based therapies, defined here as the delivery of cells in vivo to treat disease, have recently gained increasing public attention as a potentially promising approach to restore structure and function to musculoskeletal tissues. Although cell-based therapy has the potential to improve the treatment of disorders of the musculoskeletal system, there is also the possibility of misuse and misrepresentation of the efficacy of such treatments. The medical literature contains anecdotal reports and research studies, along with web-based marketing and patient testimonials supporting cell-based therapy. Both the American Society for Bone and Mineral Research (ASBMR) and the Orthopaedic Research Society (ORS) are committed to ensuring that the potential of cell-based therapies is realized through rigorous, reproducible, and clinically meaningful scientific discovery. The two organizations convened a multidisciplinary and international Task Force composed of physicians, surgeons, and scientists who are recognized experts in the development and use of cell-based therapies. The Task Force was charged with defining the state-of-the art in cell-based therapies and identifying the gaps in knowledge and methodologies that should guide the research agenda. The efforts of this Task Force are designed to provide researchers and clinicians with a better understanding of the current state of the science and research needed to advance the study and use of cell-based therapies for skeletal tissues. The design and implementation of rigorous, thorough protocols will be critical to leveraging these innovative treatments and optimizing clinical and functional patient outcomes. In addition to providing specific recommendations and ethical considerations for preclinical and clinical investigations, this report concludes with an outline to address knowledge gaps in how to determine the cell autonomous and nonautonomous effects of a donor population used for bone regeneration. © 2019 American Society for Bone and Mineral Research
    • 

    corecore