Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 966
Filter
2.
Nat Commun ; 15(1): 3640, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684714

ABSTRACT

Careful consideration of how we approach design is crucial to all areas of biotechnology. However, choosing or developing an effective design methodology is not always easy as biology, unlike most areas of engineering, is able to adapt and evolve. Here, we put forward that design and evolution follow a similar cyclic process and therefore all design methods, including traditional design, directed evolution, and even random trial and error, exist within an evolutionary design spectrum. This contrasts with conventional views that often place these methods at odds and provides a valuable framework for unifying engineering approaches for challenging biological design problems.


Subject(s)
Directed Molecular Evolution , Directed Molecular Evolution/methods , Bioengineering/methods , Biotechnology/methods , Biological Evolution , Synthetic Biology/methods
3.
Acta Biomater ; 179: 121-129, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38494083

ABSTRACT

Reconstruction of the human auricle remains a formidable challenge for plastic surgeons. Autologous costal cartilage grafts and alloplastic implants are technically challenging, and aesthetic and/or tactile outcomes are frequently suboptimal. Using a small animal "bioreactor", we have bioengineered full-scale ears utilizing decellularized cartilage xenograft placed within a 3D-printed external auricular scaffold that mimics the size, shape, and biomechanical properties of the native human auricle. The full-scale polylactic acid ear scaffolds were 3D-printed based upon data acquired from 3D photogrammetry of an adult ear. Ovine costal cartilage was processed either through mincing (1 mm3) or zesting (< 0.5 mm3), and then fully decellularized and sterilized. At explantation, both the minced and zested neoears maintained the size and contour complexities of the scaffold topography with steady tissue ingrowth through 6 months in vivo. A mild inflammatory infiltrate at 3 months was replaced by homogenous fibrovascular tissue ingrowth enveloping individual cartilage pieces at 6 months. All ear constructs were pliable, and the elasticity was confirmed by biomechanical analysis. Longer-term studies of the neoears with faster degrading biomaterials will be warranted for future clinical application. STATEMENT OF SIGNIFICANCE: Accurate reconstruction of the human auricle has always been a formidable challenge to plastic surgeons. In this article, we have bioengineered full-scale ears utilizing decellularized cartilage xenograft placed within a 3D-printed external auricular scaffold that mimic the size, shape, and biomechanical properties of the native human auricle. Longer-term studies of the neoears with faster degrading biomaterials will be warranted for future clinical application.


Subject(s)
Ear Auricle , Heterografts , Printing, Three-Dimensional , Tissue Scaffolds , Tissue Scaffolds/chemistry , Animals , Sheep , Humans , Tissue Engineering/methods , Ear Cartilage/physiology , Bioengineering/methods , Cartilage/physiology
4.
Sci Rep ; 14(1): 7590, 2024 03 31.
Article in English | MEDLINE | ID: mdl-38555385

ABSTRACT

Large volume soft tissue defects greatly impact patient quality of life and function while suitable repair options remain a challenge in reconstructive surgery. Engineered flaps could represent a clinically translatable option that may circumvent issues related to donor site morbidity and tissue availability. Herein, we describe the regeneration of vascularized porcine flaps, specifically of the omentum and tensor fascia lata (TFL) flaps, using a tissue engineering perfusion-decellularization and recellularization approach. Flaps were decellularized using a low concentration sodium dodecyl sulfate (SDS) detergent perfusion to generate an acellular scaffold with retained extracellular matrix (ECM) components while removing underlying cellular and nuclear contents. A perfusion-recellularization strategy allowed for seeding of acellular flaps with a co-culture of human umbilical vein endothelial cell (HUVEC) and mesenchymal stromal cells (MSC) onto the decellularized omentum and TFL flaps. Our recellularization technique demonstrated evidence of intravascular cell attachment, as well as markers of endothelial and mesenchymal phenotype. Altogether, our findings support the potential of using bioengineered porcine flaps as a novel, clinically-translatable strategy for future application in reconstructive surgery.


Subject(s)
Bioengineering , Quality of Life , Humans , Swine , Animals , Bioengineering/methods , Biomedical Engineering , Perfusion , Surgical Flaps , Extracellular Matrix , Tissue Scaffolds , Tissue Engineering/methods
5.
J Biol Chem ; 300(3): 105747, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38354783

ABSTRACT

Glycosyltransferases (GT) catalyze the glycosylation of bioactive natural products, including peptides and proteins, flavonoids, and sterols, and have been extensively used as biocatalysts to generate glycosides. However, the often narrow substrate specificity of wild-type GTs requires engineering strategies to expand it. The GT-B structural family is constituted by GTs that share a highly conserved tertiary structure in which the sugar donor and acceptor substrates bind in dedicated domains. Here, we have used this selective binding feature to design an engineering process to generate chimeric glycosyltransferases that combine auto-assembled domains from two different GT-B enzymes. Our approach enabled the generation of a stable dimer with broader substrate promiscuity than the parent enzymes that were related to relaxed interactions between domains in the dimeric GT-B. Our findings provide a basis for the development of a novel class of heterodimeric GTs with improved substrate promiscuity for applications in biotechnology and natural product synthesis.


Subject(s)
Biocatalysis , Glycosyltransferases , Flavonoids/chemistry , Glycosylation , Glycosyltransferases/chemistry , Glycosyltransferases/genetics , Substrate Specificity , Protein Domains , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Bioengineering/methods
6.
Int J Artif Organs ; 47(3): 129-139, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38253541

ABSTRACT

Liver transplantation is the only definitive treatment for end-stage liver disease and its availability is restricted by organ donor shortages. The development of liver bioengineering provides the probability to create a functional alternative to reduce the gap in organ demand and supply. Decellularized liver scaffolds have been widely applied in bioengineering because they can mimic the native liver microenvironment and retain extracellular matrix (ECM) components. Multiple approaches including chemical, physical and biological methods have been developed for liver decellularization in current studies, but a full set of unified criteria has not yet been established. Each method has its advantages and drawbacks that influence the microstructure and ligand landscape of decellularized liver scaffolds. Optimizing a decellularization method to eliminate cell material while retaining as much of the ECM intact as possible is therefore important for biological scaffold applications. Furthermore, crosslinking strategies can improve the biological performance of scaffolds, including reinforcing biomechanics, delaying degradation in vivo and reducing immune rejection, which can better promote the integration of re-cellularized scaffolds with host tissue and influence the reconstruction process. In this review, we aim to present the different liver decellularization techniques, the crosslinking methods to improve scaffold characteristics with crosslinking and the preparation of soluble ECM.


Subject(s)
Liver Transplantation , Tissue Scaffolds , Tissue Scaffolds/chemistry , Extracellular Matrix/chemistry , Liver , Bioengineering/methods , Tissue Engineering/methods
7.
Arterioscler Thromb Vasc Biol ; 44(3): e66-e81, 2024 03.
Article in English | MEDLINE | ID: mdl-38174560

ABSTRACT

Peripheral artery disease is an atherosclerotic disease associated with limb ischemia that necessitates limb amputation in severe cases. Cell therapies comprised of adult mononuclear or stromal cells have been clinically tested and show moderate benefits. Bioengineering strategies can be applied to modify cell behavior and function in a controllable fashion. Using mechanically tunable or spatially controllable biomaterials, we highlight examples in which biomaterials can increase the survival and function of the transplanted cells to improve their revascularization efficacy in preclinical models. Biomaterials can be used in conjunction with soluble factors or genetic approaches to further modulate the behavior of transplanted cells and the locally implanted tissue environment in vivo. We critically assess the advances in bioengineering strategies such as 3-dimensional bioprinting and immunomodulatory biomaterials that can be applied to the treatment of peripheral artery disease and then discuss the current challenges and future directions in the implementation of bioengineering strategies.


Subject(s)
Bioengineering , Peripheral Arterial Disease , Adult , Humans , Bioengineering/methods , Peripheral Arterial Disease/therapy , Biocompatible Materials , Cell- and Tissue-Based Therapy , Vascular Surgical Procedures , Treatment Outcome
8.
Adv Mater ; 36(18): e2311661, 2024 May.
Article in English | MEDLINE | ID: mdl-38252744

ABSTRACT

Brain infections, frequently accompanied by significant inflammation, necessitate comprehensive therapeutic approaches targeting both infections and associated inflammation. A major impediment to such combined treatment is the blood-brain barrier (BBB), which significantly restricts therapeutic agents from achieving effective concentrations within the central nervous system. Here, a neutrophil-centric dual-responsive delivery system, coined "CellUs," is pioneered. This system is characterized by live neutrophils enveloping liposomes of dexamethasone, ceftriaxone, and oxygen-saturated perfluorocarbon (Lipo@D/C/P). CellUs is meticulously engineered to co-deliver antibiotics, anti-inflammatory agents, and oxygen, embodying a comprehensive strategy against brain infections. CellUs leverages the intrinsic abilities of neutrophils to navigate through BBB, accurately target infection sites, and synchronize the release of Lipo@D/C/P with local inflammatory signals. Notably, the incorporation of ultrasound-responsive perfluorocarbon within Lipo@D/C/P ensures the on-demand release of therapeutic agents at the afflicted regions. CellUs shows considerable promise in treating Staphylococcus aureus infections in mice with meningitis, particularly when combined with ultrasound treatments. It effectively penetrates BBB, significantly eliminates bacteria, reduces inflammation, and delivers oxygen to the affected brain tissue, resulting in a substantial improvement in survival rates. Consequently, CellUs harnesses the natural chemotactic properties of neutrophils and offers an innovative pathway to improve treatment effectiveness while minimizing adverse effects.


Subject(s)
Anti-Bacterial Agents , Blood-Brain Barrier , Neutrophils , Staphylococcus aureus , Animals , Neutrophils/metabolism , Mice , Blood-Brain Barrier/metabolism , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Fluorocarbons/chemistry , Liposomes/chemistry , Dexamethasone/pharmacology , Staphylococcal Infections/drug therapy , Brain/metabolism , Ceftriaxone/therapeutic use , Oxygen/metabolism , Humans , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Bioengineering/methods
9.
Science ; 383(6680): 247, 2024 01 19.
Article in English | MEDLINE | ID: mdl-38236976

ABSTRACT

Efficient method for creating proteins with unusual amino acids opens the door to new medicines and catalysts.


Subject(s)
Amino Acids , Bacteria , Bioengineering , Protein Biosynthesis , Proteins , Amino Acids/genetics , Amino Acids/metabolism , Bacteria/genetics , Bacteria/metabolism , Bioengineering/methods , Proteins/chemistry , Proteins/genetics , Synthetic Biology , Genetic Code
10.
Adv Healthc Mater ; 13(1): e2300984, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37694339

ABSTRACT

Pancreatic cancer is a highly lethal form of digestive malignancy that poses significant health risks to individuals worldwide. Chemotherapy-based comprehensive treatment is the primary therapeutic approach for midlife and late-life patients. Nevertheless, the heterogeneity of the tumor and individual genetic backgrounds result in substantial variations in drug sensitivity among patients, rendering a single treatment regimen unsuitable for all patients. Conventional pancreatic cancer tumor organoid models are capable of emulating the biological traits of pancreatic cancer and are utilized in drug development and screening. However, these tumor organoids can still not mimic the tumor microenvironment (TME) in vivo, and the poor controllability in the preparation process hinders translation from essential drug screening to clinical pharmacological therapy. In recent years, many engineering methods with remarkable results have been used to develop pancreatic cancer organoid models, including bio-hydrogel, co-culture, microfluidic, and gene editing. Here, this work summarizes and analyzes the recent developments in engineering pancreatic tumor organoid models. In addition, the future direction of improving engineered pancreatic cancer organoids is discussed for their application prospects in clinical treatment.


Subject(s)
Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/therapy , Pancreatic Neoplasms/pathology , Coculture Techniques , Bioengineering/methods , Tumor Microenvironment , Organoids/pathology
11.
J Transl Med ; 21(1): 926, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38129833

ABSTRACT

While cartilage tissue engineering has significantly improved the speed and quality of cartilage regeneration, the underlying metabolic mechanisms are complex, making research in this area lengthy and challenging. In the past decade, organoids have evolved rapidly as valuable research tools. Methods to create these advanced human cell models range from simple tissue culture techniques to complex bioengineering approaches. Cartilaginous organoids in part mimic the microphysiology of human cartilage and fill a gap in high-fidelity cartilage disease models to a certain extent. They hold great promise to elucidate the pathogenic mechanism of a diversity of cartilage diseases and prove crucial in the development of new drugs. This review will focus on the research progress of cartilaginous organoids and propose strategies for cartilaginous organoid construction, study directions, and future perspectives.


Subject(s)
Organoids , Tissue Engineering , Humans , Organoids/metabolism , Tissue Engineering/methods , Bioengineering/methods , Cartilage
12.
J Vis Exp ; (199)2023 09 29.
Article in English | MEDLINE | ID: mdl-37843266

ABSTRACT

Lung transplantation is often the only option for patients in the later stages of severe lung disease, but this is limited both due to the supply of suitable donor lungs and both acute and chronic rejection after transplantation. Ascertaining novel bioengineering approaches for the replacement of diseased lungs is imperative for improving patient survival and avoiding complications associated with current transplantation methodologies. An alternative approach involves the use of decellularized whole lungs lacking cellular constituents that are typically the cause of acute and chronic rejection. Since the lung is such a complex organ, it is of interest to examine the extracellular matrix components of specific regions, including the vasculature, airways, and alveolar tissue. The purpose of this approach is to establish simple and reproducible methods by which researchers may dissect and isolate region-specific tissue from fully decellularized lungs. The current protocol has been devised for pig and human lungs, but may be applied to other species as well. For this protocol, four regions of the tissue were specified: airway, vasculature, alveoli, and bulk lung tissue. This procedure allows for the procurement of samples of tissue that more accurately represent the contents of the decellularized lung tissue as opposed to traditional bulk analysis methods.


Subject(s)
Lung Diseases , Tissue Scaffolds , Humans , Animals , Swine , Lung/surgery , Lung/blood supply , Bioengineering/methods , Biomedical Engineering , Tissue Engineering/methods , Extracellular Matrix
13.
Biomaterials ; 298: 122143, 2023 07.
Article in English | MEDLINE | ID: mdl-37146365

ABSTRACT

Aging-associated neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases remain poorly understood and no disease-modifying treatments exist despite decades of investigation. Predominant in vitro (e.g., 2D cell culture, organoids) and in vivo (e.g., mouse) models of these diseases are insufficient mimics of human brain tissue structure and function and of human neurodegenerative pathobiology, and have thus contributed to this collective translational failure. This has been a longstanding challenge in the field, and new strategies are required to address both fundamental and translational needs. Bioengineered tissue culture models constitute a class of promising alternatives, as they can overcome the low cell density, poor nutrient exchange, and long term culturability limitations of existing in vitro models. Further, they can reconstruct the structural, mechanical, and biochemical cues of native brain tissue, providing a better mimic of human brain tissues for in vitro pathobiological investigation and drug development. We discuss bioengineering techniques for the generation of these neurodegenerative tissue models, including biomaterials-, organoid-, and microfluidics-based approaches, and design considerations for their construction. To aid the development of the next generation of functional neurodegenerative disease models, we discuss approaches to incorporate greater cellular diversity and simulate aging processes within bioengineered brain tissues.


Subject(s)
Neurodegenerative Diseases , Animals , Mice , Humans , Biomedical Engineering , Organoids , Cell Culture Techniques/methods , Bioengineering/methods , Disease Models, Animal
14.
J Immunother Cancer ; 11(5)2023 05.
Article in English | MEDLINE | ID: mdl-37192783

ABSTRACT

BACKGROUND: Immunosuppressive tumor microenvironment (ITM) remains an obstacle that jeopardizes clinical immunotherapy. METHODS: To address this concern, we have engineered an exosome inherited from M1-pheototype macrophages, which thereby retain functions and ingredients of the parent M1-phenotype macrophages. The delivered RSL3 that serves as a common ferroptosis inducer can reduce the levels of ferroptosis hallmarkers (eg, glutathione and glutathione peroxidase 4), break the redox homeostasis to magnify oxidative stress accumulation, promote the expression of ferroptosis-related proteins, and induce robust ferroptosis of tumor cells, accompanied with which systematic immune response activation can bbe realized. M1 macrophage-derived exosomes can inherit more functions and genetic substances than nanovesicles since nanovesicles inevitably suffer from substance and function loss caused by extrusion-arised structural damage. RESULTS: Inspired by it, spontaneous homing to tumor and M2-like macrophage polarization into M1-like ones are attained, which not only significantly magnify oxidative stress but also mitigate ITM including M2-like macrophage polarization and regulatory T cell decrease, and regulate death pathways. CONCLUSIONS: All these actions accomplish a synergistic antitumor enhancement against tumor progression, thus paving a general route to mitigate ITM, activate immune responses, and magnify ferroptosis.


Subject(s)
Carbolines , Exosomes , Ferroptosis , Macrophages , Neoplasms , Macrophages/metabolism , Exosomes/chemistry , Exosomes/metabolism , Immunotherapy , Ferroptosis/drug effects , Tumor Microenvironment , Animals , Mice , Carbolines/pharmacology , Bioengineering/methods , Neoplasms/immunology , Neoplasms/therapy , Cell Line, Tumor
15.
Cell ; 186(10): 2062-2077.e17, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37075755

ABSTRACT

Entry of enveloped viruses into cells is mediated by viral fusogenic proteins that drive membrane rearrangements needed for fusion between viral and target membranes. Skeletal muscle development also requires membrane fusion events between progenitor cells to form multinucleated myofibers. Myomaker and Myomerger are muscle-specific cell fusogens but do not structurally or functionally resemble classical viral fusogens. We asked whether the muscle fusogens could functionally substitute for viral fusogens, despite their structural distinctiveness, and fuse viruses to cells. We report that engineering of Myomaker and Myomerger on the membrane of enveloped viruses leads to specific transduction of skeletal muscle. We also demonstrate that locally and systemically injected virions pseudotyped with the muscle fusogens can deliver µDystrophin to skeletal muscle of a mouse model of Duchenne muscular dystrophy and alleviate pathology. Through harnessing the intrinsic properties of myogenic membranes, we establish a platform for delivery of therapeutic material to skeletal muscle.


Subject(s)
Bioengineering , Lentivirus , Membrane Proteins , Muscle, Skeletal , Muscular Dystrophy, Duchenne , Animals , Mice , Cell Fusion , Membrane Fusion , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscle Development , Muscle, Skeletal/metabolism , Muscle, Skeletal/virology , Bioengineering/methods , Muscular Dystrophy, Duchenne/therapy , Disease Models, Animal , Viral Tropism , Lentivirus/genetics
16.
Angew Chem Int Ed Engl ; 62(17): e202218613, 2023 04 17.
Article in English | MEDLINE | ID: mdl-36855015

ABSTRACT

Probes allowing high-contrast discrimination of cancer cells and effective retention are powerful tools for the early diagnosis and treatment of cancer. However, conventional small-molecule probes often show limited performance in both aspects. Herein, we report an ingenious molecular engineering strategy for tuning the cellular uptake and retention of rhodamine dyes. Introduction of polar aminoethyl leads to the increased brightness and reduced cellular uptake of dyes, and this change can be reversed by amino acetylation. Moreover, these modifications allow cancer cells to take up more dyes than normal cells (16-fold) through active transport. Specifically, we further improve the signal contrast (56-fold) between cancer and normal cells by constructing activatable probes and confirm that the released fluorophore can remain in cancer cells with extended time, enabling long-term and specific tumor imaging.


Subject(s)
Neoplasms , Humans , Cell Line, Tumor , Bioengineering/methods , Rhodamines/analysis , Rhodamines/chemistry , Rhodamines/metabolism , Animals , Mice
17.
Nat Protoc ; 18(1): 108-135, 2023 01.
Article in English | MEDLINE | ID: mdl-36261633

ABSTRACT

Tissue engineering is an interdisciplinary field that combines stem cells and matrices to form functional constructs that can be used to repair damaged tissues or regenerate whole organs. Tissue stem cells can be expanded and functionally differentiated to form 'mini-organs' resembling native tissue architecture and function. The choice of the scaffold is also pivotal to successful tissue reconstruction. Scaffolds may be broadly classified into synthetic or biological depending upon the purpose of the engineered organ. Bioengineered intestinal grafts represent a potential source of transplantable tissue for patients with intestinal failure, a condition resulting from extensive anatomical and functional loss of small intestine and therefore digestive and absorptive capacity. Prior strategies in intestinal bioengineering have predominantly used either murine or pluripotent cells and synthetic or decellularized rodent scaffolds, thus limiting their translation. Microscale models of human intestinal epithelium on shaped hydrogels and synthetic scaffolds are more physiological, but their regenerative potential is limited by scale. Here we present a protocol for bioengineering human intestinal grafts using patient-derived materials in a bioreactor culture system. This includes the isolation, expansion and biobanking of patient-derived intestinal organoids and fibroblasts, the generation of decellularized human intestinal scaffolds from native human tissue and providing a system for recellularization to form transplantable grafts. The duration of this protocol is 12 weeks, and it can be completed by scientists with prior experience of organoid culture. The resulting engineered mucosal grafts comprise physiological intestinal epithelium, matrix and surrounding niche, offering a valuable tool for both regenerative medicine and the study of human gastrointestinal diseases.


Subject(s)
Biological Specimen Banks , Tissue Scaffolds , Humans , Mice , Animals , Tissue Engineering/methods , Bioengineering/methods , Organoids , Intestinal Mucosa , Fibroblasts , Extracellular Matrix
18.
Proc Natl Acad Sci U S A ; 119(33): e2201776119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35943987

ABSTRACT

Many natural organisms, such as fungal hyphae and plant roots, grow at their tips, enabling the generation of complex bodies composed of natural materials as well as dexterous movement and exploration. Tip growth presents an exemplary process by which materials synthesis and actuation are coupled, providing a blueprint for how growth could be realized in a synthetic system. Herein, we identify three underlying principles essential to tip-based growth of biological organisms: a fluid pressure driving force, localized polymerization for generating structure, and fluid-mediated transport of constituent materials. In this work, these evolved features inspire a synthetic materials growth process called extrusion by self-lubricated interface photopolymerization (E-SLIP), which can continuously fabricate solid profiled polymer parts with tunable mechanical properties from liquid precursors. To demonstrate the utility of E-SLIP, we create a tip-growing soft robot, outline its fundamental governing principles, and highlight its capabilities for growth at speeds up to 12 cm/min and lengths up to 1.5 m. This growing soft robot is capable of executing a range of tasks, including exploration, burrowing, and traversing tortuous paths, which highlight the potential for synthetic growth as a platform for on-demand manufacturing of infrastructure, exploration, and sensing in a variety of environments.


Subject(s)
Bioengineering , Biomimetics , Polymerization , Robotics , Agaricales/growth & development , Bioengineering/methods , Biomimetics/methods , Movement , Plant Development
19.
Curr Opin Biotechnol ; 77: 102756, 2022 10.
Article in English | MEDLINE | ID: mdl-35930844

ABSTRACT

Recent breakthroughs in biofabrication of bioasemblies, consisting of the engineered structures composed of biological or biosynthetic components into a single construct, have found a wide range of practical applications in medicine and engineering. This review presents an overview of how the bottom-up assembly of living entities could drive advances in medicine, by developing tunable biological models and more precise methods for quantifying biological events. Moreover, we delve into advances beyond biomedical applications, where bioassemblies can be manipulated as functional robots and construction materials. Finally, we address the potential challenges and opportunities in the field of engineering living bioassemblies, toward building new design principles for the next generation of bioengineering applications.


Subject(s)
Bioengineering , Biomedical Engineering , Bioengineering/methods
20.
Science ; 377(6602): 148-150, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35857553

ABSTRACT

Scalable biofabrication of heart helical tissue pattern augments pumping function.


Subject(s)
Bioengineering , Heart, Artificial , Heart , Prosthesis Design , Bioengineering/methods , Humans , Myocardial Contraction
SELECTION OF CITATIONS
SEARCH DETAIL
...