Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.605
1.
BMC Biotechnol ; 24(1): 36, 2024 May 25.
Article En | MEDLINE | ID: mdl-38796454

BACKGROUND: To establish a strategy for stem cell-related tissue regeneration therapy, human gingival mesenchymal stem cells (hGMSCs) were loaded with three-dimensional (3D) bioengineered Matrigel matrix scaffolds in high-cell density microtissues to promote local tissue restoration. METHODS: The biological performance and stemness of hGMSCs under 3D culture conditions were investigated by viability and multidirectional differentiation analyses. A Sprague‒Dawley (SD) rat full-thickness buccal mucosa wound model was established, and hGMSCs/Matrigel were injected into the submucosa of the wound. Autologous stem cell proliferation and wound repair in local tissue were assessed by histomorphometry and immunohistochemical staining. RESULTS: Three-dimensional suspension culture can provide a more natural environment for extensions and contacts between hGMSCs, and the viability and adipogenic differentiation capacity of hGMSCs were significantly enhanced. An animal study showed that hGMSCs/Matrigel significantly accelerated soft tissue repair by promoting autologous stem cell proliferation and enhancing the generation of collagen fibers in local tissue. CONCLUSION: Three-dimensional cell culture with hydrogel scaffolds, such as Matrigel, can effectively improve the biological function and maintain the stemness of stem cells. The therapeutic efficacy of hGMSCs/Matrigel was confirmed, as these cells could effectively stimulate soft tissue repair to promote the healing process by activating the host microenvironment and autologous stem cells.


Collagen , Drug Combinations , Laminin , Mesenchymal Stem Cells , Proteoglycans , Rats, Sprague-Dawley , Tissue Scaffolds , Wound Healing , Animals , Laminin/chemistry , Proteoglycans/chemistry , Collagen/chemistry , Humans , Rats , Mesenchymal Stem Cells/cytology , Tissue Scaffolds/chemistry , Cell Differentiation , Cell Proliferation , Gingiva/cytology , Cell Culture Techniques, Three Dimensional/methods , Cells, Cultured , Tissue Engineering/methods , Male , Mouth Mucosa/cytology
2.
Biofabrication ; 16(3)2024 Jun 07.
Article En | MEDLINE | ID: mdl-38810618

The evaluation of anti-tumor drugs is critical for their development and clinical guidance. Tumor organoid models are gaining increased attention due to their ability to better mimic real tumor tissues, as well as lower time and economic costs, which makes up for the shortcomings of cell lines and xenograft models. However, current tumor organoid cultures based on the Matrigel have limitations in matching with high-throughput engineering methods due to slow gelation and low mechanical strength. Here, we present a novel composite bioink for culturing colorectal cancer organoids that provides an environment close to real tissue growth conditions and exhibits excellent photocrosslinking properties for rapid gel formation. Most importantly, the tumor organoids viability in the composite bioink after printing was as high as 97%, which also kept multicellular polar structures consistent with traditional culture methods in the Matrigel. Using 3D bioprinting with this composite bioink loaded with organoids, we demonstrated the feasibility of this drug evaluation model by validating it with clinically used colorectal cancer treatment drugs. Our results suggested that the composite bioink could effectively cultivate tumor organoids using 3D bioprinting, which had the potential to replace less reliable manual operations in promoting the application of tumor organoids in drug development and clinical guidance.


Bioprinting , Organoids , Printing, Three-Dimensional , Organoids/cytology , Organoids/drug effects , Humans , Colorectal Neoplasms/pathology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Laminin/chemistry , Laminin/pharmacology , Proteoglycans/chemistry , Proteoglycans/pharmacology , Collagen , Drug Combinations
3.
Biomater Adv ; 160: 213847, 2024 Jun.
Article En | MEDLINE | ID: mdl-38657288

Three-dimensional (3D) organoid models have been instrumental in understanding molecular mechanisms responsible for many cellular processes and diseases. However, established organic biomaterial scaffolds used for 3D hydrogel cultures, such as Matrigel, are biochemically complex and display significant batch variability, limiting reproducibility in experiments. Recently, there has been significant progress in the development of synthetic hydrogels for in vitro cell culture that are reproducible, mechanically tuneable, and biocompatible. Self-assembling peptide hydrogels (SAPHs) are synthetic biomaterials that can be engineered to be compatible with 3D cell culture. Here we investigate the ability of PeptiGel® SAPHs to model the mammary epithelial cell (MEC) microenvironment in vitro. The positively charged PeptiGel®Alpha4 supported MEC viability, but did not promote formation of polarised acini. Modifying the stiffness of PeptiGel® Alpha4 stimulated changes in MEC viability and changes in protein expression associated with altered MEC function, but did not fully recapitulate the morphologies of MECs grown in Matrigel. To supply the appropriate biochemical signals for MEC organoids, we supplemented PeptiGels® with laminin. Laminin was found to require negatively charged PeptiGel® Alpha7 for functionality, but was then able to provide appropriate signals for correct MEC polarisation and expression of characteristic proteins. Thus, optimisation of SAPH composition and mechanics allows tuning to support tissue-specific organoids.


Cell Culture Techniques, Three Dimensional , Collagen , Drug Combinations , Epithelial Cells , Hydrogels , Laminin , Peptides , Proteoglycans , Laminin/pharmacology , Laminin/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Proteoglycans/pharmacology , Proteoglycans/chemistry , Collagen/chemistry , Collagen/pharmacology , Peptides/pharmacology , Peptides/chemistry , Epithelial Cells/drug effects , Epithelial Cells/cytology , Humans , Female , Cell Culture Techniques, Three Dimensional/methods , Cell Survival/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Mammary Glands, Human/cytology , Organoids/drug effects , Organoids/cytology , Cell Culture Techniques/methods
4.
ACS Biomater Sci Eng ; 10(5): 3203-3217, 2024 May 13.
Article En | MEDLINE | ID: mdl-38557027

The intricate electrophysiological functions and anatomical structures of spinal cord tissue render the establishment of in vitro models for spinal cord-related diseases highly challenging. Currently, both in vivo and in vitro models for spinal cord-related diseases are still underdeveloped, complicating the exploration and development of effective therapeutic drugs or strategies. Organoids cultured from human induced pluripotent stem cells (hiPSCs) hold promise as suitable in vitro models for spinal cord-related diseases. However, the cultivation of spinal cord organoids predominantly relies on Matrigel, a matrix derived from murine sarcoma tissue. Tissue-specific extracellular matrices are key drivers of complex organ development, thus underscoring the urgent need to research safer and more physiologically relevant organoid culture materials. Herein, we have prepared a rat decellularized brain extracellular matrix hydrogel (DBECMH), which supports the formation of hiPSC-derived spinal cord organoids. Compared with Matrigel, organoids cultured in DBECMH exhibited higher expression levels of markers from multiple compartments of the natural spinal cord, facilitating the development and maturation of spinal cord organoid tissues. Our study suggests that DBECMH holds potential to replace Matrigel as the standard culture medium for human spinal cord organoids, thereby advancing the development of spinal cord organoid culture protocols and their application in in vitro modeling of spinal cord-related diseases.


Brain , Hydrogels , Induced Pluripotent Stem Cells , Organoids , Spinal Cord , Organoids/drug effects , Organoids/cytology , Organoids/metabolism , Humans , Animals , Spinal Cord/cytology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Brain/metabolism , Rats , Decellularized Extracellular Matrix/chemistry , Decellularized Extracellular Matrix/pharmacology , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Laminin/pharmacology , Laminin/chemistry , Proteoglycans/chemistry , Rats, Sprague-Dawley , Drug Combinations , Collagen
5.
ACS Biomater Sci Eng ; 10(5): 3218-3231, 2024 May 13.
Article En | MEDLINE | ID: mdl-38593429

Spinal cord organoids are of significant value in the research of spinal cord-related diseases by simulating disease states, thereby facilitating the development of novel therapies. However, the complexity of spinal cord structure and physiological functions, along with the lack of human-derived inducing components, presents challenges in the in vitro construction of human spinal cord organoids. Here, we introduce a novel human decellularized placenta-derived extracellular matrix hydrogel (DPECMH) and, combined with a new induction protocol, successfully construct human spinal cord organoids. The human placenta-sourced decellularized extracellular matrix (dECM), verified through hematoxylin and eosin staining, DNA quantification, and immunofluorescence staining, retained essential ECM components such as elastin, fibronectin, type I collagen, laminin, and so forth. The temperature-sensitive hydrogel made from human placenta dECM demonstrated good biocompatibility and promoted the differentiation of human induced pluripotent stem cell (hiPSCs)-derived spinal cord organoids into neurons. It displayed enhanced expression of laminar markers in comparison to Matrigel and showed higher expression of laminar markers compared to Matrigel, accelerating the maturation process of spinal cord organoids and demonstrating its potential as an organoid culture substrate. DPECMH has the potential to replace Matrigel as the standard additive for human spinal cord organoids, thus advancing the development of spinal cord organoid culture protocols and their application in the in vitro modeling of spinal cord-related diseases.


Cell Differentiation , Decellularized Extracellular Matrix , Hydrogels , Induced Pluripotent Stem Cells , Organoids , Placenta , Spinal Cord , Humans , Organoids/cytology , Organoids/metabolism , Organoids/drug effects , Female , Placenta/cytology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Pregnancy , Hydrogels/chemistry , Hydrogels/pharmacology , Spinal Cord/cytology , Spinal Cord/metabolism , Cell Differentiation/drug effects , Decellularized Extracellular Matrix/pharmacology , Decellularized Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Laminin/pharmacology , Laminin/chemistry
6.
Chembiochem ; 25(8): e202300835, 2024 Apr 16.
Article En | MEDLINE | ID: mdl-38390634

Self-assembled hydrogels, fabricated through diverse non-covalent interactions, have been extensively studied in regenerative medicines. Inspired from bioactive functional motifs of ECM protein, short peptide sequences have shown remarkable abilities to replicate the intrinsic features of the natural extracellular milieu. In this direction, we have fabricated two short hydrophobic bioactive sequences derived from the laminin protein i. e., IKVAV and YIGSR. Based on the substantial hydrophobicity of these peptides, we selected a co-solvent approach as a suitable gelation technique that included different concentrations of DMSO as an organic phase along with an aqueous solution containing 0.1 % TFA. These hydrophobic laminin-based bioactive peptides with limited solubility in aqueous physiological environment showed significantly enhanced solubility with higher DMSO content in water. The enhanced solubility resulted in extensive intermolecular interactions that led to the formation of hydrogels with a higher-order entangled network along with improved mechanical properties. Interestingly, by simply modulating DMSO content, highly tunable gels were accessed in the same gelator domain that displayed differential physicochemical properties. Further, the cellular studies substantiated the potential of these laminin-derived hydrogels in enhancing cell-matrix interactions, thereby reinforcing their applications in tissue engineering.


Dimethyl Sulfoxide , Hydrogels , Hydrogels/chemistry , Solvents , Peptides/chemistry , Laminin/chemistry
7.
Small ; 20(23): e2308815, 2024 Jun.
Article En | MEDLINE | ID: mdl-38161254

Non-neural extracellular matrix (ECM) has limited application in humanized physiological neural modeling due to insufficient brain-specificity and safety concerns. Although brain-derived ECM contains enriched neural components, certain essential components are partially lost during the decellularization process, necessitating augmentation. Here, it is demonstrated that the laminin-augmented porcine brain-decellularized ECM (P-BdECM) is xenogeneic factor-depleted as well as favorable for the regulation of human neurons, astrocytes, and microglia. P-BdECM composition is comparable to human BdECM regarding brain-specificity through the matrisome and gene ontology-biological process analysis. As augmenting strategy, laminin 111 supplement promotes neural function by synergic effect with laminin 521 in P-BdECM. Annexin A1(ANXA1) and Peroxiredoxin(PRDX) in P-BdECM stabilized microglial and astrocytic behavior under normal while promoting active neuroinflammation in response to neuropathological factors. Further, supplementation of the brain-specific molecule to non-neural matrix also ameliorated glial cell inflammation as in P-BdECM. In conclusion, P-BdECM-augmentation strategy can be used to recapitulate humanized pathophysiological cerebral environments for neurological study.


Brain , Cell Differentiation , Extracellular Matrix , Laminin , Humans , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Laminin/chemistry , Brain/metabolism , Animals , Neurons/metabolism , Neuroinflammatory Diseases/metabolism , Swine , Astrocytes/metabolism , Microglia/metabolism , Inflammation/pathology
8.
Sci Rep ; 13(1): 14556, 2023 09 04.
Article En | MEDLINE | ID: mdl-37666868

Deriving stem cells to regenerate full-thickness human skin is important for treating skin disorders without invasive surgical procedures. Our previous protocol to differentiate human induced pluripotent stem cells (iPSCs) into skin-derived precursor cells (SKPs) as a source of dermal stem cells employs mouse fibroblasts as feeder cells and is therefore unsuitable for clinical use. Herein, we report a feeder-free method for differentiating iPSCs into SKPs by customising culture substrates. We immunohistochemically screened for laminins expressed in dermal papillae (DP) and explored the conditions for inducing the differentiation of iPSCs into SKPs on recombinant laminin E8 (LM-E8) fragments with or without conjugation to domain I of perlecan (PDI), which binds to growth factors through heparan sulphate chains. Several LM-E8 fragments, including those of LM111, 121, 332, 421, 511, and 521, supported iPSC differentiation into SKPs without PDI conjugation. However, the SKP yield was significantly enhanced on PDI-conjugated LM-E8 fragments. SKPs induced on PDI-conjugated LM111-E8 fragments retained the gene expression patterns characteristic of SKPs, as well as the ability to differentiate into adipocytes, osteocytes, and Schwann cells. Thus, PDI-conjugated LM-E8 fragments are promising agents for inducing iPSC differentiation into SKPs in clinical settings.


Cell Differentiation , Heparan Sulfate Proteoglycans , Induced Pluripotent Stem Cells , Intercellular Signaling Peptides and Proteins , Laminin , Peptide Fragments , Protein Domains , Skin , Humans , Adipocytes/cytology , Adipocytes/drug effects , Cell Differentiation/drug effects , Heparan Sulfate Proteoglycans/chemistry , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Intercellular Signaling Peptides and Proteins/metabolism , Laminin/chemistry , Laminin/pharmacology , Osteocytes/cytology , Osteocytes/drug effects , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Schwann Cells/cytology , Schwann Cells/drug effects , Skin/cytology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology
9.
Biophys J ; 122(21): 4194-4206, 2023 11 07.
Article En | MEDLINE | ID: mdl-37766428

Bladder, colon, gastric, prostate, and uterine cancers originate in organs surrounded by laminin-coated smooth muscle. In human prostate cancer, tumors that are organ confined, without extracapsular extension through muscle, have an overall cancer survival rate of up to 97% compared with 32% for metastatic disease. Our previous work modeling extracapsular extension reported the blocking of tumor invasion by mutation of a laminin-binding integrin called α6ß1. Expression of the α6AA mutant resulted in a biophysical switch from cell-ECM (extracellular matrix) to cell-cell adhesion with drug sensitivity properties and an inability to invade muscle. Here we used different admixtures of α6AA and α6WT cells to test the cell heterogeneity requirements for muscle invasion. Time-lapse video microscopy revealed that tumor mixtures self-assembled into invasive networks in vitro, whereas α6AA cells assembled only as cohesive clusters. Invasion of α6AA cells into and through live muscle occurred using a 1:1 mixture of α6AA and α6WT cells. Electric cell-substrate impedance sensing measurements revealed that compared with α6AA cells, invasion-competent α6WT cells were 2.5-fold faster at closing a cell-ECM or cell-cell wound, respectively. Cell-ECM rebuilding kinetics show that an increased response occurred in mixtures since the response was eightfold greater compared with populations containing only one cell type. A synthetic cell adhesion cyclic peptide called MTI-101 completely blocked electric cell-substrate impedance sensing cell-ECM wound recovery that persisted in vitro up to 20 h after the wound. Treatment of tumor-bearing animals with 10 mg/kg MTI-101 weekly resulted in a fourfold decrease of muscle invasion by tumor and a decrease of the depth of invasion into muscle comparable to the α6AA cells. Taken together, these data suggest that mixed biophysical phenotypes of tumor cells within a population can provide functional advantages for tumor invasion into and through muscle that can be potentially inhibited by a synthetic cell adhesion molecule.


Extranodal Extension , Laminin , Male , Animals , Humans , Laminin/chemistry , Laminin/genetics , Laminin/metabolism , Integrin alpha6/genetics , Integrin alpha6/metabolism , Cell Adhesion , Muscles/metabolism , Phenotype
10.
Nat Commun ; 14(1): 317, 2023 01 19.
Article En | MEDLINE | ID: mdl-36658135

Laminin polymerization is the major step in basement membranes assembly. Its failures cause laminin N-terminal domain lamininopathies including Pierson syndrome. We have employed cryo-electron microscopy to determine a 3.7 Å structure of the trimeric laminin polymer node containing α1, ß1 and γ1 subunits. The structure reveals the molecular basis of calcium-dependent formation of laminin lattice, and provides insights into polymerization defects manifesting in human disease.


Nephrotic Syndrome , Pupil Disorders , Humans , Laminin/chemistry , Cryoelectron Microscopy , Polymerization , Basement Membrane/chemistry
11.
Org Biomol Chem ; 20(43): 8489-8500, 2022 Nov 09.
Article En | MEDLINE | ID: mdl-36268609

Matriglycan, a polysaccharide that is a pivotal part of the core M3 O-mannosyl glycan composed of the repeating disaccharide -3Xylα1-3GlcAß1-, interacts with laminin to stabilize muscle tissue. We herein report the synthesis of matriglycan-repeating hexasaccharides equipped with an alkyne linker to form glycoconjugates. The key step in the formation of an α-linked xylosyl glycoside was resolved by solvent-specific separation from an anomeric mixture. Successful glycan elongation was regio- and stereoselectively performed to obtain (-3Xylα1-3GlcAß1)3-O(C2H4O)3CH2CCH and the biotin conjugate. We also investigated interactions between matriglycan hexasaccharides and laminin-G-like domains 4 and 5 of laminin-α2 using saturation transfer difference-NMR. The dissociation constant obtained from bio-layer interferometry was estimated to be 7.5 × 10-8 M. These results indicate that a chemical approach may be applied to the reconstruction of muscle tissue.


Laminin , Polysaccharides , Laminin/chemistry , Laminin/metabolism , Glycosylation
12.
Adv Healthc Mater ; 11(22): e2201646, 2022 11.
Article En | MEDLINE | ID: mdl-36099430

Spatiotemporal control of vascularization and innervation is a desired hallmark in advanced tissue regeneration. For this purpose, we design a 3D model scaffold, based on elastin-like recombinamer (ELR) hydrogels. This contains two interior and well-defined areas, small cylinders, with differentiated bioactivities with respect to the bulk. Both are constructed on a protease sensitive ELR with a fast-proteolyzed domain, but one bears a VEGF-mimetic peptide (QK) and the other a laminin-derived pentapeptide (IKVAV), to promote angiogenesis and neurogenesis, respectively. The outer bulk is based on a slow proteolytic sequence and RGD cell adhesion domains. In vitro studies show the effect of QK and IKVAV peptides on the promotion of endothelial cell and axon spreading, respectively. The subcutaneous implantation of the final 3D scaffold demonstrates the ability to spatiotemporally control angiogenesis and neurogenesis in vivo. Specifically, the inner small cylinder containing the QK peptide promotes fast endothelialization, whereas the one with IKVAV peptide promotes fast neurogenesis. Both, vascularization and innervation take place in advance of the bulk scaffold infiltration. This scaffold shows that it is possible to induce vascularization and innervation in predetermined areas of the scaffold well ahead to the bulk infiltration. That significantly increases the efficiency of the regenerative activity.


Elastin , Laminin , Elastin/pharmacology , Elastin/chemistry , Laminin/pharmacology , Laminin/chemistry , Vascular Endothelial Growth Factor A/pharmacology , Peptide Hydrolases , Peptides/pharmacology , Peptides/chemistry , Hydrogels/pharmacology , Hydrogels/chemistry , Neurogenesis
13.
Molecules ; 27(13)2022 Jun 27.
Article En | MEDLINE | ID: mdl-35807362

Synthetic bioactive aromatic peptide amphiphiles have been recognized as key elements of emerging biomedical strategies due to their biocompatibility, design flexibility, and functionality. Inspired by natural proteins, we synthesized two supramolecular materials of phenyl-capped Ile-Lys-Val-Ala-Val (Ben-IKVAV) and perfluorophenyl-capped Ile-Lys-Val-Ala-Val (PFB-IKVAV). We employed UV-vis absorption, fluorescence, circular dichroism, and Fourier-transform infrared spectroscopy to examine the driving force in the self-assembly of the newly discovered materials. It was found that both compounds exhibited ordered π-π interactions and secondary structures, especially PFB-IKVAV. The cytotoxicity of human mesenchymal stem cells (hMSCs) and cell differentiation studies was also performed. In addition, the immunofluorescent staining for neuronal-specific markers of MAP2 was 4.6 times (neural induction medium in the presence of PFB-IKVAV) that of the neural induction medium (control) on day 7. From analyzing the expression of neuronal-specific markers in hMSCs, it can be concluded that PFB-IKVAV may be a potential supramolecular biomaterial for biomedical applications.


Laminin , Peptide Fragments , Humans , Hydrogels/chemistry , Laminin/chemistry , Peptide Fragments/chemistry , Peptides/chemistry , Peptides/pharmacology
14.
J Mater Sci Mater Med ; 33(6): 50, 2022 May 31.
Article En | MEDLINE | ID: mdl-35639181

Multichannel structures in the design of nerve conduits offer potential advantages for regeneration of damaged nerves. However, lack of biochemical cues and electrical stimulation could hamper satisfactory nerve regeneration. The aim of this study was to simultaneously evaluate the effects of topographical, biological, and electrical cues on sciatic nerve regeneration. Accordingly, a series of multichannel nerve conduit was made using longitudinally-aligned laminin-coated poly (lactic-co-glycolic acid) (PLGA)/carbon nanotubes (CNT) nanofibers (NF, mean diameter: 455 ± 362 nm) in the lumen and randomly-oriented polycaprolactone (PCL) NF (mean diameter: 340 ± 200 nm) on the outer surface. In vitro studies revealed that the materials were nontoxic and able to promote cell attachment and proliferation on nanofibers and on fibrin gel. To determine the influence of laminin as biological and CNT as electrical cues on nerve regeneration, either of hollow PCL conduits, PLGA NF-embedded, PLGA/CNT NF-embedded or laminin-coated PLGA/CNT NF-embedded PCL conduits were implanted in rats. A new surgery method was utilized and results were compared with an autograft. The results of motor and sensory tests in addition to histopathological examination of the regenerated nerves demonstrated the formation of nerve fibers in laminin-coated PLGA/CNT NF-embedded PCL conduits. Results suggested that these conduits have the potential to improve sciatic nerve regeneration. Graphical abstract.


Nanofibers , Nanotubes, Carbon , Animals , Laminin/chemistry , Nanofibers/chemistry , Nanotubes, Carbon/chemistry , Nerve Regeneration , Rats , Sciatic Nerve/physiology
15.
Nat Protoc ; 16(12): 5652-5672, 2021 12.
Article En | MEDLINE | ID: mdl-34759383

Heart-forming organoids (HFOs) derived from human pluripotent stem cells (hPSCs) are a complex, highly structured in vitro model of early heart, foregut and vasculature development. The model represents a potent tool for various applications, including teratogenicity studies, gene function analysis and drug discovery. Here, we provide a detailed protocol describing how to form HFOs within 14 d. In an initial 4 d preculture period, hPSC aggregates are individually formed in a 96-well format and then Matrigel-embedded. Subsequently, the chemical WNT pathway modulators CHIR99021 and IWP2 are applied, inducing directed differentiation. This highly robust protocol can be used on many different hPSC lines and be combined with manipulation technologies such as gene targeting and drug testing. HFO formation can be assessed by numerous complementary methods, ranging from various imaging approaches to gene expression studies. Here, we highlight the flow cytometry-based analysis of individual HFOs, enabling the quantitative monitoring of lineage formation.


Flow Cytometry/methods , Organogenesis/genetics , Organoids/cytology , Pluripotent Stem Cells/cytology , Tissue Scaffolds , Wnt Signaling Pathway/drug effects , Benzothiazoles/pharmacology , Cell Differentiation/drug effects , Collagen/chemistry , Collagen/pharmacology , Drug Combinations , Drug Discovery/methods , Gene Targeting/methods , Heart/diagnostic imaging , Heart/drug effects , Humans , Laminin/chemistry , Laminin/pharmacology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Organogenesis/drug effects , Organoids/diagnostic imaging , Organoids/drug effects , Organoids/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Proteoglycans/chemistry , Proteoglycans/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Teratogens/toxicity
16.
Science ; 374(6569): 848-856, 2021 Nov 12.
Article En | MEDLINE | ID: mdl-34762454

The signaling of cells by scaffolds of synthetic molecules that mimic proteins is known to be effective in the regeneration of tissues. Here, we describe peptide amphiphile supramolecular polymers containing two distinct signals and test them in a mouse model of severe spinal cord injury. One signal activates the transmembrane receptor ß1-integrin and a second one activates the basic fibroblast growth factor 2 receptor. By mutating the peptide sequence of the amphiphilic monomers in nonbioactive domains, we intensified the motions of molecules within scaffold fibrils. This resulted in notable differences in vascular growth, axonal regeneration, myelination, survival of motor neurons, reduced gliosis, and functional recovery. We hypothesize that the signaling of cells by ensembles of molecules could be optimized by tuning their internal motions.


Nanofibers , Peptides , Spinal Cord Injuries/therapy , Spinal Cord Regeneration , Tissue Scaffolds , Animals , Cell Survival , Computer Simulation , Human Umbilical Vein Endothelial Cells/physiology , Humans , Integrin beta1/metabolism , Laminin/chemistry , Laminin/metabolism , Mice , Motor Neurons/physiology , Neovascularization, Physiologic , Neural Stem Cells/physiology , Peptides/chemistry , Peptidomimetics/chemistry , Polymers/chemistry , Protein Conformation, beta-Strand , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Recovery of Function , Signal Transduction , Surface-Active Agents
17.
Nat Commun ; 12(1): 6967, 2021 11 29.
Article En | MEDLINE | ID: mdl-34845227

Breast cancer is now globally the most frequent cancer and leading cause of women's death. Two thirds of breast cancers express the luminal estrogen receptor-positive (ERα + ) phenotype that is initially responsive to antihormonal therapies, but drug resistance emerges. A major barrier to the understanding of the ERα-pathway biology and therapeutic discoveries is the restricted repertoire of luminal ERα + breast cancer models. The ERα + phenotype is not stable in cultured cells for reasons not fully understood. We examine 400 patient-derived breast epithelial and breast cancer explant cultures (PDECs) grown in various three-dimensional matrix scaffolds, finding that ERα is primarily regulated by the matrix stiffness. Matrix stiffness upregulates the ERα signaling via stress-mediated p38 activation and H3K27me3-mediated epigenetic regulation. The finding that the matrix stiffness is a central cue to the ERα phenotype reveals a mechanobiological component in breast tissue hormonal signaling and enables the development of novel therapeutic interventions. Subject terms: ER-positive (ER + ), breast cancer, ex vivo model, preclinical model, PDEC, stiffness, p38 SAPK.


Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Mechanotransduction, Cellular/genetics , Transcriptome , p38 Mitogen-Activated Protein Kinases/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Case-Control Studies , Cell Line, Tumor , Cinnamates/pharmacology , Collagen/chemistry , Collagen/pharmacology , Drug Combinations , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Female , Fulvestrant/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Histones/genetics , Histones/metabolism , Humans , Indazoles/pharmacology , Laminin/chemistry , Laminin/pharmacology , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Phenotype , Proteoglycans/chemistry , Proteoglycans/pharmacology , Tamoxifen/pharmacology , Tissue Culture Techniques , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Sci Rep ; 11(1): 21547, 2021 11 03.
Article En | MEDLINE | ID: mdl-34732783

Bioprinting is increasingly used to create complex tissue constructs for an array of research applications, and there are also increasing efforts to print tissues for transplantation. Bioprinting may also prove valuable in the context of drug screening for personalized medicine for treatment of diseases such as cancer. However, the rapidly expanding bioprinting research field is currently limited by access to bioprinters. To increase the availability of bioprinting technologies we present here an open source extrusion bioprinter based on the E3D motion system and tool changer to enable high-resolution multimaterial bioprinting. As proof of concept, the bioprinter is used to create collagen constructs using freeform reversible embedding of suspended hydrogels (FRESH) methodology, as well as multimaterial constructs composed of distinct sections of laminin and collagen. Data is presented demonstrating that the bioprinted constructs support growth of cells either seeded onto printed constructs or included in the bioink prior to bioprinting. This open source bioprinter is easily adapted for different bioprinting applications, and additional tools can be incorporated to increase the capabilities of the system.


Bioprinting/instrumentation , Bioprinting/methods , Neoplasms/physiopathology , Printing, Three-Dimensional , Tissue Engineering/methods , Breast Neoplasms/physiopathology , Calibration , Cell Line, Tumor , Cell Survival , Collagen/chemistry , Equipment Design , Humans , Hydrogels/chemistry , Laminin/chemistry , Temperature , Tissue Scaffolds
19.
Commun Biol ; 4(1): 1183, 2021 10 14.
Article En | MEDLINE | ID: mdl-34650188

A key to enhance the low translatability of preclinical drug discovery are in vitro human three-dimensional (3D) microphysiological systems (MPS). Here, we show a new method for automated engineering of 3D human skeletal muscle models in microplates and functional compound screening to address the lack of muscle wasting disease medication. To this end, we adapted our recently described 24-well plate 3D bioprinting platform with a printhead cooling system to allow microvalve-based drop-on-demand printing of cell-laden Matrigel containing primary human muscle precursor cells. Mini skeletal muscle models develop within a week exhibiting contractile, striated myofibers aligned between two attachment posts. As an in vitro exercise model, repeated high impact stimulation of contractions for 3 h by a custom-made electrical pulse stimulation (EPS) system for 24-well plates induced interleukin-6 myokine expression and Akt hypertrophy pathway activation. Furthermore, the known muscle stimulators caffeine and Tirasemtiv acutely increase EPS-induced contractile force of the models. This validated new human muscle MPS will benefit development of drugs against muscle wasting diseases. Moreover, our Matrigel 3D bioprinting platform will allow engineering of non-self-organizing complex human 3D MPS.


Bioprinting/methods , Caffeine/pharmacology , Collagen/chemistry , Exercise/physiology , Imidazoles/pharmacology , Laminin/chemistry , Muscle Contraction/physiology , Muscle, Skeletal/physiology , Proteoglycans/chemistry , Pyrazines/pharmacology , Drug Combinations , Electric Stimulation , Humans , Printing, Three-Dimensional
20.
Biochem Biophys Res Commun ; 579: 8-14, 2021 11 19.
Article En | MEDLINE | ID: mdl-34583196

α-Dystroglycan (α-DG) is a glycoprotein specifically modified with O-mannosyl glycans bearing long polysaccharides, termed matriglycans, which comprise repeating units of glucuronic acid and xylose. The matriglycan is linked to the O-mannosyl glycan core through two ribitol phosphate units that can be replaced with glycerol phosphate (GroP) units synthesized by fukutin and fukutin-related protein that transfer GroP from CDP-Gro. Here, we found that forced expression of the bacterial CDP-Gro synthase, TagD, from Bacillus subtilis could result in the overproduction of CDP-Gro in human colon carcinoma HCT116 cells. Western blot and liquid chromatography-tandem mass spectrometry analyses indicated that α-DG prepared from the TagD-expressing HCT116 cells contained abundant GroP and lacked matriglycans. Using the GroP-containing recombinant α-DG-Fc, we developed a novel monoclonal antibody, termed DG2, that reacts with several truncated glycoforms of α-DG, including GroP-terminated glycoforms lacking matriglycans; we verified the reactivity of DG2 against various types of knockout cells deficient in the biosynthesis of matriglycans. Accordingly, forced expression of TagD in HCT116 cells resulted in the reduction of matriglycans and an increase in DG2 reactivity. Collectively, our results indicate that DG2 could serve as a useful tool to determine tissue distribution and function of α-DG lacking matriglycans under physiological and pathophysiological conditions.


Antibodies, Monoclonal/chemistry , Dystroglycans/chemistry , Laminin/chemistry , Protein Isoforms/chemistry , Animals , Bacillus subtilis , CRISPR-Cas Systems , Chromatography, Liquid , DNA, Complementary/metabolism , Female , Glucuronic Acid/chemistry , Glycopeptides/chemistry , HCT116 Cells , Humans , Mass Spectrometry , Mice , Mice, Inbred BALB C , Phosphates , Polysaccharides , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Ribitol/chemistry , Xylose
...