Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 16.221
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 3946, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38729950

RESUMO

Disease modeling with isogenic Induced Pluripotent Stem Cell (iPSC)-differentiated organoids serves as a powerful technique for studying disease mechanisms. Multiplexed coculture is crucial to mitigate batch effects when studying the genetic effects of disease-causing variants in differentiated iPSCs or organoids, and demultiplexing at the single-cell level can be conveniently achieved by assessing natural genetic barcodes. Here, to enable cost-efficient time-series experimental designs via multiplexed bulk and single-cell RNA-seq of hybrids, we introduce a computational method in our Vireo Suite, Vireo-bulk, to effectively deconvolve pooled bulk RNA-seq data by genotype reference, and thereby quantify donor abundance over the course of differentiation and identify differentially expressed genes among donors. Furthermore, with multiplexed scRNA-seq and bulk RNA-seq, we demonstrate the usefulness and necessity of a pooled design to reveal donor iPSC line heterogeneity during macrophage cell differentiation and to model rare WT1 mutation-driven kidney disease with chimeric organoids. Our work provides an experimental and analytic pipeline for dissecting disease mechanisms with chimeric organoids.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas , Organoides , RNA-Seq , Análise de Célula Única , Organoides/metabolismo , Análise de Célula Única/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Humanos , Diferenciação Celular/genética , RNA-Seq/métodos , Análise de Sequência de RNA/métodos , Macrófagos/metabolismo , Macrófagos/citologia , Animais , Análise da Expressão Gênica de Célula Única
2.
Methods Mol Biol ; 2800: 1-10, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38709473

RESUMO

The fruit fly Drosophila is a well-established invertebrate model that enables in vivo imaging of innate immune cell (e.g., macrophage) migration and signaling at high spatiotemporal resolution within the intact, living animal. While optimized methods already exist to enable flow cytometry-based macrophage isolation from Drosophila at various stages of development, there remains a need for more rapid and gentle methods to isolate living macrophages for downstream ex vivo applications. Here, we describe techniques for rapid and direct isolation of living macrophages from mature Drosophila pupae and their downstream ex vivo preparation for live imaging and immunostaining. This strategy enables straightforward access to physiologically relevant innate immune cells, both circulating and tissue-resident populations, for subsequent imaging of signal transduction.


Assuntos
Macrófagos , Pupa , Animais , Pupa/citologia , Macrófagos/citologia , Macrófagos/metabolismo , Drosophila , Separação Celular/métodos , Citometria de Fluxo/métodos , Drosophila melanogaster/citologia
3.
Stem Cell Res Ther ; 15(1): 132, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38702808

RESUMO

BACKGROUND: Induced pluripotent stem cells (iPSCs)-derived kidney organoids are a promising model for studying disease mechanisms and renal development. Despite several protocols having been developed, further improvements are needed to overcome existing limitations and enable a wider application of this model. One of the approaches to improve the differentiation of renal organoids in vitro is to include in the system cell types important for kidney organogenesis in vivo, such as macrophages. Another approach could be to improve cell survival. Mesodermal lineage differentiation is the common initial step of the reported protocols. The glycogen synthase kinase-3 (GSK-3) activity inhibitor, CHIR99021 (CHIR), is applied to induce mesodermal differentiation. It has been reported that CHIR simultaneously induces iPSCs apoptosis that can compromise cell differentiation. We thought to interfere with CHIR-induced apoptosis of iPSCs using rapamycin. METHODS: Differentiation of kidney organoids from human iPSCs was performed. Cell survival and autophagy were analyzed using Cell counting kit 8 (CCK8) kit and Autophagy detection kit. Cells were treated with rapamycin or co-cultured with human monocytes isolated from peripheral blood or iPSCs-macrophages using a transwell co-culture system. Monocyte-derived extracellular vesicles (EVs) were isolated using polyethylene glycol precipitation. Expression of apoptotic markers cleaved Caspase 3, Poly [ADP-ribose] polymerase 1 (PARP-1) and markers of differentiation T-Box Transcription Factor 6 (TBX6), odd-skipped related 1 (OSR1), Nephrin, E-Cadherin, Paired box gene 2 (Pax2) and GATA Binding Protein 3 (Gata3) was assessed by RT-PCR and western blotting. Organoids were imaged by 3D-confocal microscopy. RESULTS: We observed that CHIR induced apoptosis of iPSCs during the initial stage of renal organoid differentiation. Underlying mechanisms implied the accumulation of reactive oxygen species and decreased autophagy. Activation of autophagy by rapamacin and by an indirect co-culture of differentiating iPSCs with iPSCs-macrophages and human peripheral blood monocytes prevented apoptosis induced by CHIR. Furthermore, monocytes (but not rapamycin) strongly promoted expression of renal differentiation markers and organoids development via released extracellular vesicles. CONCLUSION: Our data suggest that co-culturing of iPSCs with human monocytes strongly improves differentiation of kidney organoids. An underlying mechanism of monocytic action implies, but not limited to, an increased autophagy in CHIR-treated iPSCs. Our findings enhance the utility of kidney organoid models.


Assuntos
Apoptose , Diferenciação Celular , Células-Tronco Pluripotentes Induzidas , Rim , Monócitos , Organoides , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Organoides/citologia , Organoides/metabolismo , Organoides/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Rim/citologia , Rim/metabolismo , Monócitos/metabolismo , Monócitos/citologia , Monócitos/efeitos dos fármacos , Piridinas/farmacologia , Pirimidinas/farmacologia , Sirolimo/farmacologia , Autofagia/efeitos dos fármacos , Técnicas de Cocultura/métodos , Macrófagos/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos
4.
Nanoscale ; 16(16): 7874-7883, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38563323

RESUMO

Anisotropic gold (Au) nanostructures have been widely explored for various nanomedicine applications. While these nanomaterials have shown great promise for disease theranostics, particularly for cancer diagnosis and treatment, the utilization and clinical translation of anisotropic Au nanostructures have been limited by their high phagocytic uptake and clearance and low cancer targeting specificity. Numerous efforts have thus been made toward mitigating these challenges. Many conventional strategies, however, rely on all-synthetic materials, involve complex chemical processes, or have low product throughput and reproducibility. Herein, by integrating cell membrane coating and microfluidic technologies, a high-throughput bioinspired approach for synthesizing biomimetic anisotropic Au nanostructures with minimized phagocytic uptake and improved cancer cell targeting is reported. Through continuous hydrodynamic flow focusing, mixing, and sonication, Au nanostructures are encapsulated within the macrophage and cancer cell membrane vesicles effectively. The fabricated nanostructures are uniform and highly stable in serum. Importantly, the macrophage membrane vesicle-encapsulated Au nanostructures can be preferentially internalized by breast cancer cells, but not by macrophages. Overall, this study has demonstrated the feasibility of employing an integrated microfluidic-sonication technique to formulate uniform and highly stable biomimetic anisotropic nanostructures for enhanced cancer theranostic applications.


Assuntos
Membrana Celular , Ouro , Ouro/química , Humanos , Anisotropia , Membrana Celular/metabolismo , Membrana Celular/química , Animais , Camundongos , Macrófagos/metabolismo , Macrófagos/citologia , Nanopartículas Metálicas/química , Células RAW 264.7 , Linhagem Celular Tumoral , Nanoestruturas/química , Células MCF-7
5.
Anal Chem ; 96(18): 6922-6929, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38653330

RESUMO

We report the development and validation of an untargeted single-cell lipidomics method based on microflow chromatography coupled to a data-dependent mass spectrometry method for fragmentation-based identification of lipids. Given the absence of single-cell lipid standards, we show how the methodology should be optimized and validated using a dilute cell extract. The methodology is applied to dilute pancreatic cancer and macrophage cell extracts and standards to demonstrate the sensitivity requirements for confident assignment of lipids and classification of the cell type at the single-cell level. The method is then coupled to a system that can provide automated sampling of live, single cells into capillaries under microscope observation. This workflow retains the spatial information and morphology of cells during sampling and highlights the heterogeneity in lipid profiles observed at the single-cell level. The workflow is applied to show changes in single-cell lipid profiles as a response to oxidative stress, coinciding with expanded lipid droplets. This demonstrates that the workflow is sufficiently sensitive to observing changes in lipid profiles in response to a biological stimulus. Understanding how lipids vary in single cells will inform future research into a multitude of biological processes as lipids play important roles in structural, biophysical, energy storage, and signaling functions.


Assuntos
Lipidômica , Lipídeos , Análise de Célula Única , Lipidômica/métodos , Humanos , Lipídeos/análise , Lipídeos/química , Animais , Cromatografia Líquida , Camundongos , Linhagem Celular Tumoral , Espectrometria de Massas , Macrófagos/metabolismo , Macrófagos/citologia
6.
Nature ; 629(8010): 184-192, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38600378

RESUMO

Glucocorticoids represent the mainstay of therapy for a broad spectrum of immune-mediated inflammatory diseases. However, the molecular mechanisms underlying their anti-inflammatory mode of action have remained incompletely understood1. Here we show that the anti-inflammatory properties of glucocorticoids involve reprogramming of the mitochondrial metabolism of macrophages, resulting in increased and sustained production of the anti-inflammatory metabolite itaconate and consequent inhibition of the inflammatory response. The glucocorticoid receptor interacts with parts of the pyruvate dehydrogenase complex whereby glucocorticoids provoke an increase in activity and enable an accelerated and paradoxical flux of the tricarboxylic acid (TCA) cycle in otherwise pro-inflammatory macrophages. This glucocorticoid-mediated rewiring of mitochondrial metabolism potentiates TCA-cycle-dependent production of itaconate throughout the inflammatory response, thereby interfering with the production of pro-inflammatory cytokines. By contrast, artificial blocking of the TCA cycle or genetic deficiency in aconitate decarboxylase 1, the rate-limiting enzyme of itaconate synthesis, interferes with the anti-inflammatory effects of glucocorticoids and, accordingly, abrogates their beneficial effects during a diverse range of preclinical models of immune-mediated inflammatory diseases. Our findings provide important insights into the anti-inflammatory properties of glucocorticoids and have substantial implications for the design of new classes of anti-inflammatory drugs.


Assuntos
Anti-Inflamatórios , Glucocorticoides , Inflamação , Macrófagos , Mitocôndrias , Succinatos , Animais , Feminino , Humanos , Masculino , Camundongos , Anti-Inflamatórios/farmacologia , Carboxiliases/metabolismo , Carboxiliases/antagonistas & inibidores , Ciclo do Ácido Cítrico/efeitos dos fármacos , Ciclo do Ácido Cítrico/genética , Citocinas/imunologia , Citocinas/metabolismo , Glucocorticoides/farmacologia , Glucocorticoides/metabolismo , Hidroliases/deficiência , Hidroliases/genética , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Complexo Piruvato Desidrogenase/metabolismo , Receptores de Glucocorticoides/metabolismo , Succinatos/metabolismo , Ativação Enzimática/efeitos dos fármacos
7.
Biofabrication ; 16(3)2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38663394

RESUMO

Extracellular matrix (ECM) rich whole organ bio-scaffolds, preserving structural integrity and essential growth factors, has potential towards regeneration and reconstruction. Women with cervical anomalies or trauma can benefit from clinical cervicovaginal repair using constructs rich in site specific ECM. In this study, complete human cervix decellularization was achieved using a modified perfusion-based stir bench top decellularization method. This was followed by physico-chemical processes including perfusion of ionic agents, enzymatic treatment and washing using detergent solutions for a duration of 10-12 d. Histopathological analysis, as well as DNA quantification confirmed the efficacy of the decellularization process. Tissue ultrastructure integrity was preserved and the same was validated via scanning electron microscopy and transmission electron microscopy studies. Biochemical analysis and structural characterizations like Fourier transform infrared, Raman spectroscopy of decellularized tissues demonstrated preservation of important proteins, crucial growth factors, collagen, and glycosaminoglycans.In vitrostudies, using THP-1 and human umbilical vein endothelial cell (HUVEC) cells, demonstrated macrophage polarization from M1 to M2 and vascular functional genes enhancement, respectively, when treated with decellularized human cervical matrix (DHCp). Crosslinked DHC scaffolds were recellularized with site specific human cervical epithelial cells and HUVEC, showing non-cytotoxic cell viability and enhanced proliferation. Furthermore, DHC scaffolds showed immunomodulatory effectsin vivoon small rodent model via upregulation of M2 macrophage genes as compared to decellularized rat cervix matrix scaffolds (DRC). DHC scaffolds underwent neo-vascularization followed by ECM remodeling with enhanced tissue integration.


Assuntos
Colo do Útero , Matriz Extracelular Descelularizada , Células Endoteliais da Veia Umbilical Humana , Alicerces Teciduais , Humanos , Feminino , Colo do Útero/citologia , Animais , Matriz Extracelular Descelularizada/química , Matriz Extracelular Descelularizada/farmacologia , Alicerces Teciduais/química , Matriz Extracelular/metabolismo , Matriz Extracelular/química , Ratos , Engenharia Tecidual , Células THP-1 , Macrófagos/metabolismo , Macrófagos/citologia , Ratos Sprague-Dawley
8.
Sci Signal ; 17(834): eadq0353, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38687824

RESUMO

Displacement of the glycocalyx by membrane blebbing enables macrophages to recognize apoptotic cells.


Assuntos
Apoptose , Glicocálix , Macrófagos , Humanos , Glicocálix/metabolismo , Animais , Macrófagos/metabolismo , Macrófagos/citologia , Fagócitos/metabolismo , Fagócitos/citologia , Fagocitose , Camundongos
9.
Tissue Eng Regen Med ; 21(4): 587-594, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38451425

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) have been highlighted as a potent therapeutic option for conditions with excessive osteoclast activity such as systemic and local bone loss in rheumatic disease. In addition to their immunomodulatory functions, MSCs also directly suppress osteoclast differentiation and activation by secreting osteoprotegerin (OPG) and IL-10 but the underlying mechanisms are still to be clarified. Tumor necrosis factor-stimulated gene-6 (TSG-6) is a potent anti-inflammatory molecule that inhibits osteoclast activation and has been shown to mediate MSC's immunomodulatory functions. In this study, we aimed to determine whether adipose tissue-derived MSC (ADMSC) inhibits the differentiation from osteoclast precursors to mature osteoclasts through TSG-6. METHODS: Human ADMSCs were co-cultured with bone marrow-derived monocyte/macrophage (BMMs) from DBA/1J or B6 mouse in the presence of osteoclastogenic condition (M-CSF 10 ng/mL and RANKL 10 ng/mL). In some co-culture groups, ADMSCs were transfected with siRNA targeting TSG-6 or OPG to determine their role in osteoclastogenesis. Tartrate-resistant acid phosphatase (TRAP) activity in culture supernatant and mRNA expression of osteoclast markers were investigated. TRAP+ multinucleated cells and F-actin ring formation were counted. RESULTS: ADMSCs significantly inhibited osteoclast differentiation under osteoclastogenic conditions. Suppression of TSG-6 significantly reversed the inhibition of osteoclast differentiation in a degree similar to that of OPG based on TRAP activity, mRNA expression of osteoclast markers, and numbers of TRAP+ multinucleated cell and F-actin ring formation. CONCLUSION: This study demonstrated that ADMSCs inhibit osteoclast differentiation through TSG-6 under osteoclastogenic conditions.


Assuntos
Tecido Adiposo , Moléculas de Adesão Celular , Diferenciação Celular , Células-Tronco Mesenquimais , Osteoclastos , Osteoclastos/metabolismo , Osteoclastos/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Diferenciação Celular/efeitos dos fármacos , Humanos , Animais , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Camundongos , Moléculas de Adesão Celular/metabolismo , Osteoprotegerina/metabolismo , Técnicas de Cocultura , Camundongos Endogâmicos C57BL , Osteogênese/efeitos dos fármacos , Fosfatase Ácida Resistente a Tartarato/metabolismo , Macrófagos/metabolismo , Macrófagos/citologia
10.
Stem Cell Rev Rep ; 20(4): 1121-1134, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38478316

RESUMO

Macrophages are key regulators in bone repair and regeneration. Recent studies have shown that long-term epigenetic changes and metabolic shifts occur during specific immune training of macrophages that affect their functional state, resulting in heightened (trained) or reduced (tolerant) responses upon exposure to a second stimulus. This is known as innate immune memory. Here, we study the impact of macrophages' memory trait on osteoblast differentiation of human mesenchymal stromal cells (hMSCs) and osteoclast differentiation. An in vitro trained immunity protocol of monocyte-derived macrophages was employed using inactivated Candida albicans and Bacillus Calmette-Guérin (BCG) to induce a 'trained' state and Pam3CSK4 (PAM) and Lipopolysaccharides (LPS) to induce a 'tolerance' state. Macrophages were subsequently cocultured with hMSCs undergoing osteogenic differentiation during either resting (unstimulated) or inflammatory conditions (restimulated with LPS). Alkaline phosphatase activity, mineralization, and cytokine levels (TNF, IL-6, oncostatin M and SDF-1α) were measured. In addition, macrophages underwent osteoclast differentiation. Our findings show that trained and tolerized macrophages induced opposing results. Under resting conditions, BCG-trained macrophages enhanced ALP levels (threefold), while under inflammatory conditions this was found in the LPS-tolerized macrophages (fourfold). Coculture of hMSCs with trained macrophages showed mineralization while tolerized macrophages inhibited the process under both resting and inflammatory conditions. While osteoclast differentiation was not affected in trained-macrophages, this ability was significantly loss in tolerized ones. This study further confirms the intricate cross talk between immune cells and bone cells, highlighting the need to consider this interaction in the development of personalized approaches for bone regenerative medicine.


Assuntos
Diferenciação Celular , Técnicas de Cocultura , Imunidade Inata , Lipopolissacarídeos , Macrófagos , Células-Tronco Mesenquimais , Osteoblastos , Osteoclastos , Humanos , Osteoclastos/metabolismo , Osteoclastos/citologia , Osteoblastos/metabolismo , Osteoblastos/citologia , Macrófagos/metabolismo , Macrófagos/imunologia , Macrófagos/citologia , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Lipopolissacarídeos/farmacologia , Osteogênese , Células Cultivadas , Citocinas/metabolismo , Candida albicans/imunologia , Lipopeptídeos/farmacologia , Imunidade Treinada
11.
Biophys J ; 123(9): 1069-1084, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38532625

RESUMO

Macrophage phagocytosis is critical for the immune response, homeostasis regulation, and tissue repair. This intricate process involves complex changes in cell morphology, cytoskeletal reorganization, and various receptor-ligand interactions controlled by mechanical constraints. However, there is a lack of comprehensive theoretical and computational models that investigate the mechanical process of phagocytosis in the context of cytoskeletal rearrangement. To address this issue, we propose a novel coarse-grained mesoscopic model that integrates a fluid-like cell membrane and a cytoskeletal network to study the dynamic phagocytosis process. The growth of actin filaments results in the formation of long and thin pseudopods, and the initial cytoskeleton can be disassembled upon target entry and reconstructed after phagocytosis. Through dynamic changes in the cytoskeleton, our macrophage model achieves active phagocytosis by forming a phagocytic cup utilizing pseudopods in two distinct ways. We have developed a new algorithm for modifying membrane area to prevent membrane rupture and ensure sufficient surface area during phagocytosis. In addition, the bending modulus, shear stiffness, and cortical tension of the macrophage model are investigated through computation of the axial force for the tubular structure and micropipette aspiration. With this model, we simulate active phagocytosis at the cytoskeletal level and investigate the mechanical process during the dynamic interplay between macrophage and target particles.


Assuntos
Macrófagos , Modelos Biológicos , Fagocitose , Pseudópodes , Macrófagos/citologia , Macrófagos/metabolismo , Pseudópodes/metabolismo , Membrana Celular/metabolismo , Fenômenos Biomecânicos , Citoesqueleto/metabolismo
12.
Int Immunopharmacol ; 130: 111766, 2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38452411

RESUMO

OBJECTIVES: This study aimed to investigate the effect of calcitonin gene-related peptide (CGRP) on the temporal alteration of macrophage phenotypes and macrophage-regulated angiogenesis duringearlybonehealing and preliminarily elucidate the mechanism. METHODS: In vivo, the rat mandibular defect models were established with inferior alveolar nerve transection (IANT) or CGRP receptor antagonist injection. Radiographicandhistologic assessments for osteogenesis, angiogenesis, and macrophage phenotypic alteration within bone defects were performed. In vitro, the effect and mechanism of CGRP on macrophage polarization and phenotypic alteration were analyzed. Then the conditioned medium (CM) from CGRP-treated M1 or M2 macrophages was used to culture human umbilical vein endothelial cells (HUVECs), and the CGRP's effect on macrophage-regulated angiogenesis was detected. RESULTS: Comparable changes following IANT and CGRP blockade within bone defects were observed, including the suppression of early osteogenesis and angiogenesis, the prolonged M1 macrophage infiltration and the prohibited transition toward M2 macrophages around vascular endothelium. In vitro experiments showed that CGRP promoted M2 macrophage polarization while upregulating the expression of interleukin 6 (IL-6), a major cytokine that facilitates the transition from M1 to M2-dominant stage, in M1 macrophages via the activation of Yes-associated protein 1. Moreover, CGRP-treated macrophage-CM showed an anabolic effect on HUVECs angiogenesis compared with macrophage-CM and might prevail over the direct effect of CGRP on HUVECs. CONCLUSIONS: Collectively, our results reveal the effect of CGRP on M1 to M2 macrophage phenotypic alteration possibly via upregulating IL-6 in M1 macrophages, and demonstrate the macrophage-regulated pro-angiogenic potential of CGRP in early bone healing.


Assuntos
Regeneração Óssea , Osso e Ossos , Peptídeo Relacionado com Gene de Calcitonina , Interleucina-6 , Macrófagos , Neovascularização Fisiológica , Animais , Humanos , Ratos , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Interleucina-6/metabolismo , Macrófagos/citologia , Macrófagos/fisiologia , Fenótipo , Ratos Sprague-Dawley , Feminino , Osso e Ossos/irrigação sanguínea
13.
J Dent ; 144: 104957, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38527517

RESUMO

INTRODUCTION: Osteoclasts (OCs) play a crucial role in maintaining bone health. Changes in OC activity are linked to different bone diseases, making them an intriguing focus for research. However, most studies on OCs have relied on 2D cultures, limiting our understanding of their behavior. Yet, there's a lack of knowledge regarding platforms that effectively support osteoclast formation in 3D cultures. METHODS: In our investigation, we explored the capacity of collagen and GelMA hydrogels to facilitate osteoclast development in 3D culture settings. We assessed the osteoclast development by using different hydrogels and cell seeding strategies and optimizing cell seeding density and cytokine concentration. The osteoclast development in 3D cultures was further validated by biochemical assays and immunochemical staining. RESULTS: Our findings revealed that 0.3 % (w/v) collagen was conducive to osteoclast formation in both 2D and 3D cultures, demonstrated by increased multinucleation and higher TRAP activity compared to 0.6 % collagen and 5 % to 10 % (w/v) GelMA hydrogels. Additionally, we devised a "sandwich" technique using collagen substrates and augmented the initial macrophage seeding density and doubling cytokine concentrations, significantly enhancing the efficiency of OC culture in 3D conditions. Notably, we validated osteoclasts derived from macrophages in our 3D cultures express key osteoclast markers like cathepsin K and TRAP. CONCLUSIONS: To conclude, our study contributes to establishing an effective method for cultivating osteoclasts in 3D environments in vitro. This innovative approach not only promises a more physiologically relevant platform to study osteoclast behavior during bone remodeling but also holds potential for applications in bone tissue engineering. CLINICAL SIGNIFICANCE: This study introduces an efficient method for cultivating osteoclasts in 3D environments in vitro. It offers a more physiologically relevant platform to investigate osteoclast behavior and holds promise to advance research in bone biology and regenerative dentistry.


Assuntos
Técnicas de Cultura de Células , Hidrogéis , Osteoclastos , Osteoclastos/citologia , Animais , Diferenciação Celular , Colágeno , Camundongos , Técnicas de Cultura de Células em Três Dimensões/métodos , Macrófagos/citologia , Catepsina K , Citocinas/metabolismo , Células Cultivadas
14.
ACS Appl Mater Interfaces ; 16(17): 21415-21426, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38445580

RESUMO

Effective tissue regeneration and immune responses are essential for the success of biomaterial implantation. Although the interaction between synthetic materials and biological systems is well-recognized, the role of surface topographical cues in regulating the local osteoimmune microenvironment─specifically, their impact on host tissue and immune cells, and their dynamic interactions─remains underexplored. This study addresses this gap by investigating the impact of surface topography on osteogenesis and immunomodulation. We fabricated MXene/hydroxyapatite (HAP)-coated surfaces with controlled 2.5D nano-, submicro-, and microscale topographical patterns using our custom bottom-up patterning method. These engineered surfaces were employed to assess the behavior of osteoblast precursor cells and macrophage polarization. Our results demonstrate that MXene/HAP-coated surfaces with microscale crumpled topography significantly influence osteogenic activity and macrophage polarization: these surfaces notably enhanced osteoblast precursor cell spreading, proliferation, and differentiation and facilitated a shift in macrophages toward an anti-inflammatory, prohealing M2 phenotype. The observed cell responses indicate that the physical cues from the crumpled topographies, combined with the chemical cues from the MXene/HAP coatings, synergistically create a favorable osteoimmune microenvironment. This study presents the first evidence of employing MXene/HAP-multilayer coated surfaces with finely crumpled topography to concurrently facilitate osteogenesis and immunomodulation for improved implant-to-tissue integration. The tunable topographic patterns of these coatings coupled with a facile and scalable fabrication process make them widely applicable for various biomedical purposes. Our results highlight the potential of these multilayer coatings with controlled topography to improve the in vivo performance and fate of implants by modulating the host response at the material interface.


Assuntos
Materiais Revestidos Biocompatíveis , Macrófagos , Osteogênese , Osteogênese/efeitos dos fármacos , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Animais , Durapatita/química , Durapatita/farmacologia , Diferenciação Celular/efeitos dos fármacos , Propriedades de Superfície , Células RAW 264.7 , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Nanopartículas/química , Proliferação de Células/efeitos dos fármacos , Humanos
15.
Nature ; 628(8008): 604-611, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38538784

RESUMO

The immune system has a critical role in orchestrating tissue healing. As a result, regenerative strategies that control immune components have proved effective1,2. This is particularly relevant when immune dysregulation that results from conditions such as diabetes or advanced age impairs tissue healing following injury2,3. Nociceptive sensory neurons have a crucial role as immunoregulators and exert both protective and harmful effects depending on the context4-12. However, how neuro-immune interactions affect tissue repair and regeneration following acute injury is unclear. Here we show that ablation of the NaV1.8 nociceptor impairs skin wound repair and muscle regeneration after acute tissue injury. Nociceptor endings grow into injured skin and muscle tissues and signal to immune cells through the neuropeptide calcitonin gene-related peptide (CGRP) during the healing process. CGRP acts via receptor activity-modifying protein 1 (RAMP1) on neutrophils, monocytes and macrophages to inhibit recruitment, accelerate death, enhance efferocytosis and polarize macrophages towards a pro-repair phenotype. The effects of CGRP on neutrophils and macrophages are mediated via thrombospondin-1 release and its subsequent autocrine and/or paracrine effects. In mice without nociceptors and diabetic mice with peripheral neuropathies, delivery of an engineered version of CGRP accelerated wound healing and promoted muscle regeneration. Harnessing neuro-immune interactions has potential to treat non-healing tissues in which dysregulated neuro-immune interactions impair tissue healing.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina , Macrófagos , Neutrófilos , Nociceptores , Cicatrização , Animais , Camundongos , Comunicação Autócrina , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Eferocitose , Macrófagos/citologia , Macrófagos/metabolismo , Monócitos/citologia , Monócitos/metabolismo , Músculo Esquelético , Canal de Sódio Disparado por Voltagem NAV1.8/deficiência , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Canal de Sódio Disparado por Voltagem NAV1.8/metabolismo , Neutrófilos/citologia , Neutrófilos/metabolismo , Nociceptores/metabolismo , Comunicação Parácrina , Doenças do Sistema Nervoso Periférico/complicações , Proteína 1 Modificadora da Atividade de Receptores/metabolismo , Regeneração/efeitos dos fármacos , Pele , Trombospondina 1/metabolismo , Cicatrização/efeitos dos fármacos , Cicatrização/imunologia , Humanos , Masculino , Feminino
16.
Sci Data ; 11(1): 252, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38418823

RESUMO

RNA modifications have emerged as central regulators of gene expression programs. Amongst RNA modifications are N6-methyladenosine (m6A) and RNA 5-hydroxymethylcytosine (5hmC). While m6A is established as a versatile regulator of RNA metabolism, the functions of RNA 5hmC are unclear. Despite some evidence linking RNA modifications to immunity, their implications in gene expression control in macrophage development and functions remain unclear. Here we present a multi-omics dataset capturing different layers of the gene expression programs driving macrophage differentiation and polarisation. We obtained mRNA-Seq, m6A-IP-Seq, 5hmC-IP-Seq, Polyribo-Seq and LC-MS/MS data from monocytes and resting-, pro- and anti-inflammatory-like macrophages. We present technical validation showing high quality and correlation between samples for all datasets, and evidence of biological consistency of modelled macrophages at the transcriptomic, epitranscriptomic, translational and proteomic levels. This multi-omics dataset provides a resource for the study of RNA m6A and 5hmC in the context of macrophage biology and spans the gene expression process from transcripts to proteins.


Assuntos
Macrófagos , Multiômica , RNA , Humanos , Cromatografia Líquida , Macrófagos/citologia , RNA/metabolismo , Espectrometria de Massas em Tandem , Diferenciação Celular , Polaridade Celular
17.
J Virol ; 98(3): e0156323, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38323811

RESUMO

Macrophages are important target cells for diverse viruses and thus represent a valuable system for studying virus biology. Isolation of primary human macrophages is done by culture of dissociated tissues or from differentiated blood monocytes, but these methods are both time consuming and result in low numbers of recovered macrophages. Here, we explore whether macrophages derived from human induced pluripotent stem cells (iPSCs)-which proliferate indefinitely and potentially provide unlimited starting material-could serve as a faithful model system for studying virus biology. Human iPSC-derived monocytes were differentiated into macrophages and then infected with HIV-1, dengue virus, or influenza virus as model human viruses. We show that iPSC-derived macrophages support the replication of these viruses with kinetics and phenotypes similar to human blood monocyte-derived macrophages. These iPSC-derived macrophages were virtually indistinguishable from human blood monocyte-derived macrophages based on surface marker expression (flow cytometry), transcriptomics (RNA sequencing), and chromatin accessibility profiling. iPSC lines were additionally generated from non-human primate (chimpanzee) fibroblasts. When challenged with dengue virus, human and chimpanzee iPSC-derived macrophages show differential susceptibility to infection, thus providing a valuable resource for studying the species-tropism of viruses. We also show that blood- and iPSC-derived macrophages both restrict influenza virus at a late stage of the virus lifecycle. Collectively, our results substantiate iPSC-derived macrophages as an alternative to blood monocyte-derived macrophages for the study of virus biology. IMPORTANCE: Macrophages have complex relationships with viruses: while macrophages aid in the removal of pathogenic viruses from the body, macrophages are also manipulated by some viruses to serve as vessels for viral replication, dissemination, and long-term persistence. Here, we show that iPSC-derived macrophages are an excellent model that can be exploited in virology.


Assuntos
Vírus da Dengue , HIV-1 , Células-Tronco Pluripotentes Induzidas , Macrófagos , Modelos Biológicos , Orthomyxoviridae , Virologia , Animais , Humanos , Diferenciação Celular/genética , HIV-1/crescimento & desenvolvimento , HIV-1/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Macrófagos/citologia , Macrófagos/metabolismo , Macrófagos/virologia , Orthomyxoviridae/crescimento & desenvolvimento , Orthomyxoviridae/fisiologia , Pan troglodytes , Vírus da Dengue/crescimento & desenvolvimento , Vírus da Dengue/fisiologia , Fibroblastos/citologia , Monócitos/citologia , Replicação Viral , Citometria de Fluxo , Perfilação da Expressão Gênica , Montagem e Desmontagem da Cromatina , Tropismo Viral , Virologia/métodos , Biomarcadores/análise , Biomarcadores/metabolismo
18.
Adv Mater ; 36(19): e2310876, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38321645

RESUMO

Structural and physiological cues provide guidance for the directional migration and spatial organization of endogenous cells. Here, a microchannel scaffold with instructive niches is developed using a circumferential freeze-casting technique with an alkaline salting-out strategy. Thereinto, polydopamine-coated nano-hydroxyapatite is employed as a functional inorganic linker to participate in the entanglement and crystallization of chitosan molecules. This scaffold orchestrates the advantage of an oriented porous structure for rapid cell infiltration and satisfactory immunomodulatory capacity to promote stem cell recruitment, retention, and subsequent osteogenic differentiation. Transcriptomic analysis as well as its in vitro and in vivo verification demonstrates that essential colony-stimulating factor-1 (CSF-1) factor is induced by this scaffold, and effectively bound to the target colony-stimulating factor-1 receptor (CSF-1R) on the macrophage surface to activate the M2 phenotype, achieving substantial endogenous bone regeneration. This strategy provides a simple and efficient approach for engineering inducible bone regenerative biomaterials.


Assuntos
Regeneração Óssea , Durapatita , Fator Estimulador de Colônias de Macrófagos , Osteogênese , Polímeros , Receptor de Fator Estimulador de Colônias de Macrófagos , Alicerces Teciduais , Regeneração Óssea/efeitos dos fármacos , Alicerces Teciduais/química , Animais , Camundongos , Durapatita/química , Fator Estimulador de Colônias de Macrófagos/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/química , Polímeros/química , Diferenciação Celular , Quitosana/química , Indóis/química , Transdução de Sinais , Engenharia Tecidual/métodos , Macrófagos/metabolismo , Macrófagos/citologia , Células RAW 264.7
19.
Adv Mater ; 36(18): e2312740, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38272455

RESUMO

The epithelium, an essential barrier to protect organisms against infection, exists in many organs. However, rapid re-epithelialization to restore tissue integrity and function in an adverse environment is challenging. In this work, a long-term anti-inflammatory and antioxidant hydrogel with mechanical stimulation for rapid re-epithelialization, mainly composed of the small molecule thioctic acid, biocompatible glycine, and γ-Fe2O3 nanoparticles is reported. Glycine-modified supramolecular thioctic acid is stable and possesses outstanding mechanical properties. The incorporating γ-Fe2O3 providing the potential contrast function for magnetic resonance imaging observation, can propel hydrogel reconfiguration to enhance the mechanical properties of the hydrogel underwater due to water-initiated release of Fe3+. In vitro experiments show that the hydrogels effectively reduced intracellular reactive oxygen species, guided macrophages toward M2 polarization, and alleviated inflammation. The effect of rapid re-epithelialization is ultimately demonstrated in a long urethral injury model in vivo, and the mechanical stimulation of hydrogels achieves effective functional replacement and ultimately accurate remodeling of the epithelium. Notably, the proposed strategy provides an advanced alternative treatment for patients in need of large-area epithelial reconstruction.


Assuntos
Anti-Inflamatórios , Antioxidantes , Hidrogéis , Hidrogéis/química , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Antioxidantes/química , Antioxidantes/farmacologia , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Reepitelização/efeitos dos fármacos , Células RAW 264.7 , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/citologia , Glicina/química , Glicina/farmacologia , Humanos , Compostos Férricos/química
20.
J Biol Chem ; 300(1): 105561, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38097183

RESUMO

Chronic inflammation is the underlying cause of many diseases, including type 1 diabetes, obesity, and non-alcoholic fatty liver disease. Macrophages are continuously recruited to tissues during chronic inflammation where they exacerbate or resolve the pro-inflammatory environment. Although leukotriene B4 receptor 2 (BLT2) has been characterized as a low affinity receptor to several key eicosanoids and chemoattractants, its precise roles in the setting of inflammation and macrophage function remain incompletely understood. Here we used zebrafish and mouse models to probe the role of BLT2 in macrophage function during inflammation. We detected BLT2 expression in bone marrow derived and peritoneal macrophages of mouse models. Transcriptomic analysis of Ltb4r2-/- and WT macrophages suggested a role for BLT2 in macrophage migration, and studies in vitro confirmed that whereas BLT2 does not mediate macrophage polarization, it is required for chemotactic function, possibly mediated by downstream genes Ccl5 and Lgals3. Using a zebrafish model of tailfin injury, we demonstrated that antisense morpholino-mediated knockdown of blt2a or chemical inhibition of BLT2 signaling impairs macrophage migration. We further replicated these findings in zebrafish models of islet injury and liver inflammation. Moreover, we established the applicability of our zebrafish findings to mammals by showing that macrophages of Ltb4r2-/- mice have defective migration during lipopolysaccharide stimulation in vivo. Collectively, our results demonstrate that BLT2 mediates macrophage migration during inflammation, which implicates it as a potential therapeutic target for inflammatory pathologies.


Assuntos
Movimento Celular , Macrófagos , Receptores do Leucotrieno B4 , Animais , Camundongos , Inflamação/genética , Inflamação/metabolismo , Leucotrieno B4/genética , Leucotrieno B4/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Receptores do Leucotrieno B4/genética , Receptores do Leucotrieno B4/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA