Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.664
Filter
1.
Biomaterials ; 312: 122711, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39088911

ABSTRACT

The unsuitable deformation stimulus, harsh urine environment, and lack of a regenerative microenvironment (RME) prevent scaffold-based urethral repair and ultimately lead to irreversible urethral scarring. The researchers clarify the optimal elastic modulus of the urethral scaffolds for urethral repair and design a multilayered PVA hydrogel scaffold for urethral scar-free healing. The inner layer of the scaffold has self-healing properties, which ensures that the wound effectively resists harsh urine erosion, even when subjected to sutures. In addition, the scaffold's outer layer has an extracellular matrix-like structure that synergizes with adipose-derived stem cells to create a favorable RME. In vivo experiments confirm successful urethral scar-free healing using the PVA multilayered hydrogel scaffold. Further mechanistic study shows that the PVA multilayer hydrogel effectively resists the urine-induced inflammatory response and accelerates the transition of urethral wound healing to the proliferative phase by regulating macrophage polarization, thus providing favorable conditions for urethral scar-free healing. This study provides mechanical criteria for the fabrication of urethral tissue-engineered scaffolds, as well as important insights into their design.


Subject(s)
Elastic Modulus , Hydrogels , Tissue Scaffolds , Urethra , Wound Healing , Tissue Scaffolds/chemistry , Animals , Hydrogels/chemistry , Tissue Engineering/methods , Mice , Regeneration , Cicatrix/pathology , Male , Cellular Microenvironment , Rats, Sprague-Dawley , Stem Cells/cytology
2.
Front Immunol ; 15: 1432841, 2024.
Article in English | MEDLINE | ID: mdl-39267742

ABSTRACT

Traumatic spinal cord injury (tSCI) is a severe injury to the central nervous system that is categorized into primary and secondary injuries. Among them, the local microenvironmental imbalance in the spinal cord caused by secondary spinal cord injury includes accumulation of cytokines and chemokines, reduced angiogenesis, dysregulation of cellular energy metabolism, and dysfunction of immune cells at the site of injury, which severely impedes neurological recovery from spinal cord injury (SCI). In recent years, single-cell techniques have revealed the heterogeneity of multiple immune cells at the genomic, transcriptomic, proteomic, and metabolomic levels after tSCI, further deepening our understanding of the mechanisms underlying tSCI. However, spatial information about the tSCI microenvironment, such as cell location and cell-cell interactions, is lost in these approaches. The application of spatial multi-omics technology can solve this problem by combining the data obtained from immunohistochemistry and multiparametric analysis to reveal the changes in the microenvironment at different times of secondary injury after SCI. In this review, we systematically review the progress of spatial multi-omics techniques in the study of the microenvironment after SCI, including changes in the immune microenvironment and discuss potential future therapeutic strategies.


Subject(s)
Cellular Microenvironment , Proteomics , Spinal Cord Injuries , Spinal Cord Injuries/immunology , Spinal Cord Injuries/metabolism , Humans , Cellular Microenvironment/immunology , Proteomics/methods , Animals , Metabolomics/methods , Genomics/methods , Transcriptome , Single-Cell Analysis , Multiomics
3.
Nat Commun ; 15(1): 7809, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39242588

ABSTRACT

Innate lymphoid cells (ILCs) are critical in maintaining tissue homeostasis, and during infection and inflammation. Here we identify, by using combinatorial reporter mice, a rare ILC progenitor (ILCP) population, resident to the small intestinal lamina propria (siLP) in adult mice. Transfer of siLP-ILCP into recipients generates group 1 ILCs (including ILC1 and NK cells), ILC2s and ILC3s within the intestinal microenvironment, but almost exclusively group 1 ILCs in the liver, lung and spleen. Single cell gene expression analysis and high dimensional spectral cytometry analysis of the siLP-ILCPs and ILC progeny indicate that the phenotype of the group 1 ILC progeny is also influenced by the tissue microenvironment. Thus, a local pool of siLP-ILCP can contribute to pan-ILC generation in the intestinal microenvironment but has more restricted potential in other tissues, with a greater propensity than bone marrow-derived ILCPs to favour ILC1 and ILC3 production. Therefore, ILCP potential is influenced by both tissue of origin and the microenvironment during development. This may provide additional flexibility during the tuning of immune reactions.


Subject(s)
Immunity, Innate , Intestinal Mucosa , Lymphoid Progenitor Cells , Mice, Inbred C57BL , Animals , Mice , Intestinal Mucosa/immunology , Intestinal Mucosa/cytology , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Lymphoid Progenitor Cells/immunology , Cellular Microenvironment/immunology , Lymphocytes/immunology , Intestine, Small/immunology , Intestine, Small/cytology , Female , Male
4.
Nat Commun ; 15(1): 7010, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237549

ABSTRACT

Kidney injury disrupts the intricate renal architecture and triggers limited regeneration, together with injury-invoked inflammation and fibrosis. Deciphering the molecular pathways and cellular interactions driving these processes is challenging due to the complex tissue structure. Here, we apply single cell spatial transcriptomics to examine ischemia-reperfusion injury in the mouse kidney. Spatial transcriptomics reveals injury-specific and spatially-dependent gene expression patterns in distinct cellular microenvironments within the kidney and predicts Clcf1-Crfl1 in a molecular interplay between persistently injured proximal tubule cells and their neighboring fibroblasts. Immune cell types play a critical role in organ repair. Spatial analysis identifies cellular microenvironments resembling early tertiary lymphoid structures and associated molecular pathways. Collectively, this study supports a focus on molecular interactions in cellular microenvironments to enhance understanding of injury, repair and disease.


Subject(s)
Cell Communication , Cellular Microenvironment , Kidney , Regeneration , Reperfusion Injury , Transcriptome , Animals , Mice , Regeneration/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Kidney/metabolism , Kidney/pathology , Mice, Inbred C57BL , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Fibroblasts/metabolism , Gene Expression Profiling , Single-Cell Analysis , Fibrosis
5.
Am J Reprod Immunol ; 92(3): e13921, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39225584

ABSTRACT

PROBLEM: Endometrial immune cells are essential for maintaining homeostasis and the endometrial receptivity to embryo implantation. Understanding regional variations in endometrial immune cell populations is crucial for comprehending normal endometrial function and the pathophysiology of endometrial disorders. Despite previous studies focusing on the overall immune cell composition and function in the endometrium, regional variations in premenopausal women remain unclear. METHOD OF STUDY: Endometrial biopsies were obtained from four regions (anterior, posterior, left lateral, and right lateral) of premenopausal women undergoing hysteroscopy with no abnormalities. A 15-color human endometrial immune cell-focused flow cytometry panel was used for analysis. High-dimensional flow cytometry combined with a clustering algorithm was employed to unravel the complexity of endometrial immune cells. Additionally, multiplex immunofluorescent was performed for further validation. RESULTS: Our findings revealed no significant variation in the distribution and abundance of immune cells across different regions under normal conditions during the proliferative phase. Each region harbored similar immune cell subtypes, indicating a consistent immune microenvironment. However, when comparing normal regions to areas with focal hemorrhage, significant differences were observed. An increase in CD8+ T cells highlights the impact of localized abnormalities on the immune microenvironment. CONCLUSIONS: Our study demonstrates that the endometrial immune cell landscape is consistent across different anatomical regions during the proliferative phase in premenopausal women. This finding has important implications for understanding normal endometrial function and the pathophysiology of endometrial disorders.


Subject(s)
Cellular Microenvironment , Endometrium , Humans , Female , Endometrium/immunology , Endometrium/pathology , Adult , Cellular Microenvironment/immunology , Flow Cytometry , Premenopause/immunology , CD8-Positive T-Lymphocytes/immunology , Biopsy
6.
Sci Rep ; 14(1): 18939, 2024 08 15.
Article in English | MEDLINE | ID: mdl-39147768

ABSTRACT

Rheumatoid arthritis (RA) and arthrofibrosis (AF) are both chronic synovial hyperplasia diseases that result in joint stiffness and contractures. They shared similar symptoms and many common features in pathogenesis. Our study aims to perform a comprehensive analysis between RA and AF and identify novel drugs for clinical use. Based on the text mining approaches, we performed a correlation analysis of 12 common joint diseases including arthrofibrosis, gouty arthritis, infectious arthritis, juvenile idiopathic arthritis, osteoarthritis, post infectious arthropathies, post traumatic osteoarthritis, psoriatic arthritis, reactive arthritis, rheumatoid arthritis, septic arthritis, and transient arthritis. 5 bulk sequencing datasets and 4 single-cell sequencing datasets of RA and AF were integrated and analyzed. A novel drug repositioning method was found for drug screening, and text mining approaches were used to verify the identified drugs. RA and AF performed the highest gene similarity (0.77) and functional ontology similarity (0.84) among all 12 joint diseases. We figured out that they share the same key pathogenic cell including CD34 + sublining fibroblasts (CD34-SLF) and DKK3 + sublining fibroblasts (DKK3-SLF). Potential therapeutic target database (PTTD) was established with the differential expressed genes (DEGs) of these key pathogenic cells. Based on the PTTD, 15 potential drugs for AF and 16 potential drugs for RA were identified. This work provides a new perspective on AF and RA study which enhances our understanding of their pathogenesis. It also shed light on their underlying mechanism and open new avenues for drug repositioning studies.


Subject(s)
Arthritis, Rheumatoid , Fibrosis , Synovial Membrane , Humans , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/metabolism , Synovial Membrane/pathology , Synovial Membrane/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Drug Repositioning , Cellular Microenvironment/drug effects , Data Mining
7.
J Nanobiotechnology ; 22(1): 491, 2024 Aug 18.
Article in English | MEDLINE | ID: mdl-39155382

ABSTRACT

Cerium oxide (CeO2) nanospheres have limited enzymatic activity that hinders further application in catalytic therapy, but they have an "oxidation switch" to enhance their catalytic activity by increasing oxygen vacancies. In this study, according to the defect-engineering strategy, we developed PtCuOX/CeO2-X nanozymes as highly efficient SOD/CAT mimics by introducing bimetallic copper (Cu) and platinum (Pt) into CeO2 nanospheres to enhance the oxygen vacancies, in an attempt to combine near-infrared (NIR) irradiation to regulate microenvironment for osteoarthritis (OA) therapy. As expected, the Cu and Pt increased the Ce3+/Ce4+ ratio of CeO2 to significantly enhance the oxygen vacancies, and simultaneously CeO2 (111) facilitated the uniform dispersion of Cu and Pt. The strong metal-carrier interaction synergy endowed the PtCuOX/CeO2-X nanozymes with highly efficient SOD/CAT-like activity by the decreased formation energy of oxygen vacancy, promoted electron transfer, the increased adsorption energy of intermediates, and the decreased reaction activation energy. Besides, the nanozymes have excellent photothermal conversion efficiency (55.41%). Further, the PtCuOX/CeO2-X antioxidant system effectively scavenged intracellular ROS and RNS, protected mitochondrial function, and inhibited the inflammatory factors, thus reducing chondrocyte apoptosis. In vivo, experiments demonstrated the biosafety of PtCuOX/CeO2-X and its potent effect on OA suppression. In particular, NIR radiation further enhanced the effects. Mechanistically, PtCuOX/CeO2-X nanozymes reduced ras-related C3 botulinum toxin substrate 1 (Rac-1) and p-p65 protein expression, as well as ROS levels to remodel the inflammatory microenvironment by inhibiting the ROS/Rac-1/nuclear factor kappa-B (NF-κB) signaling pathway. This study introduces new clinical concepts and perspectives that can be applied to inflammatory diseases.


Subject(s)
Cerium , Copper , Osteoarthritis , Platinum , Superoxide Dismutase , Cerium/chemistry , Cerium/pharmacology , Copper/chemistry , Copper/pharmacology , Animals , Superoxide Dismutase/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Platinum/chemistry , Platinum/pharmacology , Mice , Oxygen/metabolism , Oxygen/chemistry , Reactive Oxygen Species/metabolism , Catalase/metabolism , Catalase/chemistry , Humans , Chondrocytes/metabolism , Chondrocytes/drug effects , Antioxidants/pharmacology , Antioxidants/chemistry , Cellular Microenvironment/drug effects , Male
8.
Front Immunol ; 15: 1403458, 2024.
Article in English | MEDLINE | ID: mdl-39161767

ABSTRACT

Genetic mutations in the ß-globin gene lead to a decrease or removal of the ß-globin chain, causing the build-up of unstable alpha-hemoglobin. This condition is referred to as beta-thalassemia (BT). The present treatment strategies primarily target the correction of defective erythropoiesis, with a particular emphasis on gene therapy and hematopoietic stem cell transplantation. However, the presence of inefficient erythropoiesis in BT bone marrow (BM) is likely to disturb the previously functioning BM microenvironment. This includes accumulation of various macromolecules, damage to hematopoietic function, destruction of bone cell production and damage to osteoblast(OBs), and so on. In addition, the changes of BT BM microenvironment may have a certain correlation with the occurrence of hematological malignancies. Correction of the microenvironment can be achieved through treatments such as iron chelation, antioxidants, hypoglycemia, and biologics. Hence, This review describes damage in the BT BM microenvironment and some potential remedies.


Subject(s)
Bone Marrow , Cellular Microenvironment , beta-Thalassemia , Humans , Bone Marrow/pathology , Bone Marrow/metabolism , beta-Thalassemia/therapy , Genetic Therapy , Animals , Thalassemia/therapy , Erythropoiesis , Hematopoietic Stem Cell Transplantation/adverse effects , Iron Chelating Agents/therapeutic use , beta-Globins/genetics
9.
J Immunol Res ; 2024: 4751168, 2024.
Article in English | MEDLINE | ID: mdl-39104594

ABSTRACT

In recent years, as the aging population continues to grow, osteoarthritis (OA) has emerged as a leading cause of disability, with its incidence rising annually. Current treatments of OA include exercise and medications in the early stages and total joint replacement in the late stages. These approaches only relieve pain and reduce inflammation; however, they have significant side effects and high costs. Therefore, there is an urgent need to identify effective treatment methods that can delay the pathological progression of this condition. The changes in the articular cartilage microenvironment, which are complex and diverse, can aggravate the pathological progression into a vicious cycle, inhibiting the repair and regeneration of articular cartilage. Understanding these intricate changes in the microenvironment is crucial for devising effective treatment modalities. By searching relevant research articles and clinical trials in PubMed according to the keywords of articular cartilage, microenvironment, OA, mechanical force, hypoxia, cytokine, and cell senescence. This study first summarizes the factors affecting articular cartilage regeneration, then proposes corresponding treatment strategies, and finally points out the future research direction. We find that regulating the opening of mechanosensitive ion channels, regulating the expression of HIF-1, delivering growth factors, and clearing senescent cells can promote the formation of articular cartilage regeneration microenvironment. This study provides a new idea for the treatment of OA in the future, which can promote the regeneration of articular cartilage through the regulation of the microenvironment so as to achieve the purpose of treating OA.


Subject(s)
Cartilage, Articular , Cellular Microenvironment , Osteoarthritis , Regeneration , Humans , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cartilage, Articular/physiology , Osteoarthritis/therapy , Osteoarthritis/pathology , Animals , Chondrocytes/metabolism , Chondrocytes/physiology , Cellular Senescence
10.
Nat Commun ; 15(1): 6651, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39103330

ABSTRACT

Myocardial infarction (MI) has a 5-year mortality rate of more than 50% due to the lack of effective treatments. Interactions between cardiomyocytes and the MI microenvironment (MIM) can determine the progression and fate of infarcted myocardial tissue. Here, a specially designed Melanin-based composite nanomedicines (MCN) is developed to effectively treat MI by reprogramming the MIM. MCN is a nanocomposite composed of polydopamine (P), Prussian blue (PB) and cerium oxide (CexOy) with a Mayuan-like structure, which reprogramming the MIM by the efficient conversion of detrimental substances (H+, reactive oxygen species, and hypoxia) into beneficial status (O2 and H2O). In coronary artery ligation and ischemia reperfusion models of male mice, intravenously injecting MCN specifically targets the damaged area, resulting in restoration of cardiac function. With its promising therapeutic effects, MCN constitutes a new agent for MI treatment and demonstrates potential for clinical application.


Subject(s)
Cerium , Indoles , Melanins , Myocardial Infarction , Nanomedicine , Polymers , Animals , Melanins/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Male , Mice , Nanomedicine/methods , Indoles/chemistry , Polymers/chemistry , Cerium/chemistry , Cerium/pharmacology , Cerium/administration & dosage , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Nanocomposites/chemistry , Disease Models, Animal , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL , Cellular Microenvironment/drug effects , Ferrocyanides
11.
BMC Cardiovasc Disord ; 24(1): 432, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152369

ABSTRACT

BACKGROUND: Heart failure (HF), which is caused by cardiac overload and injury, is linked to significant mortality. Writers of RNA modification (WRMs) play a crucial role in the regulation of epigenetic processes involved in immune response and cardiovascular disease. However, the potential roles of these writers in the immunological milieu of HF remain unknown. METHODS: We comprehensively characterized the expressions of 28 WRMs using datasets GSE145154 and GSE141910 to map the cardiac immunological microenvironment in HF patients. Based on the expression of WRMs, the immunological cells in the datasets were scored. RESULTS: Single-cell transcriptomics analysis (GSE145154) revealed immunological dysregulation in HF as well as differential expression of WRMs in immunological cells from HF and non-HF (NHF) samples. WRM-scored immunological cells were positively correlated with the immunological response, and the high WRM score group exhibited elevated immunological cell infiltration. WRMs are involved in the differentiation of T cells and myeloid cells. WRM scores of T cell and myeloid cell subtypes were significantly reduced in the HF group compared to the NHF group. We identified a myogenesis-related resident macrophage population in the heart, Macro-MYL2, that was characterized by an increased expression of cardiomyocyte structural genes (MYL2, TNNI3, TNNC1, TCAP, and TNNT2) and was regulated by TRMT10C. Based on the WRM expression pattern, the transcriptomics data (GSE141910) identified two distinct clusters of HF samples, each with distinct functional enrichments and immunological characteristics. CONCLUSION: Our study demonstrated a significant relationship between the WRMs and immunological microenvironment in HF, as well as a novel resident macrophage population, Macro-MYL2, characterized by myogenesis. These results provide a novel perspective on the underlying mechanisms and therapeutic targets for HF. Further experiments are required to validate the regulation of WRMs and Macro-MYL2 macrophage subtype in the cardiac immunological milieu.


Subject(s)
Gene Expression Profiling , Heart Failure , Macrophages , Single-Cell Analysis , Transcriptome , Humans , Heart Failure/genetics , Heart Failure/immunology , Heart Failure/metabolism , Macrophages/metabolism , Macrophages/immunology , Databases, Genetic , Cellular Microenvironment , RNA Processing, Post-Transcriptional , Animals , Case-Control Studies , Gene Expression Regulation
12.
CNS Neurosci Ther ; 30(8): e14903, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39139089

ABSTRACT

INTRODUCTION: Excessive neuroinflammation, apoptosis, glial scar, and demyelination triggered by spinal cord injury (SCI) are major obstacles to SCI repair. Fucoidan, a natural marine plant extract, possesses broad-spectrum anti-inflammatory and immunomodulatory effects and is regarded as a potential therapeutic for various diseases, including neurological disorders. However, its role in SCI has not been investigated. METHODS: In this study, we established an SCI model in mice and intervened in injury repair by daily intraperitoneal injections of different doses of fucoidan (10 and 20 mg/kg). Concurrently, primary oligodendrocyte precursor cells (OPCs) were treated in vitro to validate the differentiation-promoting effect of fucoidan on OPCs. Basso Mouse Scale (BMS), Louisville Swim Scale (LSS), and Rotarod test were carried out to measure the functional recovery. Immunofluorescence staining, and transmission electron microscopy (TEM) were performed to assess the neuroinflammation, apoptosis, glial scar, and remyelination. Western blot analysis was conducted to clarify the underlying mechanism of remyelination. RESULTS: Our results indicate that in the SCI model, fucoidan exhibits significant anti-inflammatory effects and promotes the transformation of pro-inflammatory M1-type microglia/macrophages into anti-inflammatory M2-type ones. Fucoidan enhances the survival of neurons and axons in the injury area and improves remyelination. Additionally, fucoidan promotes OPCs differentiation into mature oligodendrocytes by activating the PI3K/AKT/mTOR pathway. CONCLUSION: Fucoidan improves SCI repair by modulating the microenvironment and promoting remyelination.


Subject(s)
Mice, Inbred C57BL , Polysaccharides , Recovery of Function , Remyelination , Spinal Cord Injuries , Animals , Polysaccharides/pharmacology , Mice , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Remyelination/drug effects , Remyelination/physiology , Recovery of Function/drug effects , Oligodendrocyte Precursor Cells/drug effects , Female , Cellular Microenvironment/drug effects
13.
Int J Mol Sci ; 25(16)2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39201715

ABSTRACT

Androgenetic alopecia (AGA) is characterized by microinflammation and abnormal immune responses, particularly in the upper segment of hair follicles (HFs). However, the precise patterns of immune dysregulation remain unclear, partly due to limitations in current analysis techniques to preserve tissue architecture. The infundibulum, a major part of the upper segment of HFs, is associated with significant clusters of immune cells. In this study, we investigated immune cells around the infundibulum, referred to as peri-infundibular immune infiltration (PII). We employed spatial transcriptome profiling, a high-throughput analysis technology, to investigate the immunological disruptions within the PII region. Our comprehensive analysis included an evaluation of overall immune infiltrates, gene set enrichment analysis (GSEA), cellular deconvolution, differential expression analysis, over-representation analysis, protein-protein interaction (PPI) networks, and upstream regulator analysis to identify cell types and molecular dysregulation in immune cells. Our results demonstrated significant differences in immune signatures between the PII of AGA patients (PII-A) and the PII of control donors (PII-C). Specifically, PII-A exhibited an enrichment of CD4+ helper T cells, distinct immune response patterns, and a bias toward a T helper (Th) 2 response. Immunohistochemistry revealed disruptions in T cell subpopulations, with more CD4+ T cells displaying an elevated Th2 response and a reduced Th1-cytotoxic response compared to PII-C. These findings reveal the unique immune landscapes of PII-A and PII-C, suggesting potential for the development of innovative treatment approaches.


Subject(s)
Alopecia , Gene Expression Profiling , Hair Follicle , Transcriptome , Humans , Alopecia/genetics , Alopecia/immunology , Alopecia/metabolism , Alopecia/pathology , Hair Follicle/immunology , Hair Follicle/metabolism , Hair Follicle/pathology , Male , Adult , Protein Interaction Maps , Middle Aged , Female , Cellular Microenvironment/immunology , Cellular Microenvironment/genetics , Th2 Cells/immunology , Th2 Cells/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism
14.
Curr Opin Immunol ; 89: 102453, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39173413

ABSTRACT

The delicate balance between protective immunity against pathogens and the prevention of autoimmunity requires finely tuned generation and function of regulatory CD4+ T (Treg) cells. Here, we review recent progress in the understanding of a complex set of cues, which converge on Treg cells in lymphoid and nonlymphoid organs and in tumors and how these cues modulate Treg functions. We highlight the versatility of Treg cells underlying their ability to dynamically adapt to local microenvironments and perform a wide range of functions that extend beyond the archetypal role of Treg cells in moderating adverse effects of immune response-associated inflammation and in suppressing autoimmunity.


Subject(s)
T-Lymphocytes, Regulatory , Humans , T-Lymphocytes, Regulatory/immunology , Animals , Cellular Microenvironment/immunology , Autoimmunity , Tumor Microenvironment/immunology , Neoplasms/immunology , Inflammation/immunology
15.
Nat Cardiovasc Res ; 3(3): 301-316, 2024 Mar.
Article in English | MEDLINE | ID: mdl-39196111

ABSTRACT

Myocarditis is an inflammatory heart disease that leads to loss of cardiomyocytes and frequently precipitates fibrotic remodeling of the myocardium, culminating in heart failure. However, the molecular mechanisms underlying immune cell control and maintenance of tissue integrity in the inflamed cardiac microenvironment remain elusive. In this study, we found that bone morphogenic protein-4 (BMP4) gradients maintain cardiac tissue homeostasis by single-cell transcriptomics analyses of inflamed murine and human myocardial tissues. Cardiac BMP pathway dysregulation was reflected by reduced BMP4 serum concentration in patients with myocarditis. Restoration of BMP signaling by antibody-mediated neutralization of the BMP inhibitors gremlin-1 and gremlin-2 ameliorated T cell-induced myocardial inflammation in mice. Moreover, progression to inflammatory cardiomyopathy was blocked through the reduction of fibrotic remodeling and preservation of cardiomyocyte integrity. These results unveil the BMP4-gremlin axis as a druggable pathway for the treatment of myocardial inflammation, limiting the severe sequelae of cardiac fibrosis and heart failure.


Subject(s)
Autoimmune Diseases , Bone Morphogenetic Protein 4 , Disease Models, Animal , Fibrosis , Myocarditis , Myocarditis/metabolism , Myocarditis/pathology , Myocarditis/immunology , Animals , Fibrosis/pathology , Fibrosis/metabolism , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Humans , Autoimmune Diseases/pathology , Autoimmune Diseases/metabolism , Autoimmune Diseases/immunology , Male , Signal Transduction , Mice , Cellular Microenvironment , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Female , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Myocardium/immunology
17.
Life Sci ; 354: 122947, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39117138

ABSTRACT

Temporomandibular joint osteoarthritis (TMJOA) is considered to be a low-grade inflammatory disease involving multiple joint tissues. The crosstalk between synovium and cartilage plays an important role in TMJOA. Synovial cells are a group of heterogeneous cells and synovial microenvironment is mainly composed of synovial fibroblasts (SF) and synovial macrophages. In TMJOA, SF and synovial macrophages release a large number of inflammatory cytokines and extracellular vesicles and promote cartilage destruction. Cartilage wear particles stimulate SF proliferation and macrophages activation and exacerbate synovitis. In TMJOA, chondrocytes and synovial cells exhibit increased glycolytic activity and lactate secretion, leading to impaired chondrocyte matrix synthesis. Additionally, the synovium contains mesenchymal stem cells, which are the seed cells for cartilage repair in TMJOA. Co-culture of chondrocytes and synovial mesenchymal stem cells enhances the chondrogenic differentiation of stem cells. This review discusses the pathological changes of synovium in TMJOA, the means of crosstalk between synovium and cartilage, and their influence on each other. Based on the crosstalk between synovium and cartilage in TMJOA, we illustrate the treatment strategies for improving synovial microenvironment, including reducing cell adhesion, utilizing extracellular vesicles to deliver biomolecules, regulating cellular metabolism and targeting inflammatory cytokines.


Subject(s)
Cellular Microenvironment , Chondrocytes , Osteoarthritis , Synovial Membrane , Temporomandibular Joint , Humans , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/therapy , Chondrocytes/metabolism , Chondrocytes/pathology , Synovial Membrane/metabolism , Synovial Membrane/pathology , Animals , Temporomandibular Joint/metabolism , Temporomandibular Joint/pathology , Temporomandibular Joint Disorders/metabolism , Temporomandibular Joint Disorders/pathology , Temporomandibular Joint Disorders/therapy , Mesenchymal Stem Cells/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Cytokines/metabolism , Macrophages/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology
18.
J Hazard Mater ; 478: 135540, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39178783

ABSTRACT

Silicosis represents a form of interstitial lung disease induced by the inhalation of silica particles in production environments. A key pathological characteristic of silica-induced pulmonary fibrosis is its localized tissue heterogeneity, which presents significant challenges in analyzing transcriptomic data due to the loss of important spatial context. To address this, we integrate spatial gene expression data with single-cell analyses and achieve a detailed mapping of cell types within and surrounding fibrotic regions, revealing significant shifts in cell populations in normal and diseased states. Additionally, we explore cell interactions within fibrotic zones using ligand-receptor mapping, deepening our understanding of cellular dynamics in these areas. We identify a subset of fibroblasts, termed Inmt fibroblasts, that play a suppressive role in the fibrotic microenvironment. Validating our findings through a comprehensive suite of bioinformatics, histological, and cell culture studies highlights the role of monocyte-derived macrophages in shifting Inmt fibroblast populations into profibrotic Grem1 fibroblast, potentially disrupting lung homeostasis in response to external challenges. Hence, the spatially detailed deconvolution offered by our research markedly advances the comprehension of cell dynamics and environmental interactions pivotal in the development of pulmonary fibrosis.


Subject(s)
Fibroblasts , Pulmonary Fibrosis , Silicon Dioxide , Silicon Dioxide/toxicity , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Animals , Lung/pathology , Lung/drug effects , Lung/metabolism , Macrophages/drug effects , Macrophages/metabolism , Single-Cell Analysis , Humans , Mice, Inbred C57BL , Mice , Cellular Microenvironment
19.
Circ Res ; 135(7): 758-773, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39140165

ABSTRACT

BACKGROUND: Cardiac hypertrophy is characterized by remodeling of the myocardium, which involves alterations in the ECM (extracellular matrix) and cardiomyocyte structure. These alterations critically contribute to impaired contractility and relaxation, ultimately leading to heart failure. Emerging evidence implicates that extracellular signaling molecules are critically involved in the pathogenesis of cardiac hypertrophy and remodeling. The immunophilin CyPA (cyclophilin A) has been identified as a potential culprit. In this study, we aimed to unravel the interplay between eCyPA (extracellular CyPA) and myocardial dysfunction and evaluate the therapeutic potential of inhibiting its extracellular accumulation to improve heart function. METHODS: Employing a multidisciplinary approach encompassing in silico, in vitro, in vivo, and ex vivo experiments we studied a mouse model of cardiac hypertrophy and human heart specimen to decipher the interaction of CyPA and the cardiac microenvironment in highly relevant pre-/clinical settings. Myocardial expression of CyPA (immunohistology) and the inflammatory transcriptome (NanoString) was analyzed in human cardiac tissue derived from patients with nonischemic, noninflammatory congestive heart failure (n=187). These analyses were paralleled by a mouse model of Ang (angiotensin) II-induced heart failure, which was assessed by functional (echocardiography), structural (immunohistology, atomic force microscopy), and biomolecular (Raman spectroscopy) analyses. The effect of inhibiting eCyPA in the cardiac microenvironment was evaluated using a newly developed neutralizing anti-eCyPA monoclonal antibody. RESULTS: We observed a significant accumulation of eCyPA in both human and murine-failing hearts. Importantly, higher eCyPA expression was associated with poor clinical outcomes in patients (P=0.043) and contractile dysfunction in mice (Pearson correlation coefficient, -0.73). Further, myocardial expression of eCyPA was critically associated with an increase in myocardial hypertrophy, inflammation, fibrosis, stiffness, and cardiac dysfunction in vivo. Antibody-based inhibition of eCyPA prevented (Ang II)-induced myocardial remodeling and dysfunction in mice. CONCLUSIONS: Our study provides strong evidence of the pathogenic role of eCyPA in remodeling, myocardial stiffening, and dysfunction in heart failure. The findings suggest that antibody-based inhibition of eCyPA may offer a novel therapeutic strategy for nonischemic heart failure. Further research is needed to evaluate the translational potential of these interventions in human patients with cardiac hypertrophy.


Subject(s)
Cyclophilin A , Heart Failure , Mice, Inbred C57BL , Animals , Heart Failure/metabolism , Heart Failure/physiopathology , Heart Failure/pathology , Humans , Cyclophilin A/metabolism , Cyclophilin A/genetics , Mice , Male , Cellular Microenvironment , Myocardium/metabolism , Myocardium/pathology , Disease Models, Animal , Female , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/drug effects , Ventricular Remodeling/drug effects
20.
ACS Biomater Sci Eng ; 10(9): 5513-5536, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39173130

ABSTRACT

Irreversible bone defects resulting from trauma, infection, and degenerative illnesses have emerged as a significant health concern. Structurally and functionally controllable hydrogels made by bone tissue engineering (BTE) have become promising biomaterials. Natural proteins are able to establish connections with autologous proteins through unique biologically active regions. Hydrogels based on proteins can simulate the bone microenvironment and regulate the biological behavior of stem cells in the tissue niche, making them candidates for research related to bone regeneration. This article reviews the biological functions of various natural macromolecular proteins (such as collagen, gelatin, fibrin, and silk fibroin) and highlights their special advantages as hydrogels. Then the latest research trends on cross-linking modified macromolecular protein hydrogels with improved mechanical properties and composite hydrogels loaded with exogenous micromolecular proteins have been discussed. Finally, the applications of protein hydrogels, such as 3D printed hydrogels, microspheres, and injectable hydrogels, were introduced, aiming to provide a reference for the repair of clinical bone defects.


Subject(s)
Hydrogels , Osteogenesis , Tissue Engineering , Hydrogels/chemistry , Humans , Osteogenesis/drug effects , Tissue Engineering/methods , Bone Regeneration/drug effects , Animals , Cellular Microenvironment , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Proteins/chemistry , Proteins/metabolism , Tissue Scaffolds/chemistry , Bone and Bones/metabolism , Bone and Bones/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL