Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cell Discov ; 8(1): 95, 2022 Sep 27.
Article in English | MEDLINE | ID: mdl-36163341

ABSTRACT

Our understanding of full-thickness endometrial regeneration after injury is limited by an incomplete molecular characterization of the cell populations responsible for the organ functions. To help fill this knowledge gap, we characterized 10,551 cells of full-thickness normal human uterine from two menstrual phases (proliferative and secretory phase) using unbiased single cell RNA-sequencing. We dissected cell heterogeneity of main cell types (epithelial, stromal, endothelial, and immune cells) of the full thickness uterine tissues, cell population architectures of human uterus cells across the menstrual cycle. We identified an SFRP4+ stromal cell subpopulation that was highly enriched in the regenerative stage of the human endometria during the menstrual cycle, and the SFRP4+ stromal cells could significantly enhance the proliferation of human endometrial epithelial organoid in vitro, and promote the regeneration of endometrial epithelial glands and full-thickness endometrial injury through IGF1 signaling pathway in vivo. Our cell atlas of full-thickness uterine tissues revealed the cellular heterogeneities, cell population architectures, and their cell-cell communications during the monthly regeneration of the human endometria, which provide insight into the biology of human endometrial regeneration and the development of regenerative medicine treatments against endometrial damage and intrauterine adhesion.

3.
Bone Res ; 10(1): 38, 2022 Apr 27.
Article in English | MEDLINE | ID: mdl-35477573

ABSTRACT

Articular cartilage damage is a universal health problem. Despite recent progress, chondrocyte dedifferentiation has severely compromised the clinical outcomes of cell-based cartilage regeneration. Loss-of-function changes are frequently observed in chondrocyte expansion and other pathological conditions, but the characteristics and intermediate molecular mechanisms remain unclear. In this study, we demonstrate a time-lapse atlas of chondrocyte dedifferentiation to provide molecular details and informative biomarkers associated with clinical chondrocyte evaluation. We performed various assays, such as single-cell RNA sequencing (scRNA-seq), live-cell metabolic assays, and assays for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq), to develop a biphasic dedifferentiation model consisting of early and late dedifferentiation stages. Early-stage chondrocytes exhibited a glycolytic phenotype with increased expression of genes involved in metabolism and antioxidation, whereas late-stage chondrocytes exhibited ultrastructural changes involving mitochondrial damage and stress-associated chromatin remodeling. Using the chemical inhibitor BTB06584, we revealed that early and late dedifferentiated chondrocytes possessed distinct recovery potentials from functional phenotype loss. Notably, this two-stage transition was also validated in human chondrocytes. An image-based approach was established for clinical use to efficiently predict chondrocyte plasticity using stage-specific biomarkers. Overall, this study lays a foundation to improve the quality of chondrocytes in clinical use and provides deep insights into chondrocyte dedifferentiation.

4.
Aging (Albany NY) ; 13(24): 25886-25902, 2021 12 19.
Article in English | MEDLINE | ID: mdl-34923484

ABSTRACT

Pelvic organ prolapse is a worldwide health problem to elderly women. Understanding its pathogenesis and an ideal animal model are crucial to developing promising treatments. The present study aimed to investigate new clinical significance and detailed mechanism of pelvic organ prolapse by comparing the structural, functional and molecular dysfunctions of pelvic organ prolapse in patient and Loxl1 deficient mice. Our results showed that human vagina tissues from prolapsed site showed disarranged collagen and elastic fibers compared with the non-prolapse tissue. A gene ontology (GO) analysis of differentially expressed genes revealed molecular changes mainly related to inflammatory response and extracellular matrix (ECM) organization. While the mice lacking Loxl1 developed stable POP phenotype and disordered ECM structure in histology. Such Loxl1 knockout mice exhibited a significantly urinary dysfunction and decreased mechanical properties of the pelvic floor tissues, implying that POP in human condition might be induced by progressively decreased mechanics of pelvic tissues following ECM catabolism. Similarly, we not only identified significant up-regulated ECM catabolism processes and down-regulated ECM synthesis processes, but also characterized high level of inflammatory response in vagina tissue of the Loxl1 deficient mice. Thus, all these pathological changes in the POP mice model was consistent with those of the clinical elderly patients. These findings provide new insight into remodeling of POP by LOXL1 regulation and be of great importance to develop combination treatments of ECM metabolism and inflammation regulation strategy.


Subject(s)
Amino Acid Oxidoreductases/genetics , Gene Ontology , Pelvic Organ Prolapse , Aged , Aged, 80 and over , Animals , Collagen/metabolism , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Pelvic Organ Prolapse/genetics , Pelvic Organ Prolapse/physiopathology
5.
Front Cell Neurosci ; 15: 720271, 2021.
Article in English | MEDLINE | ID: mdl-34658791

ABSTRACT

Background: Spinal cord injury (SCI) is a highly lethal and debilitating disease with a variety of etiologies. To date, there is no effective therapeutic modality for a complete cure. The pathological mechanisms of spinal cord injury at the molecular gene and protein expression levels remain unclear. Methods: This study used single-cell transcriptomic analysis and protein microarray analysis to analyzes changes in the gene expression profiles of cells and secretion of inflammatory factors respectively, around the lesion site in a rat SCI model. Results: Single-cell transcriptomic analysis found that three types of glial cells (microglia, astrocyte, and oligodendrocyte) becomes activated after acute injury, with GO exhibiting a variety of inflammatory-related terms after injury, such as metabolic processes, immune regulation, and antigen presentation. Protein microarray results showed that the levels of four inflammatory cytokines favoring SCI repair decreased while the levels of nine inflammatory cytokines hindering SCI repair increased after injury. Conclusion: These findings thus reveal the changes in cellular state from homeostatic to reactive cell type after SCI, which contribute to understand the pathology process of SCI, and the potential relationship between glial cells and inflammatory factors after SCI, and provides new theoretical foundation for further elucidating the molecular mechanisms of secondary SCI.

6.
Biomaterials ; 277: 121116, 2021 10.
Article in English | MEDLINE | ID: mdl-34478932

ABSTRACT

Macrophages play crucial roles in host tissue reaction to biomaterials upon implantation in vivo. However, the complexity of biomaterial degradation-related macrophage subpopulations that accumulate around the implanted biomaterials in situ is not fully understood. Here, using single cell RNA-seq, we analyze the transcriptome profiles of the various cell types around the scaffold to map the scaffold-induced reaction, in an unbiased approach. This enables mapping of all biomaterial degradation-associated cells at high resolution, revealing distinct subpopulations of tissue-resident macrophages as the major cellular sources of biomaterial degradation in situ. We also find that scaffold architecture can affect the mechanotransduction and catabolic activity of specific material degradation-related macrophage subpopulations in an Itgav-Mapk1-Stat3 dependent manner, eventually leading to differences in scaffold degradation rate in vivo. Our work dissects unanticipated aspects of the cellular and molecular basis of biomaterial degradation at the single-cell level, and provides a conceptual framework for developing functional tissue engineering scaffolds in future.


Subject(s)
Biocompatible Materials , Mechanotransduction, Cellular , Macrophages , RNA-Seq , Tissue Scaffolds
7.
Cell Prolif ; 54(7): e13077, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34105806

ABSTRACT

OBJECTIVE: The loss of LOXL1 expression reportedly leads to the prolapse of pelvic organs or to exfoliation syndrome glaucoma. Increasing evidence suggests that LOXL1 deficiency is associated with the pathogenesis of several other diseases. However, the characterization of the systemic functions of LOXL1 is limited by the lack of relevant investigative technologies. MATERIALS AND METHODS: To determine the functions of LOXL1, a novel method for body-wide organ transcriptome profiling, combined with single-cell mass cytometry, was developed. A body-wide organ transcriptomic (BOT) map was created by RNA-Seq of tissues from 17 organs from both Loxl1 knockout (KO) and wild-type mice. RESULTS: The BOT results indicated the systemic upregulation of genes encoding proteins associated with the immune response and proliferation processes in multiple tissues of KO mice, and histological and immune staining confirmed the hyperplasia and infiltration of local immune cells in the tissues of KO mice. Furthermore, mass cytometry analysis of peripheral blood samples revealed systemic immune changes in KO mice. These findings were well correlated with results obtained from cancer databases. Patients with tumours had higher Loxl1 mutation frequencies, and patients with Loxl1-mutant tumours showed the upregulation of immune processes and cell proliferation and lower survival rates. CONCLUSION: This study provides an effective strategy for the screening of gene functions in multiple organs and also illustrates the important biological roles of LOXL1 in the cells of multiple organs as well as in systemic immunity.


Subject(s)
Amino Acid Oxidoreductases/genetics , Transcriptome , Amino Acid Oxidoreductases/deficiency , Animals , Female , Gene Expression Profiling , Liver/metabolism , Liver/pathology , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA-Seq , Single-Cell Analysis , Skin/metabolism , Skin/pathology , Vagina/metabolism , Vagina/pathology
8.
Adv Sci (Weinh) ; 7(23): 2000938, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33304744

ABSTRACT

Although being of utmost importance for human health and mobility, stem cell identity and hierarchical organization of musculoskeletal progenitors remain largely unexplored. Here, cells from E10.5, E12.5, and E15.5 murine limbs are analyzed by high throughput single-cell RNA sequencing to illustrate the cellular architecture during limb development. Single-cell transcriptional profiling demonstrates the identity and differentiation architecture of musculoskeletal stem cells (MSSC), soft and hard tissue progenitors through expression pattern of musculoskeletal markers (scleraxis [Scx], Hoxd13, Sox9, and Col1a1). This is confirmed by genetic in vivo lineage tracing. Moreover, single-cell analyses of Scx knockout mice tissues illustrates that Scx regulates MSSC self-renewal and proliferation potential. A high-throughput and low-cost multi-tissues RNA sequencing strategy further provides evidence that musculoskeletal system tissues, including muscle, bone, meniscus, and cartilage, are all abnormally developed in Scx knockout mice. These results establish the presence of an indispensable limb Scx+Hoxd13+ MSSC population and their differentiation into soft tissue progenitors (Scx+Col1a1+) and hard tissue progenitors (Scx+Sox9+). Collectively, this study paves the way for systematically decoding the complex molecular mechanisms and cellular programs of musculoskeletal tissues morphogenesis in limb development and regeneration.

9.
Stem Cell Reports ; 14(3): 478-492, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32084387

ABSTRACT

Articular cartilage injury and degeneration causing pain and loss of quality-of-life has become a serious problem for increasingly aged populations. Given the poor self-renewal of adult human chondrocytes, alternative functional cell sources are needed. Direct reprogramming by small molecules potentially offers an oncogene-free and cost-effective approach to generate chondrocytes, but has yet to be investigated. Here, we directly reprogrammed mouse embryonic fibroblasts into PRG4+ chondrocytes using a 3D system with a chemical cocktail, VCRTc (valproic acid, CHIR98014, Repsox, TTNPB, and celecoxib). Using single-cell transcriptomics, we revealed the inhibition of fibroblast features and activation of chondrogenesis pathways in early reprograming, and the intermediate cellular process resembling cartilage development. The in vivo implantation of chemical-induced chondrocytes at defective articular surfaces promoted defect healing and rescued 63.4% of mechanical function loss. Our approach directly converts fibroblasts into functional cartilaginous cells, and also provides insights into potential pharmacological strategies for future cartilage regeneration.


Subject(s)
Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibrocartilage/cytology , Animals , Cellular Reprogramming , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis , Fibroblasts/metabolism , Mice , Organoids/cytology , Regeneration , Tissue Scaffolds/chemistry , Transcriptome/genetics
10.
Biomaterials ; 232: 119724, 2020 02.
Article in English | MEDLINE | ID: mdl-31918221

ABSTRACT

Changes in the stiffness of chondrocyte extracellular matrix (ECM) are involved in the pathological progression of osteoarthritis (OA). However, the downstream responses of cartilage ECM stiffness are still unclear. YAP (Yes-associated protein) has been extensively studied as a mechanotransducer, we thus hypothesized that by targeting the downstream molecule activity of ECM stiffness could maintain chondrocyte phenotype and prevent cartilage degeneration in OA. Here, we showed that human cartilage matrix stiffened during pathological progression of OA, and the chondrocyte YAP activity was associated with ECM stiffness. We then mimicked the physiological and pathological stiffness of human cartilage by using PDMS-based substrates, and found that YAP was activated in chondrocytes seeded on stiff substrate, gradually losing their phenotype. In addition, it was observed that YAP was also significantly activated in mice OA development, and conditional knockout (cKO) of YAP in mice preserved collagen II expression and protected cartilage from degeneration in the OA model. Furthermore, intra-articular injection of YAP-selective inhibitor, Verteporfin, significantly maintained cartilage homeostasis in mice OA model. This study indicates that the application of mechanotransducer-targeted drugs could be a potential therapeutic approach for cartilage repair in OA.


Subject(s)
Cartilage, Articular , Chitosan , Osteoarthritis , Animals , Chondrocytes , Mice , Microspheres , Osteoarthritis/drug therapy , Verteporfin
11.
Cell Death Dis ; 10(6): 427, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31160550

ABSTRACT

Characterized by their slow adhering property, skeletal muscle myogenic progenitor cells (MPCs) have been widely utilized in skeletal muscle tissue engineering for muscle regeneration, but with limited efficacy. Skeletal muscle regeneration is regulated by various cell types, including a large number of rapidly adhering cells (RACs) where their functions and mechanisms are still unclear. In this study, we explored the function of RACs by co-culturing them with MPCs in a biomimetic skeletal muscle organoid system. Results showed that RACs promoted the myogenic potential of MPCs in the organoid. Single-cell RNA-Seq was also performed, classifying RACs into 7 cell subtypes, including one newly described cell subtype: teno-muscular cells (TMCs). Connectivity map of RACs and MPCs subpopulations revealed potential growth factors (VEGFA and HBEGF) and extracellular matrix (ECM) proteins involvement in the promotion of myogenesis of MPCs during muscle organoid formation. Finally, trans-well experiments and small molecular inhibitors blocking experiments confirmed the role of RACs in the promotion of myogenic differentiation of MPCs. The RACs reported here revealed complex cell diversity and connectivity with MPCs in the biomimetic skeletal muscle organoid system, which not only offers an attractive alternative for disease modeling and in vitro drug screening but also provides clues for in vivo muscle regeneration.


Subject(s)
Muscle Development/genetics , Muscle, Skeletal/metabolism , Myoblasts/metabolism , Organoids/cytology , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cluster Analysis , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/cytology , Myoblasts/cytology , Organoids/ultrastructure , RNA-Seq , Single-Cell Analysis , Transcriptome/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
12.
Biomaterials ; 205: 38-49, 2019 06.
Article in English | MEDLINE | ID: mdl-30903824

ABSTRACT

Nanoparticles are widely developed and utilized in the pharmaceutical and medicine industry, as they can be easily distributed and infiltrated throughout the whole body once administered; however, the body wide effect of nanoparticles infiltration is still unclear. In this study, we developed a new strategy of Nano Genome Altas (NGA) of multi-tissues to study the acute Body-wide-Organ-Transcriptomic response to nanomaterials. Hydroxyapatite(HA)-Nanoparticles (HANPs) was applied in this study as an example both in vitro and in vivo. Results showed that the effect of HANPs is organ specific and mainly related to immune responses in spleen and muscle, proliferation in spleen and bone, stress and apoptosis in spleen and PBMC, ion transport in spleen, kidney, and liver tissues, metabolism in heart, spleen, and muscle, as well as tissue specific epigenetic and signal pathways. In vitro experiments also confirmed that the effects of HANPs on different tissue stem cells were tissue specific. Thus, Nano Genome Altas can provide a body-wide view of the transcriptomic response of multiple organs and tissue specific stem cells to HANPs; it could also be useful for optimizing HANPs and other nano-delivery systems.


Subject(s)
Genome , Nanoparticles/chemistry , Organ Specificity , Animals , Apoptosis , Cell Proliferation , Durapatite/chemistry , Epigenesis, Genetic , Gene Expression Regulation , Gene Ontology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunity, Innate , Mice, Inbred C57BL , Nanoparticles/ultrastructure , Signal Transduction , Tissue Distribution
13.
EBioMedicine ; 32: 223-233, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29898872

ABSTRACT

Osteoarthritis (OA) is a leading cause of physical disability among aging populations, with no available drugs able to efficiently restore the balance between cartilage matrix synthesis and degradation. Also, OA has not been accurately classified into subpopulations, hindering the development toward personalized precision medicine. In the present study, we identified a subpopulation of OA patients displaying high activation level of epidermal growth factor receptor (EGFR). With Col2a1-creERT2; Egfrf/f mice, it was found that the activation of EGFR, indicated by EGFR phosphorylation (pEGFR), led to the destruction of joints. Excitingly, EGFR inhibition prohibited cartilage matrix degeneration and promoted cartilage regeneration. The Food and Drug Administration (FDA)-approved drug gefitinib could efficiently inhibit EGFR functions in OA joints and restore cartilage structure and function in the mouse model as well as the clinical case report. Overall, our findings suggested the concept of the EGFR activated OA subpopulation and illustrated the mechanism of EGFR signaling in regulating cartilage homeostasis. Gefitinib could be a promising disease-modifying drug for this OA subpopulation treatment.


Subject(s)
ErbB Receptors/genetics , Osteoarthritis/drug therapy , Osteoarthritis/genetics , Quinazolines/administration & dosage , Animals , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Collagen Type II/genetics , Disease Models, Animal , Gefitinib , Humans , Mice , Mice, Transgenic , Osteoarthritis/classification , Osteoarthritis/pathology , Precision Medicine , Signal Transduction/drug effects
14.
Adv Healthc Mater ; 7(6): e1701014, 2018 03.
Article in English | MEDLINE | ID: mdl-29334188

ABSTRACT

Developing soft devices for invasive procedures bears great importance for human health. The softness and large strain actuation of responsive hydrogels promise the potential to fabricate soft devices, which can attach on and assist to the function of organs. The key challenges lie in the fabrication of soft devices with robust actuating ability and biocompatibility to the attached organ. This paper presents a solution that integrates the thermoresponsive hydrogel membrane with flexible electronics and silk scaffold into a balloon-like soft device. As an example, the actuation assisting function of this soft device for shrinking an animal bladder is presented. The mechanical behaviors of the balloon-like soft device are experimentally and theoretically investigated. The concepts are applicable to other applications such as soft implants, soft robotics, and microfluidics.


Subject(s)
Hydrogels/chemistry , Membranes, Artificial , Microfluidics/methods , Models, Biological , Silk/chemistry , Urinary Bladder , Animals , Humans
15.
Stem Cell Reports ; 9(1): 381-396, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28625536

ABSTRACT

The endometrial layer comprises luminal and glandular epithelia that both develop from the same simple layer of fetal uterine epithelium. Mechanisms of uterine epithelial progenitor self-renewal and differentiation are unclear. This study aims to systematically analyze the molecular and cellular mechanisms of uterine epithelial development by single-cell analysis. An integrated set of single-cell transcriptomic data of uterine epithelial progenitors and their differentiated progenies is provided. Additionally the unique molecular signatures of these cells, characterized by sequential upregulation of specific epigenetic and metabolic activities, and activation of unique signaling pathways and transcription factors, were also investigated. Finally a unique subpopulation of early progenitor, as well as differentiated luminal and glandular lineages, were identified. A complex cellular hierarchy of uterine epithelial development was thus delineated. Our study therefore systematically decoded molecular markers and a cellular program of uterine epithelial development that sheds light on uterine developmental biology.


Subject(s)
Cell Lineage , Epithelial Cells/cytology , Single-Cell Analysis/methods , Stem Cells/cytology , Transcriptome , Uterus/cytology , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Cell Differentiation , Cells, Cultured , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Developmental , Mice , Mice, Inbred ICR , Retinal Dehydrogenase , Signal Transduction , Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Uterus/growth & development , Uterus/metabolism
16.
Transl Stroke Res ; 7(6): 512-520, 2016 12.
Article in English | MEDLINE | ID: mdl-27539574

ABSTRACT

Traumatic brain injury (TBI) is a major cause of death and disability in young adults. Long-term mental disability often occurs in patients suffering moderate and severe TBI while not as frequent in the victims of mild TBI. To explore the potential mechanism underlying this severity-dependent cognitive deficit, we subjected C57/BL6 mice to different severities of controlled cortical impact (CCI) and assessed their learning-memory functions. The mice subjected to moderate and severe TBI exhibited significantly impaired long-term spatial learning-memory ability, which was accompanied by marked white matter injury and hippocampus damage. In contrast, long-term learning-memory deficits or structural abnormalities within the hippocampus or white matter were not significant in the case of mild TBI. According to a correlation analysis, the hippocampus or white matter injury severity was more relevant to Morris water maze outcome than tissue volume. This study revealed that long-term spatial learning-memory deficits are dependent on the severity of destruction in the white matter and hippocampus. Therapeutic strategies targeting both the white matter and hippocampus may be needed to improve the neurological functions in TBI victims.


Subject(s)
Brain Injuries, Traumatic/complications , Memory Disorders/etiology , Spatial Learning/physiology , Animals , Brain Injuries, Traumatic/pathology , Disease Models, Animal , Hippocampus/pathology , Male , Maze Learning , Mice , Mice, Inbred C57BL , Myelin Basic Protein/metabolism , Neurofilament Proteins/metabolism , Neurons/metabolism , Severity of Illness Index
17.
Sci Adv ; 2(11): e1600874, 2016 Nov.
Article in English | MEDLINE | ID: mdl-28138519

ABSTRACT

The repair of injured tendons remains a formidable clinical challenge because of our limited understanding of tendon stem cells and the regulation of tenogenesis. With single-cell analysis to characterize the gene expression profiles of individual cells isolated from tendon tissue, a subpopulation of nestin+ tendon stem/progenitor cells (TSPCs) was identified within the tendon cell population. Using Gene Expression Omnibus datasets and immunofluorescence assays, we found that nestin expression was activated at specific stages of tendon development. Moreover, isolated nestin+ TSPCs exhibited superior tenogenic capacity compared to nestin- TSPCs. Knockdown of nestin expression in TSPCs suppressed their clonogenic capacity and reduced their tenogenic potential significantly both in vitro and in vivo. Hence, these findings provide new insights into the identification of subpopulations of TSPCs and illustrate the crucial roles of nestin in TSPC fate decisions and phenotype maintenance, which may assist in future therapeutic strategies to treat tendon disease.


Subject(s)
Databases, Nucleic Acid , Gene Expression Regulation/physiology , Nestin/metabolism , Stem Cells/metabolism , Tendons/metabolism , Animals , Mice , Mice, Transgenic , Nestin/genetics , Stem Cells/cytology , Tendons/cytology
18.
Exp Neurol ; 272: 17-25, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25819104

ABSTRACT

Previous research on stroke and traumatic brain injury (TBI) heavily emphasized pathological alterations in neuronal cells within gray matter. However, recent studies have highlighted the equal importance of white matter integrity in long-term recovery from these conditions. Demyelination is a major component of white matter injury and is characterized by loss of the myelin sheath and oligodendrocyte cell death. Demyelination contributes significantly to long-term sensorimotor and cognitive deficits because the adult brain only has limited capacity for oligodendrocyte regeneration and axonal remyelination. In the current review, we will provide an overview of the major causes of demyelination and oligodendrocyte cell death following acute brain injuries, and discuss the crosstalk between myelin, axons, microglia, and astrocytes during the process of demyelination. Recent discoveries of molecules that regulate the processes of remyelination may provide novel therapeutic targets to restore white matter integrity and improve long-term neurological recovery in stroke or TBI patients.


Subject(s)
Brain Injuries/complications , Brain Injuries/therapy , Demyelinating Diseases/etiology , Ischemia/complications , Ischemia/therapy , Nerve Regeneration/physiology , Animals , Axons/pathology , Cell Death , Cognition Disorders/etiology , Cytokines/metabolism , Demyelinating Diseases/complications , Humans , Nerve Regeneration/drug effects , Oligodendroglia/pathology
19.
Prog Neurobiol ; 119-120: 60-84, 2014.
Article in English | MEDLINE | ID: mdl-24923657

ABSTRACT

Microglia are the first line of immune defense against central nervous system (CNS) injuries and disorders. These highly plastic cells play dualistic roles in neuronal injury and recovery and are known for their ability to assume diverse phenotypes. A broad range of surface receptors are expressed on microglia and mediate microglial 'On' or 'Off' responses to signals from other host cells as well as invading microorganisms. The integrated actions of these receptors result in tightly regulated biological functions, including cell mobility, phagocytosis, the induction of acquired immunity, and trophic factor/inflammatory mediator release. Over the last few years, significant advances have been made toward deciphering the signaling mechanisms related to these receptors and their specific cellular functions. In this review, we describe the current state of knowledge of the surface receptors involved in microglial activation, with an emphasis on their engagement of distinct functional programs and their roles in CNS injuries. It will become evident from this review that microglial homeostasis is carefully maintained by multiple counterbalanced strategies, including, but not limited to, 'On' and 'Off' receptor signaling. Specific regulation of theses microglial receptors may be a promising therapeutic strategy against CNS injuries.


Subject(s)
Central Nervous System/immunology , Microglia/physiology , Trauma, Nervous System/immunology , Animals , Humans , Receptors, Cell Surface/metabolism , Receptors, Chemokine/metabolism , Receptors, Pattern Recognition/metabolism , Receptors, Purinergic/metabolism
20.
Prog Neurobiol ; 115: 6-24, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24374228

ABSTRACT

Immune and inflammatory responses actively modulate the pathophysiological processes of acute brain injuries such as stroke. Soon after the onset of stroke, signals such as brain-derived antigens, danger-associated molecular patterns (DAMPs), cytokines, and chemokines are released from the injured brain into the systemic circulation. The injured brain also communicates with peripheral organs through the parasympathetic and sympathetic branches of the autonomic nervous system. Many of these diverse signals not only activate resident immune cells in the brain, but also trigger robust immune responses in the periphery. Peripheral immune cells then migrate toward the site of injury and release additional cytokines, chemokines, and other molecules, causing further disruptive or protective effects in the ischemic brain. Bidirectional communication between the injured brain and the peripheral immune system is now known to regulate the progression of stroke pathology as well as tissue repair. In the end, this exquisitely coordinated crosstalk helps determine the fate of animals after stroke. This article reviews the literature on ischemic brain-derived signals through which peripheral immune responses are triggered, and the potential impact of these peripheral responses on brain injury and repair. Pharmacological strategies and cell-based therapies that target the dialog between the brain and peripheral immune system show promise as potential novel treatments for stroke.


Subject(s)
Autonomic Nervous System/immunology , Brain Ischemia/complications , Brain Ischemia/diagnosis , Immune System/immunology , Inflammation/etiology , Animals , Autonomic Nervous System/physiopathology , Brain/pathology , Brain Ischemia/immunology , Cytokines/metabolism , Humans , Immune System/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...