Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Nature ; 611(7936): 585-593, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36352225

ABSTRACT

Macrophages are important players in the maintenance of tissue homeostasis1. Perivascular and leptomeningeal macrophages reside near the central nervous system (CNS) parenchyma2, and their role in CNS physiology has not been sufficiently well studied. Given their continuous interaction with the cerebrospinal fluid (CSF) and strategic positioning, we refer to these cells collectively as parenchymal border macrophages (PBMs). Here we demonstrate that PBMs regulate CSF flow dynamics. We identify a subpopulation of PBMs that express high levels of CD163 and LYVE1 (scavenger receptor proteins), closely associated with the brain arterial tree, and show that LYVE1+ PBMs regulate arterial motion that drives CSF flow. Pharmacological or genetic depletion of PBMs led to accumulation of extracellular matrix proteins, obstructing CSF access to perivascular spaces and impairing CNS perfusion and clearance. Ageing-associated alterations in PBMs and impairment of CSF dynamics were restored after intracisternal injection of macrophage colony-stimulating factor. Single-nucleus RNA sequencing data obtained from patients with Alzheimer's disease (AD) and from non-AD individuals point to changes in phagocytosis, endocytosis and interferon-γ signalling on PBMs, pathways that are corroborated in a mouse model of AD. Collectively, our results identify PBMs as new cellular regulators of CSF flow dynamics, which could be targeted pharmacologically to alleviate brain clearance deficits associated with ageing and AD.


Subject(s)
Central Nervous System , Cerebrospinal Fluid , Macrophages , Parenchymal Tissue , Animals , Mice , Alzheimer Disease/metabolism , Brain/metabolism , Central Nervous System/cytology , Central Nervous System/metabolism , Cerebrospinal Fluid/metabolism , Macrophages/physiology , Meninges/cytology , Rheology , Extracellular Matrix Proteins/metabolism , Aging/metabolism , Phagocytosis , Endocytosis , Interferon-gamma/metabolism , Parenchymal Tissue/cytology , Humans
2.
Biomaterials ; 269: 120611, 2021 02.
Article in English | MEDLINE | ID: mdl-33385685

ABSTRACT

Bioprinting is an emergent technology that has already demonstrated the capacity to create complex and/or vascularized multicellular structures with defined and organized architectures, in a reproducible and high throughput way. Here, we present the implementation of a complex liver model by the development of a three-dimensional extrusion bioprinting process, including parameters for matrix polymerization of methacrylated gelatin, using two hepatic cell lines, Huh7 and HepaRG. The printed structures exhibited long-term viability (28 days), proliferative ability, a relevant hepatocyte phenotype and functions equivalent to or better than those of their 2D counterparts using standard DMSO treatment. This work served as a basis for the bioprinting of complex multicellular models associating the hepatic parenchymal cells, HepaRG, with stellate cells (LX-2) and endothelial cells (HUVECs), able of colonizing the surface of the structure and thus recreating a pseudo endothelial barrier. When bioprinted in 3D monocultures, LX-2 expression was modulated by TGFß-1 toward the induction of myofibroblastic genes such as ACTA2 and COL1A1. In 3D multicellular bioprinted structures comprising HepaRG, LX-2 and endothelial cells, we evidenced fibrillar collagen deposition, which is never observed in monocultures of either HepaRG or LX-2 alone. These observations indicate that a precise control of cellular communication is required to recapitulate key steps of fibrogenesis. Bioprinted 3D co-cultures therefore open up new perspectives in studying the molecular and cellular basis of fibrosis development and provide better access to potential inducers and inhibitors of collagen expression and deposition.


Subject(s)
Bioprinting , Liver/cytology , Printing, Three-Dimensional , Tissue Engineering , Cell Culture Techniques , Cell Line , Endothelial Cells , Gelatin , Hepatic Stellate Cells , Humans , Parenchymal Tissue/cytology , Tissue Scaffolds
3.
J Comp Neurol ; 529(3): 553-575, 2021 02.
Article in English | MEDLINE | ID: mdl-32515035

ABSTRACT

Tanycytes are highly specialized ependymal cells that line the bottom and the lateral walls of the third ventricle. In contact with the cerebrospinal fluid through their cell bodies, they send processes into the arcuate nucleus, the ventromedial nucleus, and the dorsomedial nucleus of the hypothalamus. In the present work, we combined transgenic and immunohistochemical approaches to investigate the neuroanatomical associations between tanycytes and neural cells present in the hypothalamic parenchyma, in particular in the arcuate nucleus. The specific expression of tdTomato in tanycytes first allowed the observation of peculiar subcellular protrusions along tanycyte processes and at their endfeet such as spines, swelling, en passant boutons, boutons, or claws. Interestingly, these protrusions contact different neural cells in the brain parenchyma including blood vessels and neurons, and in particular NPY and POMC neurons in the arcuate nucleus. Using both fluorescent and electron microscopy, we finally observed that these tanycyte protrusions contain ribosomes, mitochondria, diverse vesicles, and transporters, suggesting dense tanycyte/neuron and tanycyte/blood vessel communications. Altogether, our results lay the neuroanatomical basis for tanycyte/neural cell interactions, which will be useful to further understand cell-to-cell communications involved in the regulation of neuroendocrine functions.


Subject(s)
Ependymoglial Cells/ultrastructure , Hypothalamus/ultrastructure , Neurons/ultrastructure , Parenchymal Tissue/ultrastructure , Animals , Ependymoglial Cells/chemistry , Guinea Pigs , Humans , Hypothalamus/chemistry , Hypothalamus/cytology , Male , Mice , Mice, Transgenic , Neurons/chemistry , Parenchymal Tissue/chemistry , Parenchymal Tissue/cytology , Rabbits
4.
Clin Epigenetics ; 12(1): 145, 2020 10 02.
Article in English | MEDLINE | ID: mdl-33008450

ABSTRACT

BACKGROUND: Mesenchymal fibroblasts are ubiquitous cells that maintain the extracellular matrix of organs. Within the lung, airway and parenchymal fibroblasts are crucial for lung development and are altered with disease, but it has been difficult to understand their roles due to the lack of distinct molecular markers. We studied genome-wide DNA methylation and gene expression in airway and parenchymal lung fibroblasts from healthy and asthmatic donors, to identify a robust cell marker and to determine if these cells are molecularly distinct in asthma. RESULTS: Airway (N = 8) and parenchymal (N = 15) lung fibroblasts from healthy individuals differed in the expression of 158 genes, and DNA methylation of 3936 CpGs (Bonferroni adjusted p value < 0.05). Differential DNA methylation between cell types was associated with differential expression of 42 genes, but no single DNA methylation CpG feature (location, effect size, number) defined the interaction. Replication of gene expression and DNA methylation in a second cohort identified TWIST1 gene expression, DNA methylation and protein expression as a cell marker of airway and parenchymal lung fibroblasts, with DNA methylation having 100% predictive discriminatory power. DNA methylation was differentially altered in parenchymal (112 regions) and airway fibroblasts (17 regions) with asthmatic status, with no overlap between regions. CONCLUSIONS: Differential methylation of TWIST1 is a robust cell marker of airway and parenchymal lung fibroblasts. Airway and parenchymal fibroblast DNA methylation are differentially altered in individuals with asthma, and the role of both cell types should be considered in the pathogenesis of asthma.


Subject(s)
Asthma/genetics , DNA Methylation/genetics , Fibroblasts/metabolism , Nuclear Proteins/metabolism , Parenchymal Tissue/cytology , Twist-Related Protein 1/metabolism , Aged , Airway Remodeling/genetics , Asthma/pathology , Biomarkers/metabolism , Case-Control Studies , CpG Islands/genetics , Female , Gene Expression , Genome-Wide Association Study/methods , Humans , Lung/pathology , Male , Middle Aged , Predictive Value of Tests
5.
Stem Cell Res Ther ; 11(1): 448, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097094

ABSTRACT

Gene therapy is being investigated for a range of serious lung diseases, such as cystic fibrosis and emphysema. Recombinant adeno-associated virus (rAAV) is a well-established, safe, viral vector for gene delivery with multiple naturally occurring and artificial serotypes available displaying alternate cell, tissue, and species-specific tropisms. Efficient AAV serotypes for the transduction of the conducting airways have been identified for several species; however, efficient serotypes for human lung parenchyma have not yet been identified. Here, we screened the ability of multiple AAV serotypes to transduce lung bud organoids (LBOs)-a model of human lung parenchyma generated from human embryonic stem cells. Microinjection of LBOs allowed us to model transduction from the luminal surface, similar to dosing via vector inhalation. We identified the naturally occurring rAAV2 and rAAV6 serotypes, along with synthetic rAAV6 variants, as having tropism for the human lung parenchyma. Positive staining of LBOs for surfactant proteins B and C confirmed distal lung identity and suggested the suitability of these vectors for the transduction of alveolar type II cells. Our findings establish LBOs as a new model for pulmonary gene therapy and stress the relevance of LBOs as a viral infection model of the lung parenchyma as relevant in SARS-CoV-2 research.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Human Embryonic Stem Cells/cytology , Lung Diseases/therapy , Organoids/cytology , Cell Line , Dependovirus/immunology , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Lung/metabolism , Models, Biological , Parenchymal Tissue/cytology
6.
Immunology ; 161(1): 4-17, 2020 09.
Article in English | MEDLINE | ID: mdl-32463116

ABSTRACT

Foxp3+  CD4+ regulatory T cells (Tregs) are an immune cell lineage endowed with immunosuppressive functionality in a wide array of contexts, including both anti-pathogenic and anti-self responses. In the past decades, our understanding of the functional diversity of circulating or lymphoid Tregs has grown exponentially. Only recently, the importance of Tregs residing within non-lymphoid tissues, such as visceral adipose tissue, muscle, skin and intestine, has been recognized. Not only are Tregs critical for influencing the kinetics and strength of immune responses, but the regulation of non-immune or parenchymal cells, also fall within the purview of tissue-resident or infiltrating Tregs. This review focuses on providing a systematic and comprehensive comparison of the molecular maintenance, local adaptation and functional specializations of Treg populations operating within different tissues.


Subject(s)
Immune Tolerance/immunology , Intestines/immunology , Intra-Abdominal Fat/immunology , Muscles/immunology , Skin/immunology , T-Lymphocytes, Regulatory/immunology , Autoimmunity/immunology , Humans , Inflammation/immunology , Inflammation/prevention & control , Intestines/cytology , Intra-Abdominal Fat/cytology , Muscles/cytology , Parenchymal Tissue/cytology , Parenchymal Tissue/immunology , Skin/cytology
7.
Nature ; 578(7796): 610-614, 2020 02.
Article in English | MEDLINE | ID: mdl-32076265

ABSTRACT

The sympathetic nervous system innervates peripheral organs to regulate their function and maintain homeostasis, whereas target cells also produce neurotrophic factors to promote sympathetic innervation1,2. The molecular basis of this bi-directional communication remains to be fully determined. Here we use thermogenic adipose tissue from mice as a model system to show that T cells, specifically γδ T cells, have a crucial role in promoting sympathetic innervation, at least in part by driving the expression of TGFß1 in parenchymal cells via the IL-17 receptor C (IL-17RC). Ablation of IL-17RC specifically in adipose tissue reduces expression of TGFß1 in adipocytes, impairs local sympathetic innervation and causes obesity and other metabolic phenotypes that are consistent with defective thermogenesis; innervation can be fully rescued by restoring TGFß1 expression. Ablating γδ Τ cells and the IL-17RC signalling pathway also impairs sympathetic innervation in other tissues such as salivary glands. These findings demonstrate coordination between T cells and parenchymal cells to regulate sympathetic innervation.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/innervation , Adipose Tissue/metabolism , Interleukin-17/metabolism , Sympathetic Nervous System/physiology , T-Lymphocytes/metabolism , Thermogenesis , Adipose Tissue, Brown/metabolism , Animals , Interleukin-17/deficiency , Interleukin-17/genetics , Male , Mice , Mice, Knockout , Organ Specificity , Parenchymal Tissue/cytology , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
8.
J Gastroenterol Hepatol ; 35(6): 960-966, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31782974

ABSTRACT

Performance and advances in liver surgery makes remarkable progress of the understanding of liver regeneration. Liver regeneration after liver resection has been widely researched, and the underlying mechanism mostly concerns proliferation of hepatocytes and the influence by inflammation through activation of Kupffer cells and the other parenchymal cells, the second regenerative pathway by hepatic progenitor cells (HPCs), inducing angiogenesis, remodeling of a extracellular matrix (ECM), and termination mechanisms. New clinical surgeries and the updated multiomics analysis are exploiting the remarkable progress, especially in immune regulation and metabolic process of two emerging hallmarks. This review briefly represents a systemic outline of eight hallmarks, including hepatocyte proliferation, contribution of hepatic progenitor cells, inducing angiogenesis, reprogramming of the extracellular matrix, apoptosis and termination of proliferation, inflammation, immune and metabolic regulation, which are set as organizing characteristics of postoperative liver regeneration and future directions of refining treatment targets.


Subject(s)
Digestive System Surgical Procedures , Liver Regeneration/physiology , Liver/surgery , Cell Proliferation , Extracellular Matrix/metabolism , Hepatocytes/physiology , Humans , Kupffer Cells/physiology , Liver/cytology , Liver/metabolism , Liver/physiopathology , Neovascularization, Physiologic , Parenchymal Tissue/cytology , Parenchymal Tissue/physiology , Postoperative Period , Stem Cells/physiology
9.
Stem Cells Transl Med ; 9(2): 177-188, 2020 02.
Article in English | MEDLINE | ID: mdl-31800978

ABSTRACT

Neural precursor cells (NSCs) hold great potential to treat a variety of neurodegenerative diseases and injuries to the spinal cord. However, current delivery techniques require an invasive approach in which an injection needle is advanced into the spinal parenchyma to deliver cells of interest. As such, this approach is associated with an inherent risk of spinal injury, as well as a limited delivery of cells into multiple spinal segments. Here, we characterize the use of a novel cell delivery technique that employs single bolus cell injections into the spinal subpial space. In immunodeficient rats, two subpial injections of human NSCs were performed in the cervical and lumbar spinal cord, respectively. The survival, distribution, and phenotype of transplanted cells were assessed 6-8 months after injection. Immunofluorescence staining and mRNA sequencing analysis demonstrated a near-complete occupation of the spinal cord by injected cells, in which transplanted human NSCs (hNSCs) preferentially acquired glial phenotypes, expressing oligodendrocyte (Olig2, APC) or astrocyte (GFAP) markers. In the outermost layer of the spinal cord, injected hNSCs differentiated into glia limitans-forming astrocytes and expressed human-specific superoxide dismutase and laminin. All animals showed normal neurological function for the duration of the analysis. These data show that the subpial cell delivery technique is highly effective in populating the entire spinal cord with injected NSCs, and has a potential for clinical use in cell replacement therapies for the treatment of ALS, multiple sclerosis, or spinal cord injury.


Subject(s)
Neural Stem Cells/metabolism , Parenchymal Tissue/metabolism , Animals , Parenchymal Tissue/cytology , Rats , Rats, Sprague-Dawley
10.
Nat Rev Cancer ; 19(11): 605, 2019 11.
Article in English | MEDLINE | ID: mdl-31548603
11.
Nature ; 572(7771): 589-590, 2019 08.
Article in English | MEDLINE | ID: mdl-31462800
12.
Nat Mater ; 18(6): 627-637, 2019 06.
Article in English | MEDLINE | ID: mdl-31114073

ABSTRACT

Cells are transplanted to regenerate an organs' parenchyma, but how transplanted parenchymal cells induce stromal regeneration is elusive. Despite the common use of a decellularized matrix, little is known as to the pivotal signals that must be restored for tissue or organ regeneration. We report that Alx3, a developmentally important gene, orchestrated adult parenchymal and stromal regeneration by directly transactivating Wnt3a and vascular endothelial growth factor. In contrast to the modest parenchyma formed by native adult progenitors, Alx3-restored cells in decellularized scaffolds not only produced vascularized stroma that involved vascular endothelial growth factor signalling, but also parenchymal dentin via the Wnt/ß-catenin pathway. In an orthotopic large-animal model following parenchyma and stroma ablation, Wnt3a-recruited endogenous cells regenerated neurovascular stroma and differentiated into parenchymal odontoblast-like cells that extended the processes into newly formed dentin with a structure-mechanical equivalency to native dentin. Thus, the Alx3-Wnt3a axis enables postnatal progenitors with a modest innate regenerative capacity to regenerate adult tissues. Depleted signals in the decellularized matrix may be reinstated by a developmentally pivotal gene or corresponding protein.


Subject(s)
Homeodomain Proteins/metabolism , Parenchymal Tissue/physiology , Tooth/cytology , Tooth/embryology , Adolescent , Animals , Female , Homeodomain Proteins/genetics , Humans , Incisor/cytology , Incisor/embryology , Mice, Inbred Strains , Molar, Third/cytology , Organ Culture Techniques , Parenchymal Tissue/cytology , Pregnancy , Promoter Regions, Genetic , Regeneration , Stromal Cells/physiology , Swine , Vascular Endothelial Growth Factor A/genetics , Wnt3A Protein/genetics , Wnt3A Protein/metabolism
13.
Cell Rep ; 27(4): 1073-1089.e5, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31018125

ABSTRACT

Oligodendrocyte progenitor cells (OPCs) are widely distributed cells of ramified morphology in adult brain that express PDGFRα and NG2. They retain mitotic activities in adulthood and contribute to oligodendrogenesis and myelin turnover; however, the regulatory mechanisms of their cell dynamics in adult brain largely remain unknown. Here, we found that global Pdgfra inactivation in adult mice rapidly led to elimination of OPCs due to synchronous maturation toward oligodendrocytes. Surprisingly, OPC densities were robustly reconstituted by the active expansion of Nestin+ immature cells activated in meninges and brain parenchyma, as well as a few OPCs that escaped from Pdgfra inactivation. The multipotent immature cells were induced in the meninges of Pdgfra-inactivated mice, but not of control mice. Our findings revealed powerful homeostatic control of adult OPCs, engaging dual cellular sources of adult OPC formation. These properties of the adult oligodendrocyte lineage and the alternative OPC source may be exploited in regenerative medicine.


Subject(s)
Brain/cytology , Oligodendrocyte Precursor Cells/cytology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Animals , Brain/metabolism , Cell Differentiation , Cell Lineage , Homeostasis , Meninges/cytology , Meninges/metabolism , Mice , Nestin/metabolism , Oligodendrocyte Precursor Cells/metabolism , Parenchymal Tissue/cytology
15.
Int J Mol Sci ; 19(12)2018 Dec 18.
Article in English | MEDLINE | ID: mdl-30567401

ABSTRACT

Liver regeneration is crucial for the maintenance of liver functional mass during homeostasis and diseases. In a disease context-dependent manner, liver regeneration is contributed to by hepatocytes or progenitor cells. As long as they are replicatively competent, hepatocytes are the main cell type responsible for supporting liver size homeostasisand regeneration. The concept that all hepatocytes within the lobule have the same proliferative capacity but are differentially recruited according to the localization of the wound, or whether a yet to be defined sub-population of hepatocytes supports regeneration is still debated. In a chronically or severely injured liver, hepatocytes may enter a state of replicative senescence. In such conditions, small biliary cells activate and expand, a process called ductular reaction (DR). Work in the last few decades has demonstrated that DR cells can differentiate into hepatocytes and thereby contribute to parenchymal reconstitution. In this study we will review the molecular mechanisms supporting these two processes to determine potential targets that would be amenable for therapeutic manipulation to enhance liver regeneration.


Subject(s)
Cell Differentiation/genetics , Liver Regeneration/genetics , Liver/growth & development , Stem Cells , Animals , Cell Lineage/genetics , Cell Lineage/physiology , Cellular Microenvironment/genetics , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Liver Regeneration/physiology , Parenchymal Tissue/cytology , Parenchymal Tissue/physiology
16.
Vaccine ; 36(37): 5625-5635, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30097220

ABSTRACT

Tuberculosis (TB) is the biggest cause of human mortality from an infectious disease. The only vaccine currently available, bacille Calmette-Guérin (BCG), demonstrates some protection against disseminated disease in childhood but very variable efficacy against pulmonary disease in adults. A greater understanding of protective host immune responses is required in order to aid the development of improved vaccines. Tissue-resident memory T cells (TRM) are a recently-identified subset of T cells which may represent an important component of protective immunity to TB. Here, we demonstrate that intradermal BCG vaccination induces a population of antigen-specific CD4+ T cells within the lung parenchyma which persist for >12 months post-vaccination. Comprehensive flow cytometric analysis reveals this population is phenotypically and functionally heterogeneous, and shares characteristics with lung vascular and splenic CD4+ T cells. This underlines the importance of utilising the intravascular staining technique for definitive identification of tissue-resident T cells, and also suggests that these anatomically distinct cellular subsets are not necessarily permanently resident within a particular tissue compartment but can migrate between compartments. This lung parenchymal population merits further investigation as a critical component of a protective immune response against Mycobacterium tuberculosis (M. tb).


Subject(s)
BCG Vaccine/administration & dosage , CD4-Positive T-Lymphocytes/immunology , Lung/immunology , Parenchymal Tissue/immunology , Tuberculosis, Pulmonary/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Female , Immunogenicity, Vaccine , Immunologic Memory , Interferon-gamma , Lung/cytology , Mice , Mice, Inbred BALB C , Parenchymal Tissue/cytology
17.
Clin Epigenetics ; 10: 32, 2018.
Article in English | MEDLINE | ID: mdl-29527240

ABSTRACT

Background: Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease of the lungs that is currently the fourth leading cause of death worldwide. Genetic factors account for only a small amount of COPD risk, but epigenetic mechanisms, including DNA methylation, have the potential to mediate the interactions between an individual's genetics and environmental exposure. DNA methylation is highly cell type-specific, and individual cell type studies of DNA methylation in COPD are sparse. Fibroblasts are present within the airway and parenchyma of the lung and contribute to the aberrant deposition of extracellular matrix in COPD. No assessment or comparison of genome-wide DNA methylation profiles in the airway and parenchymal fibroblasts from individuals with and without COPD has been undertaken. These data provide valuable insight into the molecular mechanisms contributing to COPD and the differing pathologies of small airways disease and emphysema in COPD. Methods: Genome-wide DNA methylation was evaluated at over 485,000 CpG sites using the Illumina Infinium HumanMethylation450 BeadChip array in the airway (non-COPD n = 8, COPD n = 7) and parenchymal fibroblasts (non-COPD n = 17, COPD n = 29) isolated from individuals with and without COPD. Targeted gene expression was assessed by qPCR in matched RNA samples. Results: Differentially methylated DNA regions were identified between cells isolated from individuals with and without COPD in both airway and parenchymal fibroblasts. Only in parenchymal fibroblasts was differential DNA methylation associated with differential gene expression. A second analysis of differential DNA methylation variability identified 359 individual differentially variable CpG sites in parenchymal fibroblasts. No differentially variable CpG sites were identified in the airway fibroblasts. Five differentially variable-methylated CpG sites, associated with three genes, were subsequently assessed for gene expression differences. Two genes (OAT and GRIK2) displayed significantly increased gene expression in cells isolated from individuals with COPD. Conclusions: Differential and variable DNA methylation was associated with COPD status in the parenchymal fibroblasts but not airway fibroblasts. Aberrant DNA methylation was associated with altered gene expression imparting biological function to DNA methylation changes. Changes in DNA methylation are therefore implicated in the molecular mechanisms underlying COPD pathogenesis and may represent novel therapeutic targets.


Subject(s)
DNA Methylation , Lung/chemistry , Ornithine-Oxo-Acid Transaminase/genetics , Parenchymal Tissue/chemistry , Pulmonary Disease, Chronic Obstructive/genetics , Receptors, Kainic Acid/genetics , Up-Regulation , Aged , Cells, Cultured , CpG Islands , Epigenesis, Genetic , Female , Fibroblasts/chemistry , Gene Expression Profiling/methods , Gene Expression Regulation , Humans , Lung/cytology , Male , Middle Aged , Organ Specificity , Parenchymal Tissue/cytology , Sequence Analysis, DNA , GluK2 Kainate Receptor
18.
Front Immunol ; 9: 44, 2018.
Article in English | MEDLINE | ID: mdl-29422896

ABSTRACT

Toxic substances and microbial or food-derived antigens continuously challenge the liver, which is tasked with their safe neutralization. This vital organ is also important for the removal of apoptotic immune cells during inflammation and has been previously described as a "graveyard" for dying lymphocytes. The clearance of apoptotic and necrotic cells is known as efferocytosis and is a critical liver function to maintain tissue homeostasis. Much of the research into this form of immunological control has focused on Kupffer cells, the liver-resident macrophages. However, hepatocytes (and other liver resident cells) are competent efferocytes and comprise 80% of the liver mass. Little is known regarding the mechanisms of apoptotic and necrotic cell capture by epithelia, which lack key receptors that mediate phagocytosis in macrophages. Herein, we discuss recent developments that increased our understanding of efferocytosis in tissues, with a special focus on the liver parenchyma. We discuss the impact of efferocytosis in health and in inflammation, highlighting the role of phagocytic epithelia.


Subject(s)
Epithelial Cells/metabolism , Kupffer Cells/immunology , Liver/metabolism , Parenchymal Tissue/metabolism , Phagocytosis/immunology , Apoptosis/immunology , Epithelium/metabolism , Hepatocytes/metabolism , Humans , Liver/cytology , Necrosis/immunology , Parenchymal Tissue/cytology
19.
Planta ; 247(5): 1067-1076, 2018 May.
Article in English | MEDLINE | ID: mdl-29344723

ABSTRACT

MAIN CONCLUSION: The close association between membranes and organelles, and the intense chloroplast remodeling in parenchyma cells of extrafloral nectaries occurred only at the secretion time and suggest a relationship with the nectar secretion. Associations between membranes and organelles have been well documented in different tissues and cells of plants, but poorly explored in secretory cells. Here, we described the close physical juxtaposition between membranes and organelles, mainly with chloroplasts, in parenchyma cells of Citharexylum myrianthum (Verbenaeceae) extrafloral nectaries under transmission electron microscopy, using conventional and microwave fixation. At the time of nectar secretion, nectary parenchyma cells exhibit a multitude of different organelle and membrane associations as mitochondria-mitochondria, mitochondria-endoplasmic reticulum, mitochondria-chloroplast, chloroplast-nuclear envelope, mitochondria-nuclear envelope, chloroplast-plasmalemma, chloroplast-chloroplast, chloroplast-tonoplast, chloroplast-peroxisome, and mitochondria-peroxisome. These associations were visualized as amorphous electron-dense material, a network of dense fibrillar material and/or dense bridges. Chloroplasts exhibited protrusions variable in shape and extension, which bring them closer to each other and to plasmalemma, tonoplast, and nuclear envelope. Parenchyma cells in the pre- and post-secretory stages did not exhibit any association or juxtaposition of membranes and organelles, and chloroplast protrusions were absent. Chloroplasts had peripheral reticulum that was more developed in the secretory stage. We propose that such subcellular phenomena during the time of nectar secretion optimize the movement of signaling molecules and the exchange of metabolites. Our results open new avenues on the potential mechanisms of organelle contact in parenchyma nectary cells, and reveal new attributes of the secretory cells on the subcellular level.


Subject(s)
Intracellular Membranes/ultrastructure , Organelles/ultrastructure , Parenchymal Tissue/cytology , Plant Structures/ultrastructure , Verbenaceae/ultrastructure , Chloroplasts/ultrastructure , Microscopy, Electron, Transmission , Parenchymal Tissue/ultrastructure , Plant Nectar/metabolism , Plant Structures/cytology , Tissue Fixation/methods
20.
Cell Physiol Biochem ; 44(4): 1295-1310, 2017.
Article in English | MEDLINE | ID: mdl-29183009

ABSTRACT

BACKGROUND/AIMS: Radiation therapy is an important treatment for thoracic cancer; however, side effects accompanied with radiotherapy lead to limited tumor control and a decline in patient quality of life. Among these side effects, radiation-induced lung injury (RILI) is the most serious and common. Hence, an effective remedy for RILI is needed. Mesenchymal stromal cells (MSCs) are multipotent adult stem cells that have been demonstrated to be an effective treatment in some disease caused by tissue damage. However, unlike other injuries, RILI received limited therapeutic effects from implanted MSCs due to local hypoxia and extensive reactive oxygen species (ROS) in irradiated lungs. Since the poor survival of MSCs is primarily due to hypoxia and ROS generation, we hypothesize that persistent and adaptive hypoxia treatment induces enhanced resistance to hypoxic stress in implanted MSC. The aim of this study is to investigate whether persistent and adaptive hypoxia treatment of bmMSCs prior to their transplantation in injured mice enhanced survival and improved curative effects in RILI. METHODS: Primary bmMSCs were obtained from the marrow of six-week-old male C57BL6/J mice and were cultured either under normoxic conditions (21% O2) or hypoxic conditions (2.5% O2). Mice were injected with normoxia/hypoxia MSCs after thoracic irradiation (20 Gy). The therapeutic effects of MSCs on RILI were assessed by pathological examinations that included H&E staining, Masson staining and α-SMA staining; meanwhile, inflammatory factors were measured using an ELISA. The morphology of MSCs in vitro was recorded using a microscope and identified by flow cytometry, cell viability was measured using the CCK-8 assay, the potential for proliferation was detected by the EdU assay, and ROS levels were measured using a ROS fluorogenic probe. In addition, HIF-1α and several survival pathway proteins (Akt, p-Akt, Caspase-3) were also detected by western blotting. RESULTS: Implanted MSCs alleviated both early radiation-induced pneumonia and late pulmonary fibrosis. However, hypoxia MSCs displayed a more pronounced therapeutic effect compared to normoxia MSCs. Compared to normoxia MSCs, the hypoxia MSCs demonstrated greater cell viability, an enhanced proliferation potential, decreased ROS levels and increased resistance to hypoxia and ROS stress. In addition, hypoxia MSCs achieved higher activation levels of HIF-1α and Akt, and HIF-1α played a critical role in the development of resistance. CONCLUSION: Hypoxia enhances the therapeutic effect of mesenchymal stromal cells on radiation-induced lung injury by promoting MSC proliferation and improving their antioxidant ability, mediated by HIF-1α.


Subject(s)
Antioxidants/metabolism , Cell Hypoxia , Lung Injury/therapy , Mesenchymal Stem Cell Transplantation , Actins/genetics , Actins/metabolism , Animals , Apoptosis/radiation effects , Bone Marrow Cells/cytology , Caspase 3/metabolism , Cell Proliferation , Cell Survival , Cells, Cultured , Gamma Rays , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Injury/pathology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Parenchymal Tissue/cytology , Parenchymal Tissue/metabolism , Parenchymal Tissue/radiation effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta/analysis , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...