Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2367, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38531868

ABSTRACT

The development of craniofacial skeletal structures is fascinatingly complex and elucidation of the underlying mechanisms will not only provide novel scientific insights, but also help develop more effective clinical approaches to the treatment and/or prevention of the numerous congenital craniofacial malformations. To this end, we performed a genome-wide analysis of RNA transcription from non-coding regulatory elements by CAGE-sequencing of the facial mesenchyme of human embryos and cross-checked the active enhancers thus identified against genes, identified by GWAS for the normal range human facial appearance. Among the identified active cis-enhancers, several belonged to the components of the PI3/AKT/mTORC1/autophagy pathway. To assess the functional role of this pathway, we manipulated it both genetically and pharmacologically in mice and zebrafish. These experiments revealed that mTORC1 signaling modulates craniofacial shaping at the stage of skeletal mesenchymal condensations, with subsequent fine-tuning during clonal intercalation. This ability of mTORC1 pathway to modulate facial shaping, along with its evolutionary conservation and ability to sense external stimuli, in particular dietary amino acids, indicate that the mTORC1 pathway may play a role in facial phenotypic plasticity. Indeed, the level of protein in the diet of pregnant female mice influenced the activity of mTORC1 in fetal craniofacial structures and altered the size of skeletogenic clones, thus exerting an impact on the local geometry and craniofacial shaping. Overall, our findings indicate that the mTORC1 signaling pathway is involved in the effect of environmental conditions on the shaping of craniofacial structures.


Subject(s)
Signal Transduction , Zebrafish , Pregnancy , Mice , Animals , Female , Humans , Proteins , Mechanistic Target of Rapamycin Complex 1 , Diet
2.
Nat Commun ; 14(1): 509, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36720873

ABSTRACT

Spatially resolved transcriptomics has enabled precise genome-wide mRNA expression profiling within tissue sections. The performance of methods targeting the polyA tails of mRNA relies on the availability of specimens with high RNA quality. Moreover, the high cost of currently available spatial resolved transcriptomics assays requires a careful sample screening process to increase the chance of obtaining high-quality data. Indeed, the upfront analysis of RNA quality can show considerable variability due to sample handling, storage, and/or intrinsic factors. We present RNA-Rescue Spatial Transcriptomics (RRST), a workflow designed to improve mRNA recovery from fresh frozen specimens with moderate to low RNA quality. First, we provide a benchmark of RRST against the standard Visium spatial gene expression protocol on high RNA quality samples represented by mouse brain and prostate cancer samples. Then, we test the RRST protocol on tissue sections collected from five challenging tissue types, including human lung, colon, small intestine, pediatric brain tumor, and mouse bone/cartilage. In total, we analyze 52 tissue sections and demonstrate that RRST is a versatile, powerful, and reproducible protocol for fresh frozen specimens of different qualities and origins.


Subject(s)
RNA , Transcriptome , Child , Male , Humans , Animals , Mice , Transcriptome/genetics , RNA, Messenger , Benchmarking , Biological Assay
3.
Nat Commun ; 13(1): 6949, 2022 11 14.
Article in English | MEDLINE | ID: mdl-36376278

ABSTRACT

There are major differences in duration and scale at which limb development and regeneration proceed, raising the question to what extent regeneration is a recapitulation of development. We address this by analyzing skeletal elements using a combination of micro-CT imaging, molecular profiling and clonal cell tracing. We find that, in contrast to development, regenerative skeletal growth is accomplished based entirely on cartilage expansion prior to ossification, not limiting the transversal cartilage expansion and resulting in bulkier skeletal parts. The oriented extension of salamander cartilage and bone appear similar to the development of basicranial synchondroses in mammals, as we found no evidence for cartilage stem cell niches or growth plate-like structures during neither development nor regeneration. Both regenerative and developmental ossification in salamanders start from the cortical bone and proceeds inwards, showing the diversity of schemes for the synchrony of cortical and endochondral ossification among vertebrates.


Subject(s)
Osteogenesis , Urodela , Animals , Bone and Bones , Cartilage , Cell Division , Mammals
4.
Elife ; 92020 10 16.
Article in English | MEDLINE | ID: mdl-33063669

ABSTRACT

Growth plate and articular cartilage constitute a single anatomical entity early in development but later separate into two distinct structures by the secondary ossification center (SOC). The reason for such separation remains unknown. We found that evolutionarily SOC appears in animals conquering the land - amniotes. Analysis of the ossification pattern in mammals with specialized extremities (whales, bats, jerboa) revealed that SOC development correlates with the extent of mechanical loads. Mathematical modeling revealed that SOC reduces mechanical stress within the growth plate. Functional experiments revealed the high vulnerability of hypertrophic chondrocytes to mechanical stress and showed that SOC protects these cells from apoptosis caused by extensive loading. Atomic force microscopy showed that hypertrophic chondrocytes are the least mechanically stiff cells within the growth plate. Altogether, these findings suggest that SOC has evolved to protect the hypertrophic chondrocytes from the high mechanical stress encountered in the terrestrial environment.


Subject(s)
Cell Differentiation , Cell Proliferation , Chondrocytes/metabolism , Growth Plate/growth & development , Osteogenesis , Animals , Biomechanical Phenomena , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Stress, Mechanical
5.
Front Cell Dev Biol ; 8: 592, 2020.
Article in English | MEDLINE | ID: mdl-32754592

ABSTRACT

With very few exceptions, all adult tissues in mammals are maintained and can be renewed by stem cells that self-renew and generate the committed progeny required. These functions are regulated by a specific and in many ways unique microenvironment in stem cell niches. In most cases disruption of an adult stem cell niche leads to depletion of stem cells, followed by impairment of the ability of the tissue in question to maintain its functions. The presence of stem cells, often referred to as mesenchymal stem cells (MSCs) or multipotent bone marrow stromal cells (BMSCs), in the adult skeleton has long been realized. In recent years there has been exceptional progress in identifying and characterizing BMSCs in terms of their capacity to generate specific types of skeletal cells in vivo. Such BMSCs are often referred to as skeletal stem cells (SSCs) or skeletal stem and progenitor cells (SSPCs), with the latter term being used throughout this review. SSPCs have been detected in the bone marrow, periosteum, and growth plate and characterized in vivo on the basis of various genetic markers (i.e., Nestin, Leptin receptor, Gremlin1, Cathepsin-K, etc.). However, the niches in which these cells reside have received less attention. Here, we summarize the current scientific literature on stem cell niches for the SSPCs identified so far and discuss potential factors and environmental cues of importance in these niches in vivo. In this context we focus on (i) articular cartilage, (ii) growth plate cartilage, (iii) periosteum, (iv) the adult endosteal compartment, and (v) the developing endosteal compartment, in that order.

6.
J Exp Med ; 217(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-31914175

ABSTRACT

The gene IL6ST encodes GP130, the common signal transducer of the IL-6 cytokine family consisting of 10 cytokines. Previous studies have identified cytokine-selective IL6ST defects that preserve LIF signaling. We describe three unrelated families with at least five affected individuals who presented with lethal Stüve-Wiedemann-like syndrome characterized by skeletal dysplasia and neonatal lung dysfunction with additional features such as congenital thrombocytopenia, eczematoid dermatitis, renal abnormalities, and defective acute-phase response. We identified essential loss-of-function variants in IL6ST (a homozygous nonsense variant and a homozygous intronic splice variant with exon skipping). Functional tests showed absent cellular responses to GP130-dependent cytokines including IL-6, IL-11, IL-27, oncostatin M (OSM), and leukemia inhibitory factor (LIF). Genetic reconstitution of GP130 by lentiviral transduction in patient-derived cells reversed the signaling defect. This study identifies a new genetic syndrome caused by the complete lack of signaling of a whole family of GP130-dependent cytokines in humans and highlights the importance of the LIF signaling pathway in pre- and perinatal development.


Subject(s)
Cytokine Receptor gp130/metabolism , Exostoses, Multiple Hereditary/metabolism , Osteochondrodysplasias/metabolism , Signal Transduction/physiology , Antigens, CD/metabolism , Cells, Cultured , HEK293 Cells , Humans , Interleukin-11/metabolism , Interleukin-6/metabolism , Leukemia Inhibitory Factor/metabolism , Oncostatin M/metabolism , Receptors, Cytokine/metabolism
7.
Pediatr Res ; 87(6): 986-990, 2020 05.
Article in English | MEDLINE | ID: mdl-31830758

ABSTRACT

Children's longitudinal growth is facilitated by the activity of the growth plates, cartilage discs located near the ends of the long-bones. In order to elongate these bones, growth plates must continuously generate chondrocytes. Two recent studies have demonstrated that there are stem cells and a stem cell niche in the growth plate, which govern the generation of chondrocytes during the postnatal growth period. The niche, which allows stem cells to renew, appears at the same time as the secondary ossification center (SOC) matures into a bone epiphysis. Thus, the mechanism of chondrocyte generation differs substantially between neonatal and postnatal age, i.e., before and after the formation of the mineralized epiphyses. Hence, at the neonatal age bone growth is based on a consumption of chondro-progenitors whereas postnatally it is based on the activity of the stem cell niche. Here we discuss potential implications of these observations in relation to longitudinal growth, including the effects of estrogens, nutrition and growth hormone.


Subject(s)
Body Height , Bone Development , Child Development , Chondrocytes/physiology , Growth Plate/physiology , Stem Cell Niche , Stem Cells/physiology , Age Factors , Cell Differentiation , Cell Proliferation , Child , Child Nutritional Physiological Phenomena , Child, Preschool , Estrogens/metabolism , Growth Plate/cytology , Human Growth Hormone/metabolism , Humans , Infant , Infant, Newborn , Nutritional Status
8.
J Vis Exp ; (152)2019 10 23.
Article in English | MEDLINE | ID: mdl-31710038

ABSTRACT

Labeling an individual cell in the body to monitor which cell types it can give rise to and track its migration through the organism or determine its longevity can be a powerful way to reveal mechanisms of tissue development and maintenance. One of the most important tools currently available to monitor cells in vivo is the Confetti mouse model. The Confetti model can be used to genetically label individual cells in living mice with various fluorescent proteins in a cell type-specific manner and monitor their fate, as well as the fate of their progeny over time, in a process called clonal genetic tracing or clonal lineage tracing. This model was generated almost a decade ago and has contributed to an improved understanding of many biological processes, particularly related to stem cell biology, development, and renewal of adult tissues. However, preserving the fluorescent signal until image collection and simultaneous capturing of various fluorescent signals is technically challenging, particularly for mineralized tissue. This publication describes a step-by-step protocol for using the Confetti model to analyze growth plate cartilage that can be applied to any mineralized or nonmineralized tissue.


Subject(s)
Calcification, Physiologic/physiology , Cell Lineage , Growth Plate/cytology , Growth Plate/growth & development , Animals , Calcification, Physiologic/genetics , Cells, Cultured , Genes, Reporter , Indicators and Reagents , Mice , Models, Animal , Stem Cells/metabolism
9.
Nature ; 567(7747): 234-238, 2019 03.
Article in English | MEDLINE | ID: mdl-30814736

ABSTRACT

Longitudinal bone growth in children is sustained by growth plates, narrow discs of cartilage that provide a continuous supply of chondrocytes for endochondral ossification1. However, it remains unknown how this supply is maintained throughout childhood growth. Chondroprogenitors in the resting zone are thought to be gradually consumed as they supply cells for longitudinal growth1,2, but this model has never been proved. Here, using clonal genetic tracing with multicolour reporters and functional perturbations, we demonstrate that longitudinal growth during the fetal and neonatal periods involves depletion of chondroprogenitors, whereas later in life, coinciding with the formation of the secondary ossification centre, chondroprogenitors acquire the capacity for self-renewal, resulting in the formation of large, stable monoclonal columns of chondrocytes. Simultaneously, chondroprogenitors begin to express stem cell markers and undergo symmetric cell division. Regulation of the pool of self-renewing progenitors involves the hedgehog and mammalian target of rapamycin complex 1 (mTORC1) signalling pathways. Our findings indicate that a stem cell niche develops postnatally in the epiphyseal growth plate, which provides a continuous supply of chondrocytes over a prolonged period.


Subject(s)
Chondrocytes/cytology , Clone Cells/cytology , Growth Plate/cytology , Stem Cell Niche/physiology , Aging , Animals , Cartilage/cytology , Cell Self Renewal , Clone Cells/metabolism , Female , Growth Plate/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice
10.
Biochem J ; 476(5): 779-781, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30842311

ABSTRACT

Colorectal cancer is one of the most common forms of cancer in the world, with more than half a million new cases annually. Amongst the most promising new therapies, niclosamide-an FDA-approved drug for treating tapeworm infections-is being assessed in a stage II clinical trial for the treatment of metastatic colorectal cancer. Despite this advanced stage of research, the underlying mechanisms behind its actions remain uncertain. Niclosamide reduces the growth of colorectal cancer cells by targeting several intracellular signalling pathways, including the ß-catenin-dependent WNT signalling pathway. In a recent paper published in the Biochemical Journal [Biochem. J. (2019) 476, 535-546], Wang and colleagues revealed that niclosamide down-regulates ß-catenin-dependent WNT signalling in colorectal cancer cells by degrading components of the pathway via autophagy. Autophagy is a catabolic process in which cellular macromolecules and organelles are recycled to their monomer units. This finding provides a further understanding of the actions of niclosamide upon colorectal cancer cells and may yield improved future treatment models for colorectal cancer patients.


Subject(s)
Autophagic Cell Death/drug effects , Colorectal Neoplasms/drug therapy , Niclosamide/therapeutic use , Wnt Signaling Pathway/drug effects , Animals , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Neoplasm Metastasis
11.
Bone Rep ; 8: 64-71, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29955624

ABSTRACT

There are several pitfalls associated with research based on transgenic mice. Here, we describe our interpretation and analysis of mTORC1 activation in growth plate chondrocytes and compare these to a recent publication (Yan et al., Nature Communications 2016, 7:11151). Both laboratories employed TSC1-floxed mice crossed with collagen type 2-driven Cre (Col2-Cre), but drew substantially different conclusions. It was reported that activation of mechanistic target of rapamycin complex 1 (mTORC1) via Tsc1 ablation promotes the hypertrophy of growth plate chondrocytes, whereas we observe only disorganization in the resting zone, with no effect on chondrocyte hypertrophy or proliferation. Here, we present our data and discuss the differences in comparison to the earlier phenotypic characterization of TSC1 ablation in cartilage. Importantly, we detect Col2-Cre activity in non-cartilaginous tissues (including the brain) and discuss it in relation to other studies reporting non-cartilaginous expression of collagen alpha(1) II. Altogether, we conclude that mouse phenotypes following genetic ablation using Col2-Cre should be interpreted with care. We also conclude that activation of mTORC1 by TSC1 ablation in postnatal chondrocytes with inducible Col2-Cre (Col2-CreERt) leads to disorganization of the resting zone but causes no changes in chondrocyte proliferation or differentiation.

12.
Elife ; 62017 04 17.
Article in English | MEDLINE | ID: mdl-28414273

ABSTRACT

Cartilaginous structures are at the core of embryo growth and shaping before the bone forms. Here we report a novel principle of vertebrate cartilage growth that is based on introducing transversally-oriented clones into pre-existing cartilage. This mechanism of growth uncouples the lateral expansion of curved cartilaginous sheets from the control of cartilage thickness, a process which might be the evolutionary mechanism underlying adaptations of facial shape. In rod-shaped cartilage structures (Meckel, ribs and skeletal elements in developing limbs), the transverse integration of clonal columns determines the well-defined diameter and resulting rod-like morphology. We were able to alter cartilage shape by experimentally manipulating clonal geometries. Using in silico modeling, we discovered that anisotropic proliferation might explain cartilage bending and groove formation at the macro-scale.


Subject(s)
Cartilage/embryology , Vertebrates/embryology , Animals , Computer Simulation , Mice , Models, Biological
13.
FASEB J ; 31(3): 1067-1084, 2017 03.
Article in English | MEDLINE | ID: mdl-27965322

ABSTRACT

Articular cartilage has little regenerative capacity. Recently, genetic lineage tracing experiments have revealed chondrocyte progenitors at the articular surface. We further characterized these progenitors by using in vivo genetic approaches. Histone H2B-green fluorescent protein retention revealed that superficial cells divide more slowly than underlying articular chondrocytes. Clonal genetic tracing combined with immunohistochemistry revealed that superficial cells renew their number by symmetric division, express mesenchymal stem cell markers, and generate chondrocytes via both asymmetric and symmetric differentiation. Quantitative analysis of cellular kinetics, in combination with phosphotungstic acid-enhanced micro-computed tomography, showed that superficial cells generate chondrocytes and contribute to the growth and reshaping of articular cartilage. Furthermore, we found that cartilage renewal occurs as the progeny of superficial cells fully replace fetal chondrocytes during early postnatal life. Thus, superficial cells are self-renewing progenitors that are capable of maintaining their own population and fulfilling criteria of unipotent adult stem cells. Furthermore, the progeny of these cells reconstitute adult articular cartilage de novo, entirely substituting fetal chondrocytes.-Li, L., Newton, P. T., Bouderlique, T., Sejnohova, M., Zikmund, T., Kozhemyakina, E., Xie, M., Krivanek, J., Kaiser, J., Qian, H., Dyachuk, V., Lassar, A. B., Warman, M. L., Barenius, B., Adameyko, I., Chagin, A. S. Superficial cells are self-renewing chondrocyte progenitors, which form the articular cartilage in juvenile mice.


Subject(s)
Adult Stem Cells/cytology , Cartilage, Articular/cytology , Chondrocytes/cytology , Chondrogenesis , Animals , Cartilage, Articular/physiology , Mice , Regeneration
14.
Ann Rheum Dis ; 75(3): 627-31, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26438374

ABSTRACT

OBJECTIVES: It has been suggested that the lysosomal recycling process called macro-autophagy plays a role in osteoarthritis development. We thus decided to genetically ablate the autophagy-indispensable Atg5 gene specifically in chondrocytes and analyse the development of osteoarthritis upon aging and in a post-traumatic model. METHODS: Mice lacking the Atg5 gene in their chondrocytes (Atg5cKO) were generated by crossing Atg5-floxed mice with transgenic mice that expressed cre recombinase driven by the collagen type 2 promoter. Animals were analysed at the age of 2, 6 and 12 months for age-related osteoarthritis or underwent mini-open partial medial meniscectomy at 2 months of age and were analysed 1 or 2 months after surgery. We evaluated osteoarthritis using the Osteoarthritis Research Society International (OARSI) scoring on safranin-O-stained samples. Cell death was evaluated by terminal deoxy-nucleotidyl-transferase-mediated deoxy-UTP nick end labelling (TUNEL) and by immunostaining of cleaved caspases. RESULTS: We observed the development of osteoarthritis in Atg5cKO mice with aging including fibrillation and loss of proteoglycans, which was particularly severe in males. The ablation of Atg5 was associated with an increased cell death as assessed by TUNEL, cleaved caspase 3 and cleaved caspase 9. Surprisingly, no difference in the development of post-traumatic osteoarthritis was observed between Atg5cKO and control mice. CONCLUSIONS: Autophagy protects from age-related osteoarthritis by facilitating chondrocyte survival.


Subject(s)
Autophagy/genetics , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Microtubule-Associated Proteins/genetics , Osteoarthritis/genetics , Animals , Autophagy-Related Protein 5 , Cartilage, Articular/cytology , Caspases/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Osteoarthritis/metabolism , Proteoglycans/metabolism , Tibial Meniscus Injuries
15.
Autophagy ; 11(9): 1594-607, 2015.
Article in English | MEDLINE | ID: mdl-26259639

ABSTRACT

Mechanistic target of rapamycin (serine/threonine kinase) complex 1 (MTORC1) is a protein-signaling complex at the fulcrum of anabolic and catabolic processes, which acts depending on wide-ranging environmental cues. It is generally accepted that lysosomes facilitate MTORC1 activation by generating an internal pool of amino acids. Amino acids activate MTORC1 by stimulating its translocation to the lysosomal membrane where it forms a super-complex involving the lysosomal-membrane-bound vacuolar-type H(+)-ATPase (v-ATPase) proton pump. This translocation and MTORC1 activation require functional lysosomes. Here we found that, in contrast to this well-accepted concept, in epiphyseal chondrocytes inhibition of lysosomal activity by v-ATPase inhibitors bafilomycin A1 or concanamycin A potently activated MTORC1 signaling. The activity of MTORC1 was visualized by phosphorylated forms of RPS6 (ribosomal protein S6) and EIF4EBP1, 2 well-known downstream targets of MTORC1. Maximal RPS6 phosphorylation was observed at 48-h treatment and reached as high as a 12-fold increase (p < 0.018). This activation of MTORC1 was further confirmed in bone organ culture and promoted potent stimulation of longitudinal growth (p < 0.001). Importantly, the same effect was observed in ATG5 (autophagy-related 5)-deficient bones suggesting a macroautophagy-independent mechanism of MTORC1 inhibition by lysosomes. Thus, our data show that in epiphyseal chondrocytes lysosomes inhibit MTORC1 in a macroautophagy-independent manner and this inhibition likely depends on v-ATPase activity.


Subject(s)
Autophagy/drug effects , Chondrocytes/metabolism , Lysosomes/metabolism , Macrolides/pharmacology , Multiprotein Complexes/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Bone Development/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chondrocytes/drug effects , Chondrocytes/pathology , Hypertrophy , Lysosomes/drug effects , Mechanistic Target of Rapamycin Complex 1 , Mice , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Rats , Ribosomal Protein S6/metabolism , Tumor Suppressor Proteins/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
16.
J Bone Miner Res ; 30(12): 2249-61, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26077727

ABSTRACT

Longitudinal bone growth takes place in epiphyseal growth plates located in the ends of long bones. The growth plate consists of chondrocytes traversing from the undifferentiated (resting zone) to the terminally differentiated (hypertrophic zone) stage. Autophagy is an intracellular catabolic process of lysosome-dependent recycling of intracellular organelles and protein complexes. Autophagy is activated during nutritionally depleted or hypoxic conditions in order to facilitate cell survival. Chondrocytes in the middle of the growth plate are hypoxic and nutritionally depleted owing to the avascular nature of the growth plate. Accordingly, autophagy may facilitate their survival. To explore the role of autophagy in chondrocyte survival and constitutional bone growth, we generated mice with cartilage-specific ablation of either Atg5 (Atg5cKO) or Atg7 (Atg7cKO) by crossing Atg5 or Atg7 floxed mice with cartilage-specific collagen type 2 promoter-driven Cre. Both Atg5cKO and Atg7cKO mice showed growth retardation associated with enhanced chondrocyte cell death and decreased cell proliferation. Similarly, inhibition of autophagy by Bafilomycin A1 (Baf) or 3-methyladenine (3MA) promoted cell death in cultured slices of human growth plate tissue. To delineate the underlying mechanisms we employed ex vivo cultures of mouse metatarsal bones and RCJ3.IC5.18 rat chondrogenic cell line. Baf or 3MA impaired metatarsal bone growth associated with processing of caspase-3 and massive cell death. Similarly, treatment of RCJ3.IC5.18 chondrogenic cells by Baf also showed massive cell death and caspase-3 cleavage. This was associated with activation of caspase-9 and cytochrome C release. Altogether, our data suggest that autophagy is important for chondrocyte survival, and inhibition of this process leads to stunted growth and caspase-dependent death of chondrocytes.


Subject(s)
Caspases/metabolism , Chondrocytes/cytology , Gene Deletion , Microtubule-Associated Proteins/genetics , Adenine/analogs & derivatives , Adenine/metabolism , Animals , Apoptosis , Autophagy , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Cell Death , Cell Line , Cell Proliferation , Cell Survival , Collagen Type II/metabolism , Cytochromes c/metabolism , Growth Plate/growth & development , Humans , Hypoxia , Immunohistochemistry , In Situ Hybridization , Macrolides/metabolism , Metatarsal Bones/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/physiology , Proteins/genetics , Rats , Ubiquitin-Activating Enzymes/genetics
17.
Mol Med ; 20: 667-75, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25365546

ABSTRACT

Surgery and critical illness often associate with cognitive decline. Surgical trauma or infection can lead independently to learning and memory impairments via similar, but not identical, cellular signaling of the innate immune system that promotes neuroinflammation. In this study we explored the putative synergism between aseptic orthopedic surgery and infection, the latter reproduced by postoperative lipopolysaccharide (LPS) administration. We observed that surgery and LPS augmented systemic inflammation up to postoperative d 3 and this was associated with further neuroinflammation (CD11b and CD68 immunoreactivity) in the hippocampus in mice compared with those receiving surgery or LPS alone. Administration of a selective α7 subtype nicotinic acetylcholine receptor (α7 nAChR) agonist 2 h after LPS significantly improved neuroinflammation and hippocampal-dependent memory dysfunction. Modulation of nuclear factor-kappa B (NF-κB) activation in monocytes and regulation of the oxidative stress response through nicotinamide adenine dinucleotide phosphate (NADPH) signaling appear to be key targets in modulating this response. Overall, these results suggest that it may be conceivable to limit and possibly prevent postoperative complications, including cognitive decline and/or infections, through stimulation of the cholinergic antiinflammatory pathway.


Subject(s)
Cholinergic Agonists/therapeutic use , Cognition Disorders/drug therapy , Endotoxemia/drug therapy , Fractures, Bone/drug therapy , Postoperative Complications/drug therapy , Tibia/injuries , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Aza Compounds/pharmacology , Aza Compounds/therapeutic use , CD11b Antigen/metabolism , Cell Line , Cells, Cultured , Cholinergic Agonists/pharmacology , Cognition Disorders/blood , Cognition Disorders/metabolism , Cytokines/blood , Dioxins/pharmacology , Dioxins/therapeutic use , Endotoxemia/blood , Endotoxemia/metabolism , Fractures, Bone/blood , Fractures, Bone/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Lipopolysaccharides , Macrophages , Male , Memory/drug effects , Mice, Inbred C57BL , NF-kappa B/metabolism , Postoperative Complications/blood , Postoperative Complications/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...