Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biology (Basel) ; 13(4)2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38666853

ABSTRACT

Overnutrition, driven by the consumption of high-fat, high-sugar diets, has reached epidemic proportions and poses a significant global health challenge. Prolonged overnutrition leads to the deposition of excessive lipids in adipose and non-adipose tissues, a condition known as lipotoxicity. The intricate interplay between overnutrition-induced lipotoxicity and the immune system plays a pivotal role in the pathogenesis of various diseases. This review aims to elucidate the consequences of impaired efferocytosis, caused by lipotoxicity-poisoned macrophages, leading to chronic inflammation and the subsequent development of severe infectious diseases, autoimmunity, and cancer, as well as chronic pulmonary and cardiovascular diseases. Chronic overnutrition promotes adipose tissue expansion which induces cellular stress and inflammatory responses, contributing to insulin resistance, dyslipidemia, and metabolic syndrome. Moreover, sustained exposure to lipotoxicity impairs the efferocytic capacity of macrophages, compromising their ability to efficiently engulf and remove dead cells. The unresolved chronic inflammation perpetuates a pro-inflammatory microenvironment, exacerbating tissue damage and promoting the development of various diseases. The interaction between overnutrition, lipotoxicity, and impaired efferocytosis highlights a critical pathway through which chronic inflammation emerges, facilitating the development of severe infectious diseases, autoimmunity, cancer, and chronic pulmonary and cardiovascular diseases. Understanding these intricate connections sheds light on potential therapeutic avenues to mitigate the detrimental effects of overnutrition and lipotoxicity on immune function and tissue homeostasis, thereby paving the way for novel interventions aimed at reducing the burden of these multifaceted diseases on global health.

2.
J Immunol Res ; 2021: 8121407, 2021.
Article in English | MEDLINE | ID: mdl-34046506

ABSTRACT

To mimic Alzheimer's disease, transgenic mice overexpressing the amyloid precursor protein (APP) were used in this study. We hypothesize that the neuroprotective effects of ETAS®50, a standardized extract of Asparagus officinalis stem produced by Amino Up Co., Ltd. (Sapporo, Japan), are linked to the inhibition of the apoptosis cascade through an enhancement of the stress-response proteins: heat shock proteins (HSPs). APP-overexpressing mice (double-transgenic APP and PS1 mouse strains with a 129s6 background), ages 6-8 weeks old, and weighing 20-24 grams were successfully bred in our laboratory. The animals were divided into 5 groups. APP-overexpressing mice and wild-type (WT) mice were pretreated with ETAS®50 powder (50% elemental ETAS and 50% destrin) at 200 mg/kg and 1000 mg/kg body weight. Saline, the vehicle for ETAS®50, was administered in APP-overexpressing mice and WT mice. ETAS®50 and saline were administered by gavage daily for 1 month. Cognitive assessments, using the Morris Water Maze, demonstrated that memory was recovered following ETAS®50 treatment as compared to nontreated APP mice. At euthanization, the brain was removed and HSPs, amyloid ß, tau proteins, and caspase-3 were evaluated through immunofluorescence staining with the appropriate antibodies. Our data indicate that APP mice have cognitive impairment along with elevated amyloid ß, tau proteins, and caspase-3. ETAS®50 restored cognitive function in these transgenic mice, increased both HSP70 and HSP27, and attenuated pathogenic level of amyloid ß, tau proteins, and caspsase-3 leading to neuroprotection. Our results were confirmed with a significant increase in HSP70 gene expression in the hippocampus.


Subject(s)
Alzheimer Disease/drug therapy , Asparagus Plant/chemistry , Neuroprotective Agents/administration & dosage , Plant Extracts/administration & dosage , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Cognition/drug effects , Disease Models, Animal , Female , HSP27 Heat-Shock Proteins/analysis , HSP27 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/analysis , HSP70 Heat-Shock Proteins/metabolism , Hippocampus/pathology , Humans , Male , Memory/drug effects , Mice , Mice, Transgenic , Morris Water Maze Test/drug effects , Presenilin-1/genetics
3.
Neurol India ; 67(3): 684-691, 2019.
Article in English | MEDLINE | ID: mdl-31347535

ABSTRACT

This review discusses the past and recent findings on how changes in gravity affect cellular and subcellular parameters of the human nervous system and the implementation of cell and tissue models of nervous tissue on space biology. In order to prepare for long duration space exploration, a focus on space life sciences research is critical. Such research not only improves our knowledge of the basic biological processes but also elucidates the mechanisms and treatment of various earthly medical conditions. However, the study of living organisms in space poses many challenges that may be negligible or nonexistent in ground-based research. In recent years, with an increase in the number of spaceflights, extended periods of stay of astronauts on the International Space station and the imminent possibility of future long term deep space exploration missions, there is a great deal of attention focused on the effects induced by altered gravitation on the human body, and in particular, on bone, skeletal muscle, immunity and brain function. The aim of this review is to collate, encapsulate and examine the effects of altered gravity on neuronal cell structure and function that have been established from data obtained during experiments performed in real microgravity and simulated microgravity like conditions.


Subject(s)
Nervous System Physiological Phenomena , Weightlessness , Animals , Cell Membrane/physiology , Humans , Neuroglia/physiology , Neurons/physiology , Space Flight
4.
Neurol India ; 67(Supplement): S198-S203, 2019.
Article in English | MEDLINE | ID: mdl-31134910

ABSTRACT

Space exploration exposes astronauts to a variety of gravitational stresses. Exposure to a reduced gravity environment affects human anatomy and physiology. Countermeasures to restore homeostatic states within the human body have begun. The pathophysiological effects of exposure to microgravity, on the neurological system, are, however, still not clear. NASA has scheduled deep space exploration of extraterrestrial locations such as the Moon and Mars in the 2030s. Adverse health effects related to the human exposure to microgravity from previous, relatively shorter missions have been documented. A lengthy deep space travel to Mars could be overburdened by significant adverse health effects. Astronauts demonstrate a significant increase in the number of many types of circulating white blood cells (neutrophils, monocytes, T-helper cells, and B-cells) but a decrease in natural killer cells. It is unclear whether these changes are due to increased production or decreased clearance of these cells. In this review, viral reactivation in astronauts will be discussed, including the occurrence of clinical cases before, during, or after spaceflight and their management during and after flight. Studies on models used in spaceflight studies such as the AKATA cells (an immortalized B-cell line derived from a Japanese patient with Burkitt's lymphoma, a tumor induced by Epstein-Barr virus) and other cell lines which shed these latent viruses, will be reviewed with specific reference to gravitational changes, radiation, and spaceflight-induced immune suppression.


Subject(s)
Astronauts , Immunosuppression Therapy , Nervous System Physiological Phenomena , Virus Activation , Weightlessness/adverse effects , Extraterrestrial Environment , Humans , Space Flight
5.
Int J Mol Sci ; 20(6)2019 Mar 18.
Article in English | MEDLINE | ID: mdl-30889841

ABSTRACT

Human cells, when exposed to both real and simulated microgravity (s-µg), form 3D tissue constructs mirroring in vivo architectures (e.g., cartilage, intima constructs, cancer spheroids and others). In this study, we exposed human foetal osteoblast (hFOB 1.19) cells to a Random Positioning Machine (RPM) for 7 days and 14 days, with the purpose of investigating the effects of s-µg on biological processes and to engineer 3D bone constructs. RPM exposure of the hFOB 1.19 cells induces alterations in the cytoskeleton, cell adhesion, extra cellular matrix (ECM) and the 3D multicellular spheroid (MCS) formation. In addition, after 7 days, it influences the morphological appearance of these cells, as it forces adherent cells to detach from the surface and assemble into 3D structures. The RPM-exposed hFOB 1.19 cells exhibited a differential gene expression of the following genes: transforming growth factor beta 1 (TGFB1, bone morphogenic protein 2 (BMP2), SRY-Box 9 (SOX9), actin beta (ACTB), beta tubulin (TUBB), vimentin (VIM), laminin subunit alpha 1 (LAMA1), collagen type 1 alpha 1 (COL1A1), phosphoprotein 1 (SPP1) and fibronectin 1 (FN1). RPM exposure also induced a significantly altered release of the cytokines and bone biomarkers sclerostin (SOST), osteocalcin (OC), osteoprotegerin (OPG), osteopontin (OPN), interleukin 1 beta (IL-1ß) and tumour necrosis factor 1 alpha (TNF-1α). After the two-week RPM exposure, the spheroids presented a bone-specific morphology. In conclusion, culturing cells in s-µg under gravitational unloading represents a novel technology for tissue-engineering of bone constructs and it can be used for investigating the mechanisms behind spaceflight-related bone loss as well as bone diseases such as osteonecrosis or bone injuries.


Subject(s)
Bone and Bones/physiology , Fetus/cytology , Osteoblasts/cytology , Tissue Engineering/methods , Bone Morphogenetic Protein 2/metabolism , Cell Shape , Cells, Cultured , Cytoskeleton/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation , Humans , Organoids/cytology , Osteoblasts/metabolism , Osteogenesis , Protein Binding , Signal Transduction , Solubility , Subcellular Fractions/metabolism , Transforming Growth Factor beta/metabolism , Weightlessness Simulation
6.
Stem Cells Dev ; 27(12): 787-804, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29596037

ABSTRACT

Experimental cell research studying three-dimensional (3D) tissues in space and on Earth using new techniques to simulate microgravity is currently a hot topic in Gravitational Biology and Biomedicine. This review will focus on the current knowledge of the use of stem cells and specialized cells for tissue engineering under simulated microgravity conditions. We will report on recent advancements in the ability to construct 3D aggregates from various cell types using devices originally created to prepare for spaceflights such as the random positioning machine (RPM), the clinostat, or the NASA-developed rotating wall vessel (RWV) bioreactor, to engineer various tissues such as preliminary vessels, eye tissue, bone, cartilage, multicellular cancer spheroids, and others from different cells. In addition, stem cells had been investigated under microgravity for the purpose to engineer adipose tissue, cartilage, or bone. Recent publications have discussed different changes of stem cells when exposed to microgravity and the relevant pathways involved in these biological processes. Tissue engineering in microgravity is a new technique to produce organoids, spheroids, or tissues with and without scaffolds. These 3D aggregates can be used for drug testing studies or for coculture models. Multicellular tumor spheroids may be interesting for radiation experiments in the future and to reduce the need for in vivo experiments. Current achievements using cells from patients engineered on the RWV or on the RPM represent an important step in the advancement of techniques that may be applied in translational Regenerative Medicine.


Subject(s)
Stem Cells/metabolism , Tissue Engineering/methods , Weightlessness Simulation , Weightlessness , Animals , Bioreactors , Bone and Bones/cytology , Bone and Bones/metabolism , Cartilage/cytology , Cartilage/metabolism , Humans , Organoids/cytology , Organoids/metabolism , Stem Cells/cytology
7.
Cell Physiol Biochem ; 40(1-2): 1-17, 2016.
Article in English | MEDLINE | ID: mdl-27842307

ABSTRACT

BACKGROUND/AIMS: Microgravity (µg) has adverse effects on the eye of humans in space. The risk of visual impairment is therefore one of the leading health concerns for NASA. The impact of µg on human adult retinal epithelium (ARPE-19) cells is unknown. METHODS: In this study we investigated the influence of simulated µg (s-µg; 5 and 10 days (d)), using a Random Positioning Machine (RPM), on ARPE-19 cells. We performed phase-contrast/fluorescent microscopy, qRT-PCR, Western blotting and pathway analysis. RESULTS: Following RPM-exposure a subset of ARPE-19 cells formed multicellular spheroids (MCS), whereas the majority of the cells remained adherent (AD). After 5d, alterations of F-actin and fibronectin were observed which reverted after 10d-exposure, suggesting a time-dependent adaptation to s-µg. Gene expression analysis of 12 genes involved in cell structure, shape, adhesion, migration, and angiogenesis suggested significant changes after a 10d-RPM-exposure. 11 genes were down-regulated in AD and MCS 10d-RPM-samples compared to 1g, whereas FLK1 was up-regulated in 5d- and 10d-RPM-MCS-samples. Similarly, TIMP1 was up-regulated in 5d-RPM-samples, whereas the remaining genes were down-regulated in 5d-RPM-samples. Western blotting revealed similar changes in VEGF, ß-actin, laminin and fibronectin of 5d-RPM-samples compared to 10d, whereas different alterations of ß-tubulin and vimentin were observed. The pathway analysis showed complementing effects of VEGF and integrin ß-1. CONCLUSIONS: These findings clearly show that s-µg induces significant alterations in the F-actin-cytoskeleton and cytoskeleton-related proteins of ARPE-19, in addition to changes in cell growth behavior and gene expression patterns involved in cell structure, growth, shape, migration, adhesion and angiogenesis.


Subject(s)
Cytoskeleton/genetics , Extracellular Matrix/genetics , Gene Expression Regulation , Retinal Pigment Epithelium/metabolism , Weightlessness , Actin Cytoskeleton/metabolism , Actins/metabolism , Adult , Cell Adhesion , Cell Proliferation , Cell Shape , Cytoskeleton/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , Models, Biological , Phenotype , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/metabolism
8.
Bone ; 87: 44-56, 2016 06.
Article in English | MEDLINE | ID: mdl-27032715

ABSTRACT

Experiencing real weightlessness in space is a dream for many of us who are interested in space research. Although space traveling fascinates us, it can cause both short-term and long-term health problems. Microgravity is the most important influence on the human organism in space. The human body undergoes dramatic changes during a long-term spaceflight. In this review, we will mainly focus on changes in calcium, sodium and bone metabolism of space travelers. Moreover, we report on the current knowledge on the mechanisms of bone loss in space, available models to simulate the effects of microgravity on bone on Earth as well as the combined effects of microgravity and cosmic radiation on bone. The available countermeasures applied in space will also be evaluated.


Subject(s)
Bone and Bones/physiology , Weightlessness , Bone Resorption/pathology , Bone and Bones/metabolism , Calcium/metabolism , Humans , Sodium/metabolism , Space Flight
9.
Article in English | MEDLINE | ID: mdl-26788109

ABSTRACT

Active Hexose Correlated Compound (AHCC) is a fermented mushroom extract and immune supplement that has been used to treat a wide range of health conditions. It helps in augmentation of the natural immune response and affects immune cell activation and outcomes. The goal of this project was to study and understand the role and mechanisms of AHCC supplementation in the prevention of immunosuppression through T cell activation. The method described here involves "in vitro" culturing of lymphocytes, exposing them to different concentrations of AHCC (0 µg/mL, 50 µg/mL, 100 µg/mL, 250 µg/mL, and 500 µg/mL) at 0 hours. Interestingly, clumping and aggregation of the cells were seen between 24 and 72 hours of incubation. The cells lay down extracellular matrix, which become adherent, and phenotypical changes from small rounded lymphocytes to large macrophage-like, spindle shaped, elongated, fibroblast-like cells even beyond 360 hours were observed. These are probably translated from genotypic changes in the cells since the cells propagate for at least 3 to 6 generations (present observations). RNA isolated was subjected to gene array analysis. We hypothesize that cell adhesion is an activation and survival pathway in lymphocytes and this could be the mechanism of AHCC activation in human lymphocytes.

SELECTION OF CITATIONS
SEARCH DETAIL
...