Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 392
1.
J Transl Med ; 22(1): 437, 2024 May 08.
Article En | MEDLINE | ID: mdl-38720345

BACKGROUND: Biological-derived hydroxyapatite is widely used as a bone substitute for addressing bone defects, but its limited osteoconductive properties necessitate further improvement. The osteo-immunomodulatory properties hold crucial promise in maintaining bone homeostasis, and precise modulation of macrophage polarization is essential in this process. Metabolism serves as a guiding force for immunity, and fluoride modification represents a promising strategy for modulating the osteoimmunological environment by regulating immunometabolism. In this context, we synthesized fluorinated porcine hydroxyapatite (FPHA), and has demonstrated its enhanced biological properties and osteogenic capacity. However, it remains unknown whether and how FPHA affects the immune microenvironment of the bone defects. METHODS: FPHA was synthesized and its composition and structural properties were confirmed. Macrophages were cultured with FPHA extract to investigate the effects of FPHA on their polarization and the related osteo-immune microenvironment. Furthermore, total RNA of these macrophages was extracted, and RNA-seq analysis was performed to explore the underlying mechanisms associated with the observed changes in macrophages. The metabolic states were evaluated with a Seahorse analyzer. Additionally, immunohistochemical staining was performed to evaluate the macrophages response after implantation of the novel bone substitutes in critical size calvarial defects in SD rats. RESULTS: The incorporation of fluoride ions in FPHA was validated. FPHA promoted macrophage proliferation and enhanced the expression of M2 markers while suppressing the expression of M1 markers. Additionally, FPHA inhibited the expression of inflammatory factors and upregulated the expression of osteogenic factors, thereby enhancing the osteogenic differentiation capacity of the rBMSCs. RNA-seq analysis suggested that the polarization-regulating function of FPHA may be related to changes in cellular metabolism. Further experiments confirmed that FPHA enhanced mitochondrial function and promoted the metabolic shift of macrophages from glycolysis to oxidative phosphorylation. Moreover, in vivo experiments validated the above results in the calvarial defect model in SD rats. CONCLUSION: In summary, our study reveals that FPHA induces a metabolic shift in macrophages from glycolysis to oxidative phosphorylation. This shift leads to an increased tendency toward M2 polarization in macrophages, consequently creating a favorable osteo-immune microenvironment. These findings provide valuable insights into the impact of incorporating an appropriate concentration of fluoride on immunometabolism and macrophage mitochondrial function, which have important implications for the development of fluoride-modified immunometabolism-based bone regenerative biomaterials and the clinical application of FPHA or other fluoride-containing materials.


Durapatite , Glycolysis , Macrophages , Oxidative Phosphorylation , Rats, Sprague-Dawley , Animals , Durapatite/chemistry , Macrophages/metabolism , Macrophages/drug effects , Oxidative Phosphorylation/drug effects , Glycolysis/drug effects , Rats , Swine , Cell Proliferation/drug effects , Male , Osteogenesis/drug effects , Skull/pathology , Skull/drug effects , Mice , Cellular Microenvironment/drug effects , RAW 264.7 Cells , Bone and Bones/metabolism , Bone and Bones/drug effects
2.
J Clin Invest ; 134(10)2024 Mar 21.
Article En | MEDLINE | ID: mdl-38512413

Elevated bone resorption and diminished bone formation have been recognized as the primary features of glucocorticoid-associated skeletal disorders. However, the direct effects of excess glucocorticoids on bone turnover remain unclear. Here, we explored the outcomes of exogenous glucocorticoid treatment on bone loss and delayed fracture healing in mice and found that reduced bone turnover was a dominant feature, resulting in a net loss of bone mass. The primary effect of glucocorticoids on osteogenic differentiation was not inhibitory; instead, they cooperated with macrophages to facilitate osteogenesis. Impaired local nutrient status - notably, obstructed fatty acid transportation - was a key factor contributing to glucocorticoid-induced impairment of bone turnover in vivo. Furthermore, fatty acid oxidation in macrophages fueled the ability of glucocorticoid-liganded receptors to enter the nucleus and then promoted the expression of BMP2, a key cytokine that facilitates osteogenesis. Metabolic reprogramming by localized fatty acid delivery partly rescued glucocorticoid-induced pathology by restoring a healthier immune-metabolic milieu. These data provide insights into the multifactorial metabolic mechanisms by which glucocorticoids generate skeletal disorders, thus suggesting possible therapeutic avenues.


Bone Remodeling , Glucocorticoids , Osteogenesis , Animals , Mice , Glucocorticoids/pharmacology , Osteogenesis/drug effects , Bone Remodeling/drug effects , Macrophages/metabolism , Macrophages/immunology , Macrophages/drug effects , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , Fatty Acids/metabolism , Bone and Bones/metabolism , Bone and Bones/drug effects , Bone and Bones/immunology , Cellular Microenvironment/drug effects
3.
Viruses ; 14(10)2022 10 09.
Article En | MEDLINE | ID: mdl-36298774

In this study, we evaluate the role of the MIF/CD74 axis in the functionality of CD4+ T lymphocytes (CD4TL) during HIV infection. MDMs from healthy donors were infected with a R5-tropic or Transmitted/Founder (T/F) HIV strain. At day 11 post-MDM infection, allogeneic co-cultures with uninfected CD4TLs plus MIF stimulus were performed. Cytokine production was evaluated by ELISA. MIF plasma levels of people with HIV (PWH) were evaluated by ELISA. The phenotype and infection rate of CD4TLs from PWH were analyzed after MIF stimulus. Intracellular cytokines and transcription factors were evaluated by flow cytometry. Data were analyzed by parametric or non-parametric methods. The MIF stimulation of HIV-infected MDMs induced an increased expression of IL-6, IL-1ß and IL-8. In CD4TL/MDM co-cultures, the MIF treatment increased IL-17A/RORγt-expressing CD4TLs. Higher concentrations of IL-17A in supernatants were also observed. These results were recapitulated using transmitted/founder (T/F) HIV-1 strains. The MIF treatment appeared to affect memory CD4TLs more than naïve CD4TLs. MIF blocking showed a negative impact on IL17A+CD4TL proportions. Higher MIF concentrations in PWH-derived plasma were correlated with higher IL-17A+CD4TL percentages. Finally, MIF stimulation in PWH-derived PBMCs led to an increase in Th17-like population. MIF may contribute to viral pathogenesis by generating a microenvironment enriched in activating mediators and Th17-like CD4TLs, which are known to be highly susceptible to HIV-1 infection and relevant to viral persistence. These observations establish a basis for considering MIF as a possible therapeutic target.


HIV Infections , Macrophage Migration-Inhibitory Factors , Th17 Cells , Humans , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/physiopathology , Interleukin-17 , Interleukin-6 , Interleukin-8 , Intramolecular Oxidoreductases , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/immunology , Macrophage Migration-Inhibitory Factors/pharmacology , Nuclear Receptor Subfamily 1, Group F, Member 3 , Transcription Factors , Th17 Cells/drug effects , Th17 Cells/immunology , Cellular Microenvironment/drug effects , Cellular Microenvironment/immunology
4.
J Nanobiotechnology ; 20(1): 12, 2022 Jan 04.
Article En | MEDLINE | ID: mdl-34983560

The management of diabetic ulcer (DU) to rescue stalled wound healing remains a paramount clinical challenge due to the spatially and temporally coupled pathological wound microenvironment that features hyperglycemia, biofilm infection, hypoxia and excessive oxidative stress. Here we present a pH-switchable nanozyme cascade catalysis (PNCC) strategy for spatial-temporal modulation of pathological wound microenvironment to rescue stalled healing in DU. The PNCC is demonstrated by employing the nanozyme of clinically approved iron oxide nanoparticles coated with a shell of glucose oxidase (Fe3O4-GOx). The Fe3O4-GOx possesses intrinsic glucose oxidase (GOx), catalase (CAT) and peroxidase (POD)-like activities, and can catalyze pH-switchable glucose-initiated GOx/POD and GOx/CAT cascade reaction in acidic and neutral environment, respectively. Specifically, the GOx/POD cascade reaction generating consecutive fluxes of toxic hydroxyl radical spatially targets the acidic biofilm (pH ~ 5.5), and eradicates biofilm to shorten the inflammatory phase and initiate normal wound healing processes. Furthermore, the GOx/CAT cascade reaction producing consecutive fluxes of oxygen spatially targets the neutral wound tissue, and accelerates the proliferation and remodeling phases of wound healing by addressing the issues of hyperglycemia, hypoxia, and excessive oxidative stress. The shortened inflammatory phase temporally coupled with accelerated proliferation and remodeling phases significantly speed up the normal orchestrated wound-healing cascades. Remarkably, this Fe3O4-GOx-instructed spatial-temporal remodeling of DU microenvironment enables complete re-epithelialization of biofilm-infected wound in diabetic mice within 15 days while minimizing toxicity to normal tissues, exerting great transformation potential in clinical DU management. The proposed PNCC concept offers a new perspective for complex pathological microenvironment remodeling, and may provide a powerful modality for the treatment of microenvironment-associated diseases.


Diabetes Mellitus, Experimental , Nanostructures , Ulcer , Animals , Biofilms/drug effects , Catalysis , Cellular Microenvironment/drug effects , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/pathology , Disease Models, Animal , Hydrogen-Ion Concentration , Mice , Nanomedicine , Ulcer/etiology , Ulcer/pathology , Wound Healing/drug effects
5.
Biomed Mater ; 17(2)2022 01 13.
Article En | MEDLINE | ID: mdl-34937006

Currentin vitrothree-dimensional (3D) models of liver tissue have been limited by the inability to study the effects of specific extracellular matrix (ECM) components on cell phenotypes. This is in part due to limitations in the availability of chemical modifications appropriate for this purpose. For example, hyaluronic acid (HA), which is a natural ECM component within the liver, lacks key ECM motifs (e.g. arginine-glycine-aspartic acid (RGD) peptides) that support cell adhesion. However, the addition of maleimide (Mal) groups to HA could facilitate the conjugation of ECM biomimetic peptides with thiol-containing end groups. In this study, we characterized a new crosslinkable hydrogel (i.e. HA-Mal) that yielded a simplified ECM-mimicking microenvironment supportive of 3D liver cell culture. We then performed a series of experiments to assess the impact of physical and biochemical signaling in the form of RGD peptide incorporation and transforming growth factorß(TGF-ß) supplementation, respectively, on hepatic functionality. Hepatic stellate cells (i.e. LX-2) exhibited increased cell-matrix interactions in the form of cell spreading and elongation within HA-Mal matrices containing RGD peptides, enabling physical adhesions, whereas hepatocyte-like cells (HepG2) had reduced albumin and urea production. We further exposed the encapsulated cells to soluble TGF-ßto elicit a fibrosis-like state. In the presence of TGF-ßbiochemical signals, LX-2 cells became activated and HepG2 functionality significantly decreased in both RGD-containing and RGD-free hydrogels. Altogether, in this study we have developed a hydrogel biomaterial platform that allows for discrete manipulation of specific ECM motifs within the hydrogel to better understand the roles of cell-matrix interactions on cell phenotype and overall liver functionality.


Biocompatible Materials , Hyaluronic Acid , Hydrogels/chemistry , Maleimides , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Adhesion/drug effects , Cell Culture Techniques , Cellular Microenvironment/drug effects , Extracellular Matrix/drug effects , Hep G2 Cells , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Maleimides/chemistry , Maleimides/pharmacology , Oligopeptides/chemistry , Surface Properties
6.
Biochem Biophys Res Commun ; 581: 46-52, 2021 12 03.
Article En | MEDLINE | ID: mdl-34653678

Excessive inflammation leads to periodontitis, which inhibits the osteogenic differentiation of human dental pulp stem cells (hDPSCs), irreversibly injured and difficultly repaired for the important dental pulp. Hence, it is necessary to study the functional gene to enhance the osteogenic differentiation of hDPSCs. Previous found that SNHG7 expression was increased in the osteogenic differentiation of hDPSCs. However, the regulatory functions of SNHG7 on osteogenic differentiation of hDPSCs in the inflammatory microenvironment still remains unknown. In this study, hDPSCs treatment with 50 ng/mL TNF-α to mimic the inflammatory microenvironment, then cultured in osteoblast differentiation medium for 14 days. SNHG7, miR-6512-3p, BSP, DSPP, DMP-1, RUNX2 and OPN in hDPSCs were detect by RT-qPCR. We found that SNHG7 expression was reduced during the osteogenic differentiation of hDPSCs after different concentrations TNF-α treatment. SNHG7 overexpression improved the TNF-α-induced suppression of calcium deposition, ALP activity, and the expression of BSP, DSPP, DMP-1, RUNX2 and OPN. Furthermore, SNHG7 can sponge with miR-6512-3p. miR-6512-3p expression was increased during the osteogenic differentiation of hDPSCs after different concentrations TNF-α treatment while inhibited after SNHG7 overexpression. knockdown of miR-6512-3p improved the TNF-α-induced suppression of calcium deposition, ALP activity, and the expression of BSP, DSPP, DMP-1, RUNX2 and OPN. Finally, miR-6512-3p overexpression reversed the effect of SNHG7 on the osteo/dentinogenic differentiation of TNF-α-treated hDPSCs. In conclusions, SNHG7 improves the osteogenic differentiation of hDPSCs by inhibiting miR-6512-3p expression under 50 ng/mL TNF-α-induced inflammatory environment, which provided potential targets for the treatment of periodontitis.


MicroRNAs/genetics , Osteoblasts/drug effects , Osteogenesis/drug effects , RNA, Small Nucleolar/genetics , Stem Cells/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Cell Differentiation , Cells, Cultured , Cellular Microenvironment/drug effects , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Culture Media/chemistry , Culture Media/pharmacology , Dental Pulp/cytology , Dental Pulp/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation , Humans , Inflammation , Integrin-Binding Sialoprotein/genetics , Integrin-Binding Sialoprotein/metabolism , MicroRNAs/metabolism , Models, Biological , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/genetics , Osteopontin/genetics , Osteopontin/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Small Nucleolar/metabolism , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism
7.
Sci Rep ; 11(1): 18418, 2021 09 16.
Article En | MEDLINE | ID: mdl-34531513

Osteoimmunity plays an important role in the process of implant osseointegration. Autophagy is a conservative metabolic pathway of eukaryotic cells, but whether the interaction between autophagy and osteoimmunity plays a key role in osseointegration remains unclear. In this study, we prepared smooth titanium disks and micro-nano topography titanium disks, to study the immune microenvironment of RAW264.7 cells, and prepared the conditioned medium to study the effect of immune microenvironment on the osteogenesis and autophagy of MC3T3-E1 cells. Autophagy inhibitor 3-MA was used to inhibit autophagy to observe the change of expression of osteogenic markers. The results showed that the micro-nano topography titanium disks could stimulate RAW264.7 cells to differentiate into M2 type, forming an anti-inflammatory immune microenvironment; compared with the control group, the anti-inflammatory immune microenvironment promoted the proliferation and differentiation of osteoblasts better. The anti-inflammatory immune environment activated the autophagy level of osteoblasts, while the expression of osteogenic markers was down-regulated after inhibition of autophagy. These results indicate that anti-inflammatory immune microenvironment can promote cell proliferation and osteogenic differentiation, autophagy plays an important role in this process. This study further explains the mechanism of implant osseointegration in osteoimmune microenvironment, and provides reference for improving implant osseointegration.


Autophagy , Nanotechnology , Osseointegration/immunology , Prostheses and Implants , Titanium/pharmacology , Animals , Autophagy/drug effects , Bone and Bones/drug effects , Bone and Bones/immunology , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Polarity/drug effects , Cell Proliferation/drug effects , Cellular Microenvironment/drug effects , Cellular Microenvironment/immunology , Gene Expression Regulation/drug effects , Inflammation/pathology , Macrophages/drug effects , Mice , Osseointegration/drug effects , RAW 264.7 Cells , Surface Properties
8.
Int J Mol Sci ; 22(15)2021 Jul 29.
Article En | MEDLINE | ID: mdl-34360901

The oxidative properties of nanomaterials arouse legitimate concerns about oxidative damage in biological systems. On the other hand, the undisputable benefits of nanomaterials promote them for biomedical applications; thus, the strategies to reduce oxidative potential are urgently needed. We aimed at analysis of nitrogen-containing carbon quantum dots (N-CQDs) in terms of their biocompatibility and internalization by different cells. Surprisingly, N-CQD uptake does not contribute to the increased oxidative stress inside cells and lacks cytotoxic influence even at high concentrations, primarily through protein corona formation. We proved experimentally that the protein coating effectively limits the oxidative capacity of N-CQDs. Thus, N-CQDs served as an immobilization support for three different enzymes with the potential to be used as therapeutics. Various kinetic parameters of immobilized enzymes were analyzed. Regardless of the enzyme structure and type of reaction catalyzed, adsorption on the nanocarrier resulted in increased catalytic efficiency. The enzymatic-protein-to-nanomaterial ratio is the pivotal factor determining the course of kinetic parameter changes that can be tailored for enzyme application. We conclude that the above properties of N-CQDs make them an ideal support for enzymatic drugs required for multiple biomedical applications, including personalized medical therapies.


Biocatalysis , Carbon/chemistry , Carbon/pharmacology , Nitrogen/chemistry , Nitrogen/pharmacology , Oxidative Stress/drug effects , Protein Corona/metabolism , Quantum Dots/chemistry , Quantum Dots/metabolism , A549 Cells , Animals , Apyrase/chemistry , Apyrase/pharmacology , Catalase/chemistry , Catalase/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cellular Microenvironment/drug effects , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/pharmacology , HeLa Cells , Humans , Rats , Reactive Oxygen Species/metabolism , beta-Galactosidase/chemistry , beta-Galactosidase/pharmacology
9.
Adv Sci (Weinh) ; 8(20): e2101526, 2021 10.
Article En | MEDLINE | ID: mdl-34436822

Reperfusion injury is still a major challenge that impedes neuronal survival in ischemic stroke. However, the current clinical treatments are remained on single pathological process, which are due to lack of comprehensive neuroprotective effects. Herein, a macrophage-disguised honeycomb manganese dioxide (MnO2 ) nanosphere loaded with fingolimod (FTY) is developed to salvage the ischemic penumbra. In particular, the biomimetic nanoparticles can accumulate actively in the damaged brain via macrophage-membrane protein-mediated recognition with cell adhesion molecules that are overexpressed on the damaged vascular endothelium. MnO2 nanosphere can consume excess hydrogen peroxide (H2 O2 ) and convert it into desiderated oxygen (O2 ), and can be decomposed in acidic lysosome for cargo release, so as to reduce oxidative stress and promote the transition of M1 microglia to M2 type, eventually reversing the proinflammatory microenvironment and reinforcing the survival of damaged neuron. This biomimetic nanomedicine raises new strategy for multitargeted combined treatment of ischemic stroke.


Inflammation/drug therapy , Ischemic Stroke/drug therapy , Nanoparticles/chemistry , Neurons/drug effects , Oxidative Stress/drug effects , Animals , Cell Line, Tumor , Cellular Microenvironment/drug effects , Fingolimod Hydrochloride/chemistry , Fingolimod Hydrochloride/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Ischemic Stroke/genetics , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Lysosomes/drug effects , Lysosomes/genetics , Macrophages/drug effects , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Nanospheres/chemistry , Neurons/pathology , Neuroprotection , Oxides/chemistry , Oxides/pharmacology , Oxygen/metabolism , Primary Cell Culture , Rats , Reperfusion Injury/drug therapy , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
10.
ACS Appl Mater Interfaces ; 13(27): 32316-32331, 2021 Jul 14.
Article En | MEDLINE | ID: mdl-34210131

Pathogenic bacterial infection has become a serious medical threat to global public health. Once the skin has serious defects, bacterial invasion and the following chain reactions will be a thorny clinical conundrum, which takes a long time to heal. Although various strategies have been used to eradicate bacteria, the treatment which can simultaneously disinfect and regulate the infection-related host responses is rarely reported. Herein, inspired by the host microenvironment, a photoenhanced dual-functional nanomedicine is constructed (Hemin@Phmg-TA-MSN) for localized bacterial ablation and host microenvironment modulation. The "NIR-triggered local microthermal therapy" and positively charged surface endow the nanomedicine with excellent bacterial capture and killing activities. Meanwhile, the nanomedicine exhibits broad-spectrum reactive oxygen species (ROS) scavenging activity via the synergistic effect of hemin and tannic acid with photoenhanced electron and hydrogen transfers. Furthermore, the in vivo experiments demonstrate that the dual-functional nanomedicine not only presents robust bacterial eradication capability, but also triggers the oxidative stress and inflammatory microenvironment regulation. The work not only shows a facile and effective way for infected wound management but also provides a new horizon for designing novel and efficient anti-infection therapy shifting focus from bacteria treatment to host microenvironment modulation.


Cellular Microenvironment/drug effects , Host-Pathogen Interactions/drug effects , Infrared Rays , Nanomedicine/methods , Wound Healing/radiation effects , Animals , Female , Mice
11.
Int J Mol Sci ; 22(11)2021 Jun 03.
Article En | MEDLINE | ID: mdl-34204881

Mesenchymal stem cells (MSCs) are broadly applied in regenerative therapy to replace cells that are lost or impaired during disease. The low survival rate of MSCs after transplantation is one of the major limitations heavily influencing the success of the therapy. Unfavorable microenvironments with inflammation and oxidative stress in the damaged regions contribute to MSCs loss. Most of the strategies developed to overcome this obstacle are aimed to prevent stress-induced apoptosis, with little attention paid to senescence-another common stress reaction of MSCs. Here, we proposed the strategy to prevent oxidative stress-induced senescence of human endometrial stem cells (hMESCs) based on deferoxamine (DFO) application. DFO prevented DNA damage and stress-induced senescence of hMESCs, as evidenced by reduced levels of reactive oxygen species, lipofuscin, cyclin D1, decreased SA-ß-Gal activity, and improved mitochondrial function. Additionally, DFO caused accumulation of HIF-1α, which may contribute to the survival of H2O2-treated cells. Importantly, cells that escaped senescence due to DFO preconditioning preserved all the properties of the initial hMESCs. Therefore, once protecting cells from oxidative damage, DFO did not alter further hMESCs functioning. The data obtained may become the important prerequisite for development of a new strategy in regenerative therapy based on MSCs preconditioning using DFO.


Deferoxamine/pharmacology , Endometrium/drug effects , Inflammation/drug therapy , Oxidative Stress/drug effects , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line , Cellular Microenvironment/drug effects , Cellular Senescence/drug effects , Cyclin D1/genetics , Endometrium/cytology , Endometrium/growth & development , Female , Gene Expression Regulation, Developmental/drug effects , Humans , Hydrogen Peroxide/toxicity , Inflammation/chemically induced , Inflammation/pathology , Lipofuscin/genetics , Mesenchymal Stem Cells/drug effects , Reactive Oxygen Species , Regenerative Medicine , Signal Transduction/drug effects
12.
Front Immunol ; 12: 697362, 2021.
Article En | MEDLINE | ID: mdl-34234785

Hepatic injury induced by ischemia and reperfusion (HIRI) is a major clinical problem after liver resection or transplantation. The polarization of macrophages plays an important role in regulating the severity of hepatic ischemia/reperfusion injury. Recent evidence had indicated that the ischemia induces an acidic microenvironment by causing increased anaerobic glycolysis and accumulation of lactic acid. We hypothesize that the acidic microenvironment might cause the imbalance of intrahepatic immunity which aggravated HIRI. The hepatic ischemia/reperfusion injury model was established to investigate the effect of the acidic microenvironment to liver injury. Liposomes were used to deplete macrophages in vivo. Macrophages were cultured under low pH conditions to analyze the polarization of macrophages in vitro. Activation of the PPAR-γ signal was determined by Western blot. PPAR-γ agonist GW1929 was administrated to functionally test the role of PPAR-γ in regulating macrophage-mediated effects in the acidic microenvironment during HIRI. We demonstrate that acidic microenvironment aggravated HIRI while NaHCO3 reduced liver injury through neutralizing the acid, besides, liposome abolished the protective ability of NaHCO3 through depleting the macrophages. In vivo and vitro experiment showed that acidic microenvironment markedly promoted M1 polarization but inhibited M2 polarization of macrophage. Furthermore, the mechanistic study proved that the PPAR-γ signal was suppressed during the polarization of macrophages under pH = 6.5 culture media. The addition of PPAR-γ agonist GW1929 inhibited M1 polarization under acidic environment and reduced HIRI. Our results indicate that acidic microenvironment is a key regulator in HIRI which promoted M1 polarization of macrophages through regulating PPAR-γ. Conversely, PPAR-γ activation reduced liver injury, which provides a novel therapeutic concept to prevent HIRI.


Liver/injuries , Liver/metabolism , Macrophages/metabolism , PPAR gamma/metabolism , Reperfusion Injury/metabolism , Animals , Benzophenones/administration & dosage , Cells, Cultured , Cellular Microenvironment/drug effects , Cellular Microenvironment/physiology , Disease Models, Animal , Hydrogen-Ion Concentration , Immunity, Innate/drug effects , Kupffer Cells/drug effects , Kupffer Cells/immunology , Kupffer Cells/metabolism , Liver/pathology , Macrophages/classification , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , PPAR gamma/agonists , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Severity of Illness Index , Signal Transduction , Sodium Bicarbonate/pharmacology , Tyrosine/administration & dosage , Tyrosine/analogs & derivatives
13.
PLoS One ; 16(7): e0253149, 2021.
Article En | MEDLINE | ID: mdl-34242238

A major obstacle for tissue engineering ear-shaped cartilage is poorly developed tissue comprising cell-scaffold constructs. To address this issue, bioresorbable scaffolds of poly-ε-caprolactone (PCL) and polyglycolic acid nanofibers (nanoPGA) were evaluated using an ethanol treatment step before auricular chondrocyte scaffold seeding, an approach considered to enhance scaffold hydrophilicity and cartilage regeneration. Auricular chondrocytes were isolated from canine ears and human surgical samples discarded during otoplasty, including microtia reconstruction. Canine chondrocytes were seeded onto PCL and nanoPGA sheets either with or without ethanol treatment to examine cellular adhesion in vitro. Human chondrocytes were seeded onto three-dimensional bioresorbable composite scaffolds (PCL with surface coverage of nanoPGA) either with or without ethanol treatment and then implanted into athymic mice for 10 and 20 weeks. On construct retrieval, scanning electron microscopy showed canine auricular chondrocytes seeded onto ethanol-treated scaffolds in vitro developed extended cell processes contacting scaffold surfaces, a result suggesting cell-scaffold adhesion and a favorable microenvironment compared to the same cells with limited processes over untreated scaffolds. Adhesion of canine chondrocytes was statistically significantly greater (p ≤ 0.05) for ethanol-treated compared to untreated scaffold sheets. After implantation for 10 weeks, constructs of human auricular chondrocytes seeded onto ethanol-treated scaffolds were covered with glossy cartilage while constructs consisting of the same cells seeded onto untreated scaffolds revealed sparse connective tissue and cartilage regeneration. Following 10 weeks of implantation, RT-qPCR analyses of chondrocytes grown on ethanol-treated scaffolds showed greater expression levels for several cartilage-related genes compared to cells developed on untreated scaffolds with statistically significantly increased SRY-box transcription factor 5 (SOX5) and decreased interleukin-1α (inflammation-related) expression levels (p ≤ 0.05). Ethanol treatment of scaffolds led to increased cartilage production for 20- compared to 10-week constructs. While hydrophilicity of scaffolds was not assessed directly in the present findings, a possible factor supporting the summary data is that hydrophilicity may be enhanced for ethanol-treated nanoPGA/PCL scaffolds, an effect leading to improvement of chondrocyte adhesion, the cellular microenvironment and cartilage regeneration in tissue-engineered auricle constructs.


Cellular Microenvironment/drug effects , Chondrocytes/drug effects , Chondrogenesis/drug effects , Ear Auricle/drug effects , Ethanol/pharmacology , Animals , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cells, Cultured , Congenital Microtia/drug therapy , Dogs , Ear Cartilage/drug effects , Ear, External/drug effects , Female , Humans , Male , Mice , Mice, Nude , Nanofibers/chemistry , Polyglycolic Acid/chemistry , Tissue Engineering/methods , Tissue Scaffolds
14.
Int J Biol Macromol ; 182: 1941-1952, 2021 Jul 01.
Article En | MEDLINE | ID: mdl-34062160

Achieving chondrogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) successfully is crucial for cartilage regeneration. To date, various hydrogels with different chemical microenvironment have been used to modulate chondrogenic differentiation of BMSCs, especially collagen and hyaluronic acid hydrogel. However, the chondro-inductive ability of collagen and hyaluronic acid hydrogel has not been evaluated yet and the different chemical and physical microenvironment of these two hydrogels increase the difficulty of comparison. In this study, three different hydrogels based on collagen and hyaluronic acid (self-assembled collagen hydrogel (Col), self-assembled collagen hydrogel cross-linked with genipin (Cgp), and methacrylated hyaluronic acid hydrogel (HA)) were prepared and their chondro-inductive ability on the encapsulated BMSCs was evaluated. Col and Cgp have the same chemical composition and similar microstructure, but are different from HA, while Cgp and HA hydrogels have the same mechanical strength. It was found that chemical and physical microenvironments of the hydrogels combined to influence cell condensation. Thanks to cell condensation was more likely to occur in collagen hydrogels in the early stage, the cartilage-induced ability was in the order of Col > Cgp > HA. However, the severe shrinkage of Col and Cgp resulted in no enough space for cell proliferation within hydrogels in the later stage. In contrast, relatively stable physical microenvironment of HA helped to maintain continuous production of cartilage-related matrix in the later stage. Overall, these results revealed that the chondro-inductive ability of collagen and hyaluronic acid hydrogel with different chemical and physical microenvironment cannot be evaluated by a particular time period. However, it provided important information for optimization and design of the future hydrogels towards successful repair of articular cartilage.


Cellular Microenvironment/drug effects , Chondrocytes/cytology , Collagen/pharmacology , Hyaluronic Acid/pharmacology , Hydrogels/pharmacology , Adsorption , Animals , Biomechanical Phenomena , Blood Proteins/metabolism , Cattle , Cell Differentiation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Chondrocytes/drug effects , Chondrogenesis/drug effects , Diffusion , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gene Expression Regulation/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Rabbits , Static Electricity , Transforming Growth Factor beta1/metabolism
15.
Int J Nanomedicine ; 16: 3509-3540, 2021.
Article En | MEDLINE | ID: mdl-34045855

The increasing incidence of bone-related disorders is causing a burden on the clinical scenario. Even though bone is one of the tissues that possess tremendous regenerative potential, certain bone anomalies need therapeutic intervention through appropriate delivery of a drug. Among several nanosystems and biologics that offer the potential to contribute towards bone healing, the exosomes from the class of extracellular vesicles are outstanding. Exosomes are extracellular nanovesicles that, apart from the various advantages, are standing out of the crowd for their ability to conduct cellular communication. The internal cargo of the exosomes is leading to its potential use in therapeutics. Exosomes are being unraveled in terms of the mechanism as well as application in targeting various diseases and tissues. Through this review, we have tried to understand and review all that is already established and the gap areas that still exist in utilizing them as drug delivery vehicles targeting the bone. The review highlights the potential of the exosomes towards their contribution to the drug delivery scenario in the bone microenvironment. A comparison of the pros and cons of exosomes with other prevalent drug delivery systems is also done. A section on the patents that have been generated so far from this field is included.


Bone and Bones/cytology , Cellular Microenvironment/drug effects , Drug Delivery Systems/methods , Exosomes/metabolism , Animals , Exosomes/drug effects , Humans
16.
Pediatr Rheumatol Online J ; 19(1): 72, 2021 May 12.
Article En | MEDLINE | ID: mdl-33980237

BACKGROUND: We examined influences of conditioned media from chondrocytes (Ch) on juvenile idiopathic arthritis synovial fibroblasts (JFLS) and potential for JFLS to undergo endochondral bone formation (EBF). METHODS: Primary cells from three control fibroblast-like synoviocytes (CFLS) and three JFLS were cultured in Ch-conditioned media and compared with untreated fibroblast-like synoviocytes (FLS). RNA was analyzed by ClariomS microarray. FLS cells cultured in conditioned media were exposed to either TGFBR1 inhibitor LY3200882 or exogenous BMP4 and compared with FLS cultured in conditioned media from Ch (JFLS-Ch). Media supernatants were analyzed by ELISA. RESULTS: In culture, JFLS downregulate BMP2 and its receptor BMPR1a while upregulating BMP antagonists (NOG and CHRD) and express genes (MMP9, PCNA, MMP12) and proteins (COL2, COLX, COMP) associated with chondrocytes. Important TGFß superfamily member gene expression (TGFBI, MMP9, COL1A1, SOX6, and MMP2) is downregulated when JFLS are cultured in Ch-conditioned media. COL2, COLX and COMP protein expression decreases in JFLS-Ch. BMP antagonist protein (NOG, CHRD, GREM, and FST) secretion is significantly increased in JFLS-Ch. Protein phosphorylation increases in JFLS-Ch exposed to exogenous BMP4, and chondrocyte-like phenotype is restored in BMP4 presence, evidenced by increased secretion of COL2 and COLX. Inhibition of TGFBR1 in JFLS-Ch results in overexpression of COL2. CONCLUSIONS: JFLS are chondrocyte-like, and Ch-conditioned media can abrogate this phenotype. The addition of exogenous BMP4 causes JFLS-Ch to restore this chondrocyte-like phenotype, suggesting that JFLS create a microenvironment favorable for endochondral bone formation, thereby contributing to joint growth disturbances in juvenile idiopathic arthritis.


Bone Morphogenetic Protein 4 , Growth Disorders , Osteogenesis , Receptor, Transforming Growth Factor-beta Type I , Synoviocytes/metabolism , TGF-beta Superfamily Proteins/metabolism , Arthritis, Juvenile/complications , Arthritis, Juvenile/metabolism , Bone Morphogenetic Protein 4/antagonists & inhibitors , Bone Morphogenetic Protein 4/metabolism , Cells, Cultured , Cellular Microenvironment/drug effects , Cellular Microenvironment/physiology , Chondrocytes/physiology , Culture Media, Conditioned/pharmacology , Gene Expression Regulation , Growth Disorders/etiology , Growth Disorders/metabolism , Humans , Osteogenesis/drug effects , Osteogenesis/physiology , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/metabolism , Signal Transduction/drug effects
17.
Stem Cell Reports ; 16(9): 2169-2181, 2021 09 14.
Article En | MEDLINE | ID: mdl-34019816

Duchenne muscular dystrophy (DMD) is a rare X-linked recessive disease that is associated with severe progressive muscle degeneration culminating in death due to cardiorespiratory failure. We previously observed an unexpected proliferation-independent telomere shortening in cardiomyocytes of a DMD mouse model. Here, we provide mechanistic insights using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Using traction force microscopy, we show that DMD hiPSC-CMs exhibit deficits in force generation on fibrotic-like bioengineered hydrogels, aberrant calcium handling, and increased reactive oxygen species levels. Furthermore, we observed a progressive post-mitotic telomere shortening in DMD hiPSC-CMs coincident with downregulation of shelterin complex, telomere capping proteins, and activation of the p53 DNA damage response. This telomere shortening is blocked by blebbistatin, which inhibits contraction in DMD cardiomyocytes. Our studies underscore the role of fibrotic stiffening in the etiology of DMD cardiomyopathy. In addition, our data indicate that telomere shortening is progressive, contraction dependent, and mechanosensitive, and suggest points of therapeutic intervention.


Muscular Dystrophies/genetics , Muscular Dystrophies/physiopathology , Myocardial Contraction/genetics , Myocytes, Cardiac/metabolism , Telomere Shortening/genetics , Biomarkers , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Cell Differentiation , Cells, Cultured , Cellular Microenvironment/drug effects , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Fibrosis , Fluorescent Antibody Technique , Gene Expression , Humans , Immunophenotyping , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mechanical Phenomena , Muscular Dystrophies/pathology , Muscular Dystrophy, Duchenne/etiology , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Myocardial Contraction/drug effects
18.
Int J Mol Sci ; 22(8)2021 Apr 13.
Article En | MEDLINE | ID: mdl-33924360

Endometriosis is a common gynecological disease. Here, we aimed to investigate the anti-fibrotic, anti-inflammatory, and anti-oxidative role of the methyl ester of 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO-Me) on endometriosis. An endometriosis rat model was constructed by intraperitoneally injecting recipient rats with an equivalent of tissue from the uterus of a donor animal. Endometriosis was allowed to develop for seven days. CDDO-Me was administered on the 7th day and for the next 7 days. On day 14, rats were sacrificed, and peritoneal fluid and endometriotic implants were collected. CDDO-Me displayed antioxidant activity by activating the Nfr2 pathway and the expression of antioxidant mediators such as NQO-1 and HO-1. Moreover, it reduced lipid peroxidation and increased glutathione (GSH) levels and superoxide dismutase (SOD) activity. CDDO-Me also showed anti-inflammatory activity by decreasing the expression of pro-inflammatory cytokines in peritoneal fluids and NFkB activation. It, in turn, reduced cyclooxygenase-2 (COX-2) expression in the endometriotic loci and prostaglandin E2 (PGE2) levels in the peritoneal fluids, leading to increased apoptosis and reduced angiogenesis. The reduced oxidative stress and pro-inflammatory microenvironment decreased implants diameter, area, and volume. In particular, CDDO-Me administration reduced the histopathological signs of endometriosis and inflammatory cells recruitment into the lesions, as shown by toluidine blue staining and myeloperoxidase (MPO) activity. CDDO-Me strongly suppressed α-SMA and fibronectin expression and collagen deposition, reducing endometriosis-associated fibrosis. In conclusion, CDDO-Me treatment resulted in a coordinated and effective suppression of endometriosis by modulating the Nrf2 and NFkB pathways.


Endometriosis/drug therapy , Endometriosis/pathology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Oleanolic Acid/analogs & derivatives , Signal Transduction , Animals , Apoptosis/drug effects , Cellular Microenvironment/drug effects , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Female , Fibrosis , Inflammation/pathology , Neovascularization, Physiologic/drug effects , Oleanolic Acid/administration & dosage , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , Oxidation-Reduction , Oxidative Stress , Rats, Sprague-Dawley
19.
Biomed Res Int ; 2021: 6625952, 2021.
Article En | MEDLINE | ID: mdl-33880372

The liver is the primary target organ for perfluorooctane sulphonate (PFOS), a recently discovered persistent organic pollutant. However, the mechanisms mediating hepatotoxicity remain unclear. Herein, we explored the relationship between reactive oxygen species (ROS) and autophagy and apoptosis induced by PFOS in L-02 cells, which are incubated with different concentrations of PFOS (0, 50, 100, 150, 200, or 250 µmol/L) for 24 or 48 hrs at 37°C. The results indicated that PFOS exposure decreased cell activities, enhanced ROS levels in a concentration-dependent manner, decreased mitochondrial membrane potential (MMP), and induced autophagy and apoptosis. Compared with the control, 200 µmol/L PFOS increased ROS levels; enhanced the expression of Bax, cleaved-caspase-3, and LC3-II; induced autophagy; decreased MMP; and lowered Bcl-2, p62, and Bcl-2/Bax ratio. The antioxidant N-acetyl cysteine (NAC) protected MMP against PFOS-induced changes and diminished apoptosis and autophagy. Compared with 200 µmol/L PFOS treatment, NAC pretreatment reversed the increase in ROS, Bax, and cleaved-caspase-3 protein caused by PFOS, lowered the apoptosis rate increased by PFOS, and increased the levels of MMP and Bcl-2/Bax ratio decreased by PFOS. The autophagy inhibitor 3-methyladenine and chloroquine decreased apoptosis and cleaved-caspase-3 protein level and increased the Bcl-2/Bax ratio. In summary, our results suggest that ROS-triggered autophagy is involved in PFOS-induced apoptosis in L-02 cells.


Alkanesulfonic Acids/pharmacology , Apoptosis , Autophagy , Embryo, Mammalian/pathology , Fluorocarbons/pharmacology , Liver/embryology , Liver/pathology , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cadaverine/pharmacology , Cell Line , Cell Survival/drug effects , Cellular Microenvironment/drug effects , Humans , Liver/drug effects , Membrane Potential, Mitochondrial/drug effects , Proteins/metabolism , Vacuoles/drug effects , Vacuoles/metabolism
20.
Biomed Pharmacother ; 139: 111545, 2021 Jul.
Article En | MEDLINE | ID: mdl-33873145

Tacrolimus (TAC, FK506) is a major calcineurin inhibitor and has been commonly used in treatments of patients with organ transplants and immune diseases. Moreover, tacrolimus is recommended by the treatment guidelines for oral potentially malignant disorders (OPMDs) such as oral lichen planus (OLP). However, whether tacrolimus increases the risk of cancer remains controversial. We observed that in a 4-Nitroquinoline N-oxide (4NQO)-induced oral carcinogenesis model, tacrolimus treatment was associated with a significantly lower ratio of cancer formation (52.94% vs. 90%) and a lower proportion of Ki67 and proliferation cell nuclear antigen (PCNA) -positive cells in lesion areas (P < 0.001). Liver, kidney, and lung functions of rats and the tumor immune microenvironment of the tongue were not affected. These observations suggest that tacrolimus blocked oral carcinogenesis through epithelial cell proliferation inhibition, independent of its immunosuppressive effects. As a processing factor, tacrolimus decreased tumor formation and cell proliferation in different stages of oral squamous cell carcinoma (OSCC) progression in vivo and in vitro. Furthermore, we investigated effects on the cell cycle and expression of related proteins. Tacrolimus induced G1/S phase arrest and significantly downregulated the expression of cyclinD1, cyclinE1, and c-Myc. These results suggest that tacrolimus induces G1/S phase arrest via inhibition of cyclinD1, cyclinE1, and c-Myc expression and retards oral cell carcinogenesis in vitro and in vivo. Thus, application of tacrolimus is a safe therapeutic strategy for treating OPMDs.


Anticarcinogenic Agents/pharmacology , Cell Cycle/drug effects , Mouth Neoplasms/prevention & control , Tacrolimus/pharmacology , 4-Nitroquinoline-1-oxide , Animals , Carcinogens , Cellular Microenvironment/drug effects , Cyclins/antagonists & inhibitors , Cyclins/biosynthesis , Genes, myc/drug effects , Ki-67 Antigen , Male , Mice , Mice, Inbred BALB C , Mouth Neoplasms/chemically induced , Mouth Neoplasms/pathology , Proliferating Cell Nuclear Antigen/analysis , Rats , Rats, Sprague-Dawley , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/prevention & control , Xenograft Model Antitumor Assays
...