Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Exp Clin Cancer Res ; 43(1): 46, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38342894

ABSTRACT

BACKGROUND: Nasopharyngeal carcinoma (NPC) poses a significant health burden in specific regions of Asia, and some of NPC patients have bone metastases at the time of initial diagnosis. Bone metastasis can cause pathologic fractures and pain, reducing patients' quality of life, and is associated with worse survival. This study aims to unravel the complex role of insulin-like growth factor 1 receptor (IGF-1R) in NPC bone metastasis, offering insights into potential therapeutic targets. METHODS: We assessed IGF-1R expression in NPC cells and explored its correlation with bone metastasis. Experiments investigated the impact of osteoclast-secreted IGF-1 on the IGF-1R/AKT/S6 pathway in promoting NPC cell proliferation within the bone marrow. Additionally, the reciprocal influence of tumor-secreted Granulocyte-macrophage colony-stimulating factor (GM-CSF) on osteoclast differentiation and bone resorption was examined. The effects of IGF-1 neutralizing antibody, IGF-1R specific inhibitor (NVP-AEW541) and mTORC inhibitor (rapamycin) on nasopharyngeal carcinoma bone metastasis were also explored in animal experiments. RESULTS: Elevated IGF-1R expression in NPC cells correlated with an increased tendency for bone metastasis. IGF-1, secreted by osteoclasts, activated the IGF-1R/AKT/S6 pathway, promoting NPC cell proliferation in the bone marrow. Tumor-secreted GM-CSF further stimulated osteoclast differentiation, exacerbating bone resorption. The IGF-1 neutralizing antibody, NVP-AEW541 and rapamycin were respectively effective in slowing down the rate of bone metastasis and reducing bone destruction. CONCLUSION: The intricate interplay among IGF-1R, IGF-1, and GM-CSF highlights potential therapeutic targets for precise control of NPC bone metastasis, providing valuable insights for developing targeted interventions.


Subject(s)
Bone Neoplasms , Bone Resorption , Nasopharyngeal Neoplasms , Animals , Humans , Nasopharyngeal Carcinoma/pathology , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor I/therapeutic use , Osteoclasts/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Quality of Life , Cell Line, Tumor , Nasopharyngeal Neoplasms/pathology , Sirolimus/pharmacology , Antibodies, Neutralizing
2.
Mol Ther ; 31(1): 174-192, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36104974

ABSTRACT

There is no effective therapy for implant-associated Staphylococcus aureus osteomyelitis, a devastating complication after orthopedic surgery. An immune-suppressive profile with up-regulated programmed cell death 1/programmed death ligand 1 (PD-1/PD-L1) was identified based on our transcriptional data (GEO: GSE166522) from a mouse model of S. aureus osteomyelitis. PD-1/PD-L1 expression was up-regulated mainly in F4/80+ macrophages surrounding the abscess in S. aureus-infected bone. Mechanistically, PD-1/PD-L1 activated mitophagy to suppress production of mitochondrial reactive oxygen species (ROS), suppressing the bactericidal function of macrophages. Using neutralizing antibodies for PD-L1 or PD-1, or knockout of PD-L1 adjuvant to gentamicin markedly reduced mitophagy in bone marrow F4/80+ cells, enhanced bacterial clearance in bone tissue and implants, and reduced bone destruction in mice. PD-1/PD-L1 expression was also increased in the bone marrow from individuals with S. aureus osteomyelitis. These findings uncover a so far unknown function of PD-1/PD-L1-mediated mitophagy in suppressing the bactericidal function of bone marrow macrophages.


Subject(s)
Antibodies , B7-H1 Antigen , Osteomyelitis , Programmed Cell Death 1 Receptor , Animals , Mice , Adjuvants, Immunologic , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , Osteomyelitis/metabolism , Osteomyelitis/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Staphylococcus aureus , Disease Models, Animal , Antibodies/therapeutic use
3.
Antioxidants (Basel) ; 11(11)2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36421451

ABSTRACT

Heterotopic ossification (HO) is defined as the generation of pathological ectopic bony structures in soft tissues, but the molecular mechanisms of tendon HO are not fully revealed. Hedgehog (Hh) signalling is reportedly critical in hereditary HO. Our study focuses on the role of Hh signalling in the formation of trauma-induced tendon ossification. In this study, samples of healthy tendons and injured tendons from C57BL/6J female mice at 1, 4, 7, and 10 weeks after Achilles tenotomy were collected for quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemical analysis (IHC). At 1, 4, 7, and 10 weeks postinjury, tendon samples from the mice administered with vehicle, GANT58 (a GLI antagonist), or SAG (a smoothened agonist) were harvested for micro-CT, histological staining, qRT-PCR, and IHC. Rat tendon-derived stem cells (TDSCs) treated with vehicle, GANT58, or SAG were used to induce osteogenic and chondrogenic differentiation in vitro for qRT-PCR, alkaline phosphatase staining, Alcian blue staining, and reactive oxygen species (ROS) levels measurement. We found that Hh signalling is remarkably activated during the formation of trauma-induced tendon ossification in the model of Achilles tenotomy. The in vitro and in vivo assays both confirm that downregulation of Hh signalling significantly suppresses osteogenesis and chondrogenesis to inhibit tendon ossification, while upregulation of Hh signalling promotes this process. Under osteogenic induction, Hh signalling regulates antioxidant pathway and affects ROS generation of TDSCs. Collectively, Hh signalling contributes to trauma-induced tendon ossification and affects ROS generation through antioxidant pathway in osteogenic differentiation of TDSCs, indicating that targeting Hh signalling by GANT58 may be a potential treatment for trauma-induced tendon ossification.

4.
Bone Res ; 10(1): 58, 2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36031625

ABSTRACT

The mechanisms that coordinate the shift from joint homeostasis to osteoarthritis (OA) remain unknown. No pharmacological intervention can currently prevent the progression of osteoarthritis. Accumulating evidence has shown that subchondral bone deterioration is a primary trigger for overlying cartilage degeneration. We previously found that H-type vessels modulate aberrant subchondral bone formation during the pathogenesis of OA. However, the mechanism responsible for the elevation of H-type vessels in OA is still unclear. Here, we found that PDGFR-ß expression, predominantly in the CD31hiEmcnhi endothelium, was substantially elevated in subchondral bones from OA patients and rodent OA models. A mouse model of OA with deletion of PDGFR-ß in endothelial cells (ECs) exhibited fewer H-type vessels, ameliorated subchondral bone deterioration and alleviated overlying cartilage degeneration. Endothelial PDGFR-ß promotes angiogenesis through the formation of the PDGFR-ß/talin1/FAK complex. Notably, endothelium-specific inhibition of PDGFR-ß by local injection of AAV9 in subchondral bone effectively attenuated the pathogenesis of OA compared with that of the vehicle-treated controls. Based on the results from this study, targeting PDGFR-ß is a novel and promising approach for the prevention or early treatment of OA.

6.
Front Microbiol ; 11: 1301, 2020.
Article in English | MEDLINE | ID: mdl-32595631

ABSTRACT

Staphylococcus aureus (S. aureus) infection-induced osteomyelitis is a great challenge in clinic treatment. Identification of the essential genes and biological processes that are specifically changed in mononuclear cells at an early stage of S. aureus osteomyelitis is of great clinical significance. Based on transcriptional dataset GSE16129 available publicly, a bioinformatic analysis was performed to identify the differentially expressed genes of osteomyelitis caused by S. aureus infection. ERBB2, TWIST1, and NANOG were screened out as the most valuable osteomyelitis-related genes (OMRGs). A mice model of implant-associated S. aureus osteomyelitis was used to verify the above genes. We found significantly up-regulated expression of TWIST1 in macrophages and accumulation of macrophages around the infected implant. Meanwhile, S. aureus infection increased the expression of TWIST1, MMP9, and MMP13, and stimulated the migration and phagocytosis function of Raw 264.7 cells. Additionally, knock-down of the expression of TWIST1 by siRNA could significantly down-regulate MMP9 and MMP13 and suppress the migration and phagocytosis ability of macrophages in response to S. aureus infection. Furthermore, we found that NF-κB signaling was activated in Raw 264.7 cells by S. aureus and that inhibition of NF-κB signaling by Bay11-7082 blocked the expression of TWIST1, MMP9, and MMP13 as well as cell migration and phagocytosis evoked by S. aureus. Our findings demonstrate that NF-κB/TWIST1 is necessary for migration and phagocytosis of macrophages in response to S. aureus infection. Our study highlights the essential role of NF-κB/TWIST1 in early innate immune response to S. aureus infection in bone.

7.
J Cell Physiol ; 235(11): 8653-8666, 2020 11.
Article in English | MEDLINE | ID: mdl-32324278

ABSTRACT

Osteoarthritis (OA), a disease of the entire joint, is characterized by abnormal bone remodeling and coalescent degradation of articular cartilage. We have previously found that elevated levels of H-type vessels in subchondral bone correlate with OA and that focal adhesion kinase (FAK) is critical for H-type vessel formation in osteoporosis. However, the potential role of FAK in OA remains unexplored. Here, we demonstrate that the p-FAK level was dramatically elevated in subchondral bone following anterior cruciate ligament transection (ACLT) in rats. Specific inhibition of FAK signaling with Y15 in subchondral bone resulted in the suppression of subchondral bone deterioration and this effect was mediated by H-type vessel-induced ectopic bone formation. Further, articular cartilage degeneration was also alleviated after Y15 treatment. In vitro, the p-FAK level was significantly elevated in mesenchymal stem cells (MSCs) from vehicle-treated ACLT rats as compared to that in MSCs from sham controls and Y15-treated ACLT rats. Elevated p-FAK level in MSCs promoted vascular endothelial growth factor (VEGF) expression, as demonstrated from the high VEGF level in the blood, subchondral bone, and conditioned medium (CM) of MSCs from vehicle-treated ACLT rats. The CM of MSCs from vehicle-treated ACLT rats might promote the angiogenesis of endothelial cells and the catabolic response of chondrocytes through the FAK-growth factor receptor-bound protein 2-mitogen-activated protein kinase-mediated expression of VEGF. The effect of the CM from MSCs of Y15-treated ACLT rats or that treated with a VEGF-neutralizing antibody on vessel formation and the catabolic response was lowered. Thus, the specific inhibition of FAK signaling may be a promising avenue for the prevention or early treatment of OA.


Subject(s)
Cartilage, Articular/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/drug effects , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Osteoarthritis/drug therapy , Alendronate/pharmacology , Animals , Anterior Cruciate Ligament/pathology , Bone Remodeling/drug effects , Bone Remodeling/physiology , Bone and Bones/pathology , Chondrocytes/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Male , Osteoarthritis/pathology , Rats, Sprague-Dawley
8.
Stem Cell Res Ther ; 11(1): 131, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32197645

ABSTRACT

BACKGROUND: Hypercholesterolemia increases the risk of tendon pain and tendon rupture. Tendon-derived stem cells (TDSCs) play a vital role in the development of tendinopathy. Our previous research found that high cholesterol inhibits tendon-related gene expression in TDSCs. Whether high cholesterol has other biological effects on TDSCs remains unknown. METHODS: TDSCs isolated from female SD rats were exposed to 10 mg/dL cholesterol for 24 h. Then, cell apoptosis was assessed using flow cytometry and fluorescence microscope. RFP-GFP-LC3 adenovirus transfection was used for measuring autophagy. Signaling transduction was measured by immunofluorescence and immunoblotting. In addition, Achilles tendons from ApoE -/- mice fed with a high-fat diet were histologically assessed using HE staining and immunohistochemistry. RESULTS: In this work, we verified that 10 mg/dL cholesterol suppressed cell proliferation and migration and induced G0/G1 phase arrest. Additionally, cholesterol induced apoptosis and autophagy simultaneously in TDSCs. Apoptosis induction was related to increased expression of cleaved caspase-3 and BAX and decreased expression of Bcl-xL. The occurrence of autophagic flux and accumulation of LC3-II demonstrated the induction of autophagy by cholesterol. Compared with the effects of cholesterol treatment alone, the autophagy inhibitor 3-methyladenine (3-MA) enhanced apoptosis, while the apoptosis inhibitor Z-VAD-FMK diminished cholesterol-induced autophagy. Moreover, cholesterol triggered reactive oxygen species (ROS) generation and activated the AKT/FOXO1 pathway, while the ROS scavenger NAC blocked cholesterol-induced activation of the AKT/FOXO1 pathway. NAC and the FOXO1 inhibitor AS1842856 rescued the apoptosis and autophagy induced by cholesterol. Finally, high cholesterol elevated the expression of cleaved caspase-3, Bax, LC3-II, and FOXO1 in vivo. CONCLUSION: The present study indicated that high cholesterol induced apoptosis and autophagy through ROS-activated AKT/FOXO1 signaling in TDSCs, providing new insights into the mechanism of hypercholesterolemia-induced tendinopathy. High cholesterol induces apoptosis and autophagy through the ROS-activated AKT/FOXO1 pathway in tendon-derived stem cells.


Subject(s)
Hypercholesterolemia , Proto-Oncogene Proteins c-akt , Animals , Apoptosis , Autophagy , Cell Line, Tumor , Cholesterol , Female , Forkhead Box Protein O1 , Mice , Nerve Tissue Proteins , Proto-Oncogene Proteins c-akt/genetics , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species , Stem Cells , Tendons
9.
FASEB J ; 33(10): 11137-11147, 2019 10.
Article in English | MEDLINE | ID: mdl-31298955

ABSTRACT

So far, there has been no effective cure for osteoporotic cortical bone, the most significant change in long bone structure during aging and the main cause of bone fragility fractures, because its underlying molecular and cellular mechanisms remain largely unknown. We used 3- and 15-mo-old mice as well as 15-mo-old mice treated with vehicle and gefitinib to evaluate structural, cellular, and molecular changes in cortical bone. We found that the senescence of osteoprogenitors was increased, whereas the expression of phosphorylated epidermal growth factor receptor (EGFR) on the endosteal surface of cortical bone down-regulated in middle-aged 15-mo-old mice compared with young 3-mo-old mice. Further decreasing EGFR signaling by gefitinib treatment in middle-aged mice resulted in promoted senescence of osteoprogenitors and accelerated cortical bone degeneration. Moreover, inhibiting EGFR signaling suppressed the expression of enhancer of zeste homolog 2 (Ezh2), the repressor of cell senescence-inducer genes, through ERK1/2 pathway, thereby promoting senescence in osteoprogenitors. Down-regulated EGFR signaling plays a physiologically significant role during aging by reducing Ezh2 expression, leading to the senescence of osteoprogenitors and the decline in bone formation on the endosteal surface of cortical bone.-Liu, G., Xie, Y., Su, J., Qin, H., Wu, H., Li, K., Yu, B., Zhang, X. The role of EGFR signaling in age-related osteoporosis in mouse cortical bone.


Subject(s)
Cortical Bone/metabolism , ErbB Receptors/metabolism , Osteoporosis/metabolism , Signal Transduction/physiology , Aging/metabolism , Animals , Cellular Senescence/physiology , Down-Regulation/physiology , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteogenesis/physiology
10.
J Cell Physiol ; 234(10): 18017-18028, 2019 08.
Article in English | MEDLINE | ID: mdl-30825206

ABSTRACT

Clinical studies have indicated that increased serum cholesterol levels raised the risk of tendinopathy in hypercholesterolemia, but the effect of cholesterol on tendon-derived stem cells (TDSCs) and its underlying mechanism have not been studied. The purpose of this study is to investigate the association between cholesterol and tendinopathy in vitro and in vivo, and its underlying molecular mechanism as well. In TDSCs, the effect of cholesterol was assessed by quantitative polymerase chain reaction, western blot analysis, and immunofluorescence staining. Intracellular levels of reactive oxygen species (ROS) was detected, using flow cytometry. The link between nuclear factor (NF)-κB signaling and the effect of cholesterol was evaluated using a representative IκB kinase (IKK) inhibitor, BAY 11-7082. In addition, Achilles tendons from apolipoprotein E mice fed with a high-fat diet were histologically assessed using hematoxylin and eosin staining and immunohistochemistry. We found that high cholesterol apparently lowered the expression of tendon cell markers (collagen 1, scleraxis, tenomodulin), and elevated ROS levels via the NF-κB pathway both in vitro and in vivo. The ROS scavenger N-acetylcysteine (NAC) and BAY 11-7082 reversed the inhibiting effect of cholesterol on the tendon-related gene expressions of TDSCs. Moreover, NAC blocked cholesterol-induced phosphorylation of IκBα and p65. Significant histological alternation in vivo was shown in Achilles tendon in the hypercholesterolemic group. These results indicated that high cholesterol may inhibit the tendon-related gene expressions in TDSCs via ROS-activated NF-кB signaling, implying pathogenesis of tendinopathy in hypercholesterolemia and suggesting a new mechanism underlying hypercholesterolemia-induced tendinopathy.


Subject(s)
Achilles Tendon/metabolism , Cholesterol/metabolism , Hypercholesterolemia/metabolism , NF-kappa B/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Stem Cells/metabolism , Tendinopathy/metabolism , Achilles Tendon/drug effects , Achilles Tendon/pathology , Animals , Antioxidants/pharmacology , Cells, Cultured , Disease Models, Animal , Female , Gene Expression Regulation , Hypercholesterolemia/drug therapy , Hypercholesterolemia/genetics , Hypercholesterolemia/pathology , Male , Mice, Knockout, ApoE , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Signal Transduction , Stem Cells/drug effects , Stem Cells/pathology , Tendinopathy/genetics , Tendinopathy/pathology , Tendinopathy/prevention & control
11.
Mol Med Rep ; 18(6): 5044-5052, 2018 12.
Article in English | MEDLINE | ID: mdl-30320384

ABSTRACT

Tendon repair follows a slow course of early inflammatory, proliferative and remodeling phases, which commonly results in the failure and loss of normal biomechanical properties. Previous studies have demonstrated that tendon­derived stem cells (TDSCs) are vital healing cells and that mRNA expression of anti­inflammatory cytokine interleukin (IL)­10 is significantly upregulated at the late inflammatory phase. To explore how IL­10 may impact tendon healing, the present study investigated the in vitro effects of IL­10 on TDSCs isolated from rat Achilles tendons. Cellular activities of TDSCs and the expression levels of tendon cell markers were measured treatment with IL­10 and subsequent performance of wound healing assays, reverse transcription­quantitative polymerase chain reaction and western blot analyses. The results demonstrated that IL­10 treatment markedly increased the proliferative capacity of TDSCs. In addition, IL­10 significantly enhanced cell migration when compared with the control cells. Furthermore, IL­10 treatment significantly activated the JAK/Stat3 signaling pathway and inhibited the protein expression of tendon cell markers, including scleraxis and tenomodulin. Notably, IL­10 treatment also reduced the gene expression levels of type 1 collagen, type 3 collagen, lumican and fibromodulin in TDSCs. These findings indicated that IL­10 enhanced cell proliferation and migration, and inhibited tenogenic differentiation in TDSCs in vitro. Reducing the negative effects whilst enhancing the positive effects of IL­10 may be a potential therapeutic target in tendon repair.


Subject(s)
Cell Differentiation , Interleukin-10/metabolism , Janus Kinases/metabolism , STAT3 Transcription Factor/metabolism , Stem Cells/metabolism , Tendons/cytology , Animals , Cell Cycle , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Female , Interleukin-10/pharmacology , Rats , Wound Healing
12.
Spectrochim Acta A Mol Biomol Spectrosc ; 205: 287-291, 2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30029191

ABSTRACT

A novel chemosensor 1 having a chiral macrocyclic dioxopolyamine of C2 symmetry as a receptor and anthracene as a signal unit has been designed and synthesized for cations and α-amino acids recognition in DMSO-HEPES buffer (1:9, v/v, pH 7.2). The ligand exhibited selective response to Cu2+ even in the presence of other metal ions with a fluorescence "switch-off" behavior. Additionally, the in situ generated 1-Cu2+ ensemble displayed specific recognition to histidine by a "switch-on" fluorescence response. For this dual functional switch, its sensing behavior via a displacement mode was confirmed by 1H NMR titration and ESI mass spectroscopy. Sequential "on-off-on" fluorescence responses of 1 to Cu2+ and histidine are successfully applied in HeLa cells.


Subject(s)
Copper/analysis , Fluorescent Dyes/chemistry , Histidine/analysis , Macrocyclic Compounds/chemistry , Spectrometry, Fluorescence/methods , HeLa Cells , Humans , Magnetic Resonance Spectroscopy , Spectrometry, Mass, Electrospray Ionization , Stereoisomerism
13.
Med Sci Monit ; 24: 1567-1573, 2018 Mar 16.
Article in English | MEDLINE | ID: mdl-29547593

ABSTRACT

BACKGROUND Previous studies demonstrated that tendon-derived stem cells (TDSCs) were vital healing cells and that mRNA expression of anti-inflammatory cytokine IL-6 was significantly upregulated in injured tendons. The aim of the present study was to investigate the effects of IL-6 on the TDSCs in vitro. MATERIAL AND METHODS TDSCs isolated from the Achilles tendons in SD rats were co-cultured with various concentrations of IL-6. Cell proliferation, cell cycle analysis, quantitative real-time PCR, western blotting analysis, and statistical analysis were used in the study. RESULTS The result showed that IL-6 strongly increased proliferation capability, and induced cell cycle activation and transition into G2/M phase from G1 phase in TDSCs. However, IL-6 treatment strongly inhibited gene expression of Scleraxis, Collagen 1, Tenomodulin, Collagen 3, Early Growth Response Protein 1, Decorin, Lumican, Biglycan and Fibromodulin in TDSCs. It also strongly inhibited protein expression of tendon cell markers like scleraxis, collagen 1, collagen 3, and tenomodulin. IL-6 treatment strongly activated the JAK/Stat3 signaling pathway in TDSCs. Furthermore, WP1066, a JAK/Stat3 signaling pathway inhibitor, abrogated the effects of IL-6 on TDSCs. CONCLUSIONS These findings indicated that IL-6 might exert dual effects on TDSCs in vitro: strongly enhancing their proliferation but inhibiting their tenogenic differentiation via the JAK/Stat3 pathway.


Subject(s)
Cell Differentiation/drug effects , Interleukin-6/pharmacology , Janus Kinases/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Tendons/cytology , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Female , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Stem Cells/drug effects
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(2): 155-161, 2018 Feb 20.
Article in Chinese | MEDLINE | ID: mdl-29502053

ABSTRACT

OBJECTIVE: To investigate the factors associated with the occurrence of transplant renal artery stenosis (TRAS). METHODS: A retrospective analysis was conducted in 26 recipients who developed TRAS and 40 concurrent renal recipients without TRAS. We also conducted a nested case-control study in 14 patients with TRAS (TRAS-SD group) and another 14 non-TRAS recipients who received the allograft from the same donor (non-TRAS-SD group). RESULTS: Compared with those in the concurrent recipients without TRAS, acute rejection (AR) occurred at a significantly higher incidence (P=0.004) and the warm ischemia time (WIT) was significantly longer (P=0.015) and the level of high?density lipoprotein cholesterol (HDL--C) significantly lower (P=0.009) in the recipients with TRAS. Logistic regression analysis suggested that AR (P=0.007) and prolonged WIT (P=0.046) were risk factors of TRAS while HDL-C (P=0.022) was the protective factor against TRAS. In recent years early diagnosis of TRAS had been made in increasing cases, the interval from transplantation to TRAS diagnosis became shortened steadily, and the recipients tended to have higher estimated glomerular filtration rate at the time of TRAS diagnosis. CONCLUSION: Apart from the surgical technique, AR and prolonged WIT are also risk factors of TRAS while a high HDL-C level is the protective factor against TRAS. The improvement of the diagnostic accuracy by ultrasound is the primary factor contributing to the increased rate of early TRAS diagnosis in recent years.


Subject(s)
Cholesterol, HDL/blood , Kidney Transplantation/adverse effects , Renal Artery Obstruction/physiopathology , Case-Control Studies , Graft Rejection/physiopathology , Humans , Protective Factors , Retrospective Studies , Treatment Outcome , Warm Ischemia
15.
PLoS One ; 10(9): e0138426, 2015.
Article in English | MEDLINE | ID: mdl-26382764

ABSTRACT

Influenza A viral (IAV) fusion peptides are known for their important role in viral-cell fusion process and membrane destabilization potential which are compatible with those of antimicrobial peptides. Thus, by replacing the negatively or neutrally charged residues of FPs with positively charged lysines, we synthesized several potent antimicrobial peptides derived from the fusogenic peptides (FPs) of hemagglutinin glycoproteins (HAs) of IAV. The biological screening identified that in addition to the potent antibacterial activities, these positively charged fusion peptides (pFPs) effectively inhibited the replication of influenza A viruses including oseltamivir-resistant strain. By employing pseudovirus-based entry inhibition assays including H5N1 influenza A virus (IAV), and VSV-G, the mechanism study indicated that the antiviral activity may be associated with the interactions between the HA2 subunit and pFP, of which, the nascent pFP exerted a strong effect to interrupt the conformational changes of HA2, thereby blocking the entry of viruses into host cells. In addition to providing new peptide "entry blockers", these data also demonstrate a useful strategy in designing potent antibacterial agents, as well as effective viral entry inhibitors. It would be meaningful in treatment of bacterial co-infection during influenza pandemic periods, as well as in our current war against those emerging pathogenic microorganisms such as IAV and HIV.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/metabolism , Viral Fusion Protein Inhibitors/pharmacology , Viral Fusion Proteins/metabolism , Virus Internalization/drug effects , Virus Replication/drug effects , Antiviral Agents/therapeutic use , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Influenza, Human/drug therapy , Viral Fusion Protein Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...