Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Reprod Biol ; 24(2): 100894, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38776742

ABSTRACT

Varicocele (VC) is a common cause of infertility in men. Pathophysiological changes caused by VC, such as testicular hypoxia, high temperatures, oxidative stress, abnormal reproductive hormones, and Cd accumulation, can induce autophagy, thus affecting the reproductive function in patients with this condition. Autophagy regulators can be classified as activators or inhibitors. Autophagy activators upregulate autophagy, reduce the damage to the testis and epididymis, inhibit spermatogenic cell apoptosis, and protect fertility. In contrast, autophagy inhibitors block autophagy and aggravate the damage to the reproductive functions. Therefore, elucidating the role of autophagy in the occurrence, development, and regulation of VC may provide additional therapeutic options for men with infertility and VC. In this review, we briefly describe the progress made in autophagy research in the context of VC.


Subject(s)
Autophagy , Varicocele , Autophagy/physiology , Varicocele/complications , Male , Humans , Animals , Infertility, Male/etiology , Testis
2.
J Cell Mol Med ; 28(1): e18009, 2024 01.
Article in English | MEDLINE | ID: mdl-37882107

ABSTRACT

The complex interplay between tumour cells and the tumour microenvironment (TME) underscores the necessity for gaining comprehensive insights into disease progression. This study centres on elucidating the elusive the elusive role of endothelial cells within the TME of head and neck squamous cell carcinoma (HNSCC). Despite their crucial involvement in angiogenesis and vascular function, the mechanistic diversity of endothelial cells among HNSCC patients remains largely uncharted. Leveraging advanced single-cell RNA sequencing (scRNA-Seq) technology and the Scissor algorithm, we aimed to bridge this knowledge gap and illuminate the intricate interplay between endothelial cells and patient prognosis within the context of HNSCC. Here, endothelial cells were categorized into Scissorhigh and Scissorlow subtypes. We identified Scissor+ endothelial cells exhibiting pro-tumorigenic profiles and constructed a prognostic risk model for HNSCC. Additionally, four biomarkers also were identified by analysing the gene expression profiles of patients with HNSCC and a prognostic risk prediction model was constructed based on these genes. Furthermore, the correlations between endothelial cells and prognosis of patients with HNSCC were analysed by integrating bulk and single-cell sequencing data, revealing a close association between SHSS and the overall survival (OS) of HNSCC patients with malignant endothelial cells. Finally, we validated the prognostic model by RT-qPCR and IHC analysis. These findings enhance our comprehension of TME heterogeneity at the single-cell level and provide a prognostic model for HNSCC.


Subject(s)
Endothelial Cells , Head and Neck Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck , Algorithms , Carcinogenesis , Tumor Microenvironment
3.
Stem Cell Res Ther ; 14(1): 211, 2023 08 21.
Article in English | MEDLINE | ID: mdl-37605271

ABSTRACT

BACKGROUND: The efficacy of human umbilical cord mesenchymal stem cell (hUC-MSC) transplantation in treating systemic lupus erythematosus (SLE) has been confirmed by small-scale clinical trials. However, these trials focused on severe or refractory SLE, while few studies focused on mild SLE. Therefore, this study focused on the therapeutic effects of hUC-MSC transplantation in early-stage or mild MRL/lpr lupus model mice. METHODS: Commercially available hUC-MSCs were transplanted into 8-week-old MRL/lpr mice by tail vein injection. Flow cytometry was used to analyze B cells and their subsets in the peripheral blood. Further, plasma inflammatory factors, autoantibodies, and plasma biochemical indices were detected using protein chip technology and ELISA kits. In addition, pathological staining and immunofluorescence were performed to detect kidney injury in mice. RESULTS: hUC-MSC transplantation did not affect the mice's body weight, and both middle and high dose hUC-MSC transplantation (MD and HD group) actually reduced spleen weight. hUC-MSC transplantation significantly decreased the proportion of plasmablasts (PB), IgG1- PB, IgG1+ PB, IgG1+ memory B (MB) cells, IgG1+ DN MB, and IgG1+ SP MB cells. The hUC-MSC transplantation had significantly reduced plasma levels of inflammatory factors, such as TNF-α, IFN-γ, IL-6, and IL-13. Pathological staining showed that the infiltration of glomerular inflammatory cells was significantly reduced and that the level of glomerular fibrosis was significantly alleviated in hUC-MSC-transplanted mice. Immunofluorescence assays showed that the deposition of IgG and IgM antibodies in the kidneys of hUC-MSC-transplanted mice was significantly lower than in the control. CONCLUSION: hUC-MSC transplantation could inhibit the proliferation and differentiation of peripheral blood B cells in the early-stage of MRL/lpr mice, thereby alleviating the plasma inflammatory environment in mice, leading to kidney injury remission. The study provides a new and feasible strategy for SLE treatment.


Subject(s)
Mesenchymal Stem Cell Transplantation , Humans , Animals , Mice , Mice, Inbred MRL lpr , Immunologic Factors , Immunoglobulin G , Kidney
4.
Front Pharmacol ; 14: 1093442, 2023.
Article in English | MEDLINE | ID: mdl-36998610

ABSTRACT

Background: This study compared the efficacy and safety of sequential immunosuppressive therapy in patients with non-end-stage IgA nephropathy (IgAN) with Lee's classification of IV ∼ V and provided evidence for the use of immunotherapy in patients with severe IgAN. Methods: We retrospectively analyzed the clinical data of patients with Lee's IV ∼ V non-end-stage IgA nephropathy. Results: 436 patients were diagnosed with IgAN, and 98 patients who met the inclusion criteria were included in this retrospective study. Of these, 17 were in the supportive care group, 20 in the P group (prednisone-only), 35 in P + CTX group (the prednisone combined with cyclophosphamide followed by mycophenolate mofetil), and 26 in the P + MMF group (prednisone combined with mycophenolate mofetil). The four groups showed differences in the segmental glomerulosclerosis score and the proportion of patients with Lee's grade IV (p < 0.05), but no differences in other indicators. Compared with the baseline values, urine protein-to-creatinine ratio (PCR) significantly decreased and serum albumin increased (p < 0.05), but there was no significant difference between the groups. The estimated Glomerular Filtration Rate (eGFR) of the P, P + MMF, and P + CTX groups were higher than that of the supportive care group at the 6th and 24th month after treatment (all p < 0.05). At the 24th month, the eGFR in the P + CTX group was higher than that in the P + MMF group (p < 0.05). The effective remission rate of the P + CTX group was higher than that of the supportive care group (p < 0.05). At 12 months, the effective remission rate of the P group was higher than that of the supportive care group (p < 0.05). At the 24th month, there was no significant difference in the effective remission rates among the three groups (P, P + MMF, and P + CTX). Nine patients with severe IgA nephropathy reached the endpoint. Conclusion: This study showed that immunosuppressive therapy insevere IgAN patient scan effectively reduce urinary protein, increase albumin, and protect renal function in the early stages of IgAN. P + CTX is the most commonly used, which has a high effective remission rate of urine protein and a low incidence of end-point events.

5.
Int J Mol Sci ; 24(3)2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36769347

ABSTRACT

Cisplatin, a widely used anticancer agent, can cause nephrotoxicity, including both acute kidney injury (AKI) and chronic kidney diseases, by accumulating in renal tubular epithelial cells (TECs). Mitochondrial pathology plays an important role in the pathogenesis of AKI. Based on the regulatory role of transcription factor EB (TFEB) in mitochondria, we investigated whether TFEB is involved in cisplatin-induced TEC damage. The results show that the expression of TFEB decreased in a concentration-dependent manner in both mouse kidney tissue and HK-2 cells when treated with cisplatin. A knockdown of TFEB aggravated cisplatin-induced renal TEC injury, which was partially reversed by TFEB overexpression in HK-2 cells. It was further observed that the TFEB knockdown also exacerbated cisplatin-induced mitochondrial damage in vitro, and included the depolarization of membrane potential, mitochondrial fragmentation and swelling, and the production of reactive oxygen species. In contrast, TFEB overexpression alleviated cisplatin-induced mitochondrial damage in TECs. These findings suggest that decreased TFEB expression may be a key mechanism of mitochondrial dysfunction in cisplatin-induced AKI, and that upregulation of TFEB has the potential to act as a therapeutic target to alleviate mitochondrial dysfunction and cisplatin-induced TEC injury. This study is important for developing therapeutic strategies to manipulate mitochondria through TFEB to delay AKI progression.


Subject(s)
Acute Kidney Injury , Cisplatin , Mice , Animals , Cisplatin/toxicity , Cisplatin/metabolism , Apoptosis , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Mitochondria/metabolism , Transcription Factors/metabolism , Mice, Inbred C57BL
6.
Front Immunol ; 13: 1007579, 2022.
Article in English | MEDLINE | ID: mdl-36341323

ABSTRACT

Animal models play an indispensable role in the study of human diseases. However, animal models of different diseases do not fully mimic the complex internal environment of humans. Immunodeficient mice are deficient in certain genes and do not express these or show reduced expression in some of their cells, facilitating the establishment of humanized mice and simulation of the human environment in vivo. Here, we summarize the developments in immunodeficient mice, from the initial nude mice lacking T lymphocytes to NOD/SCID rgnull mice lacking T, B, and NK cell populations. We describe existing humanized immune system mouse models based on immunodeficient mice in which human cells or tissues have been transplanted to establish a human immune system, including humanized-peripheral blood mononuclear cells (Hu-PBMCs), humanized hematopoietic stem cells (Hu-HSCs), and humanized bone marrow, liver, thymus (Hu-BLT) mouse models. The different methods for their development involve varying levels of complexity and humanization. Humanized mice are widely used in the study of various diseases to provide a transitional stage for clinical research. However, several challenges persist, including improving the efficiency of reconstructing the human B cell immune response, extending lifespan, improving the survival rate of mice to extend the observation period, and improving the development of standardized commercialized models and as well as their use. Overall, there are many opportunities and challenges in the development of humanized immune system mouse models which can provide novel strategies for understanding the mechanisms and treatments of human disease.


Subject(s)
Leukocytes, Mononuclear , Mice , Humans , Animals , Mice, SCID , Mice, Inbred NOD , Mice, Nude , Disease Models, Animal , Mice, Knockout
7.
Front Immunol ; 13: 986239, 2022.
Article in English | MEDLINE | ID: mdl-36189303

ABSTRACT

Systemic lupus erythematosus (SLE) is a highly heterogeneous autoimmune disease that primarily affects women. Currently, in the search for the mechanisms of SLE pathogenesis, the association of lifestyle factors such as diet, cigarette smoking, ultraviolet radiation exposure, alcohol and caffeine-rich beverage consumption with SLE susceptibility has been systematically investigated. The cellular and molecular mechanisms mediating lifestyle effects on SLE occurrence, including interactions between genetic risk loci and environment, epigenetic changes, immune dysfunction, hyper-inflammatory response, and cytotoxicity, have been proposed. In the present review of the reports published in reputable peer-reviewed journals and government websites, we consider the current knowledge about the relationships between lifestyle factors and SLE incidence and outline directions of future research in this area. Formulation of practical measures with regard to the lifestyle in the future will benefit SLE patients and may provide potential therapy strategies.


Subject(s)
Autoimmune Diseases , Cigarette Smoking , Lupus Erythematosus, Systemic , Autoimmune Diseases/complications , Caffeine , Cigarette Smoking/adverse effects , Female , Humans , Lupus Erythematosus, Systemic/genetics , Ultraviolet Rays
8.
Eur J Med Res ; 27(1): 176, 2022 Sep 10.
Article in English | MEDLINE | ID: mdl-36088368

ABSTRACT

Hyperuricemia can induce acute and chronic kidney damage, but the pathological mechanism remains unclear. The potential role of AMP-activated protein kinase (AMPK) α2 in hyperuricemia-induced renal injury was investigated in this study. Acute and chronic hyperuricemic nephropathy was induced by administering intraperitoneal injections of uric acid and oxonic acid to AMPK α2 knockout and wild-type mice. Changes in renal function, histopathology, inflammatory cell infiltration, renal interstitial fibrosis, and urate deposition were analyzed. In both acute and chronic hyperuricemic nephropathy mouse models, knockout of AMPK α2 significantly reduced serum creatinine levels and renal pathological changes. The tubular expression of kidney injury molecule-1 was also reduced in hyperuricemic nephropathy mice deficient in AMPK α2. In addition, knockout of AMPK α2 significantly suppressed the infiltration of renal macrophages and progression of renal interstitial fibrosis in mice with chronic hyperuricemic nephropathy. Knockout of AMPK α2 reduced renal urate crystal deposition, probably through increasing the expression of the uric acid transporter, multidrug resistance protein 4. In summary, AMPK α2 is involved in acute and chronic hyperuricemia-induced kidney injury and may be associated with increased urate crystal deposition in the kidney.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Hyperuricemia , Kidney Diseases , Kidney Failure, Chronic , AMP-Activated Protein Kinases/genetics , Animals , Disease Models, Animal , Fibrosis , Hyperuricemia/chemically induced , Hyperuricemia/genetics , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/metabolism , Mice , Mice, Knockout , Uric Acid/adverse effects , Uric Acid/metabolism
10.
Lupus Sci Med ; 9(1)2022 04.
Article in English | MEDLINE | ID: mdl-35414608

ABSTRACT

OBJECTIVE: Lupus nephritis (LN) is a major complication and cause of death among patients with SLE. This research used in vivo and in vitro experiments to explore the therapeutic potential of metformin in kidney injury from LN-induced inflammation. METHODS: In vivo study, 8-week-old MRL/MpJ-Faslpr/J (MRL/lpr) mice were randomly divided into two groups (n=12 each): daily administration of 0.3 mg/mL metformin in drinking water and control (water only). Body weight and urinary samples were measured biweekly. Mice were sacrificed after 8-week treatment to harvest serum, lymph nodes, spleen and kidneys. In vitro study, human kidney-2 (HK-2) cells were pretreated with 1 mM metformin for 1 hour and then stimulated with 20 µg/mL lipopolysaccharides (LPS) or 10 ng/mL tumour necrosis factor-α (TNF-α) for another 48 hours. Protein was collected for subsequent analysis. RESULTS: We found that metformin administration improved renal function in MRL/lpr lupus-prone mice, measured by decreased urea nitrogen and urinary proteins. Metformin reduced immunoglobulin G and complement C3 deposition in glomeruli. The treatment also downregulated systemic and renal inflammation, as seen in decreased renal infiltration of F4/80-positive macrophages and reduced splenic and renal MCP-1 (monocyte chemoattractant protein-1) and TNF-α, and renal IL-1ß (interleukin 1ß) expression. Metformin administration decreased renal expression of necroptosis markers p-RIPK1 (phosphorylated receptor-interacting protein kinase 1) and p-MLKL, along with tubular injury marker KIM-1 (kidney injury molecule-1) in lupus mice. In addition, metformin alleviated the necroptosis of HK-2 cells stimulated by LPS and TNF-α, evidencing by a decrease in the expression of necroptosis markers p-RIPK1, p-RIPK3 and p-MLKL, and the inflammasome-related markers NLRP3 (NLR family pyrin domain containing 3), ASC (apoptosis-associated speck-like protein containing a CARD), caspase-1. Mechanistically, metformin treatment upregulated p-AMPK (phosphorylated AMP-activated protein kinase) and downregulated p-STAT3 (phosphorylated signal transducer and activator of transcription 3) expression in the kidneys. Moreover, AMPKα2 knockdown abolished the protective effects of metformin in vitro. CONCLUSIONS: Metformin alleviated kidney injury in LN though suppressing renal necroptosis and inflammation via the AMPK/STAT3 pathway.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Metformin , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/pharmacology , Animals , Humans , Inflammation , Kidney/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/therapeutic use , Lupus Erythematosus, Systemic/complications , Lupus Erythematosus, Systemic/drug therapy , Lupus Nephritis/complications , Lupus Nephritis/drug therapy , Metformin/pharmacology , Metformin/therapeutic use , Mice , Mice, Inbred MRL lpr , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology , STAT3 Transcription Factor/therapeutic use , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/therapeutic use
11.
Arthritis Res Ther ; 24(1): 6, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34980245

ABSTRACT

BACKGROUND: Hydroxychloroquine (HCQ) has been recommended as a basic treatment for lupus nephritis (LN) during this decade based on its ability to improve LN-related renal immune-mediated inflammatory lesions. As a classical lysosomal inhibitor, HCQ may inhibit lysosomal degradation and disrupt protective autophagy in proximal tubular epithelial cells (PTECs). Therefore, the final renal effects of HCQ on LN need to be clarified. METHOD: HCQ was administered on spontaneous female MRL/lpr LN mice with severe proteinuria daily for 4 weeks. Moreover, the MRL/lpr mice with proteinuric LN were subjected to cisplatin-induced or unilateral ischemia/reperfusion (I/R)-induced acute kidney injury (AKI) after 2 weeks of HCQ preadministration. RESULTS: As expected, HCQ treatment increased the survival ratio and downregulated the levels of serum creatinine in the mice with LN, ameliorated renal lesions, and inhibited renal interstitial inflammation. Unexpectedly, HCQ preadministration significantly increased susceptibility to and delayed the recovery of AKI complicated by LN, as demonstrated by an increase in PTEC apoptosis and expression of the tubular injury marker KIM-1 as well as the retardation of PTEC replenishment. HCQ preadministration suppressed the proliferation of PTECs by arresting cells in G1/S phase and upregulated the expression of cell cycle inhibitors. Furthermore, HCQ preadministration disrupted the PTEC autophagy-lysosomal pathway and accelerated PTEC senescence. CONCLUSION: HCQ treatment may increase susceptibility and delay the recovery of AKI complicated by LN despite its ability to improve LN-related renal immune-mediated inflammatory lesions. The probable mechanism involves accelerated apoptosis and inhibited proliferation of PTECs via autophagy-lysosomal pathway disruption and senescence promotion.


Subject(s)
Acute Kidney Injury , Lupus Nephritis , Acute Kidney Injury/chemically induced , Animals , Female , Hydroxychloroquine/pharmacology , Kidney/pathology , Mice , Mice, Inbred MRL lpr
12.
Front Physiol ; 12: 786599, 2021.
Article in English | MEDLINE | ID: mdl-34950058

ABSTRACT

Nowadays, with the improvements in living standards and changes in living habits, high-fat diet (HFD) has become much more common in the populations worldwide. Recent studies have shown that HFD could induce lipid accumulation, and structural and functional abnormalities, accompanied by the release of large amounts of pro-inflammatory cytokines, in proximal tubular epithelial cells (PTECs). These findings indicate that, as an emerging risk factor, PTEC injury-induced by HFD may be closely related to inflammation; however, the potential mechanisms underlying this phenomenon is still not well-known, but may involve the several inflammatory pathways, including oxidative stress-related signaling pathways, mitochondrial dysfunction, the myeloid differentiation factor 2/Toll like receptor 4 (MD2/TLR4) signaling pathway, the ERK1/2-kidney injury molecule 1 (KIM-1)-related pathway, and nuclear factor-κB (NF-κB) activation, etc., and the detailed molecular mechanisms underlying these pathways still need further investigated in the future. Based on lipid abnormalities-induced inflammation is closely related to the development and progression of chronic kidney disease (CKD), to summarize the potential mechanisms underlying HFD-induced renal proximal tubular inflammatory injury, may provide novel approaches for CKD treatment.

13.
Front Immunol ; 12: 799788, 2021.
Article in English | MEDLINE | ID: mdl-34925385

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that was traditionally thought to be closely related to genetic and environmental risk factors. Although treatment options for SLE with hormones, immunosuppressants, and biologic drugs are now available, the rates of clinical response and functional remission of these drugs are still not satisfactory. Currently, emerging evidence suggests that gut microbiota dysbiosis may play crucial roles in the occurrence and development of SLE, and manipulation of targeting the gut microbiota holds great promises for the successful treatment of SLE. The possible mechanisms of gut microbiota dysbiosis in SLE have not yet been well identified to date, although they may include molecular mimicry, impaired intestinal barrier function and leaky gut, bacterial biofilms, intestinal specific pathogen infection, gender bias, intestinal epithelial cells autophagy, and extracellular vesicles and microRNAs. Potential therapies for modulating gut microbiota in SLE include oral antibiotic therapy, fecal microbiota transplantation, glucocorticoid therapy, regulation of intestinal epithelial cells autophagy, extracellular vesicle-derived miRNA therapy, mesenchymal stem cell therapy, and vaccination. This review summarizes novel insights into the mechanisms of microbiota dysbiosis in SLE and promising therapeutic strategies, which may help improve our understanding of the pathogenesis of SLE and provide novel therapies for SLE.


Subject(s)
Dysbiosis/complications , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Lupus Erythematosus, Systemic/microbiology , Animals , Humans , Lupus Erythematosus, Systemic/immunology
14.
Front Immunol ; 12: 728190, 2021.
Article in English | MEDLINE | ID: mdl-34659214

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic autoimmune disease. Although previous studies have demonstrated that SLE is related to the imbalance of cells in the immune system, including B cells, T cells, and dendritic cells, etc., the mechanisms underlying SLE pathogenesis remain unclear. Therefore, effective and low side-effect therapies for SLE are lacking. Recently, mesenchymal stem cell (MSC) therapy for autoimmune diseases, particularly SLE, has gained increasing attention. This therapy can improve the signs and symptoms of refractory SLE by promoting the proliferation of Th2 and Treg cells and inhibiting the activity of Th1, Th17, and B cells, etc. However, MSC therapy is also reported ineffective in some patients with SLE, which may be related to MSC- or patient-derived factors. Therefore, the therapeutic effects of MSCs should be further confirmed. This review summarizes the status of MSC therapy in refractory SLE treatment and potential reasons for the ineffectiveness of MSC therapy from three perspectives. We propose various MSC modification methods that may be beneficial in enhancing the immunosuppression of MSCs in SLE. However, their safety and protective effects in patients with SLE still need to be confirmed by further experimental and clinical evidence.


Subject(s)
Lupus Erythematosus, Systemic/surgery , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Adaptive Immunity , Animals , Cellular Microenvironment , Humans , Immune Tolerance , Immunity, Innate , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Mesenchymal Stem Cell Transplantation/adverse effects , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Phenotype , Treatment Outcome
15.
J Cell Mol Med ; 25(12): 5729-5743, 2021 06.
Article in English | MEDLINE | ID: mdl-33949118

ABSTRACT

Cyclosporine A (CsA) is an immunosuppressor widely used for the prevention of acute rejection during solid organ transplantation. However, severe nephrotoxicity has substantially limited its long-term usage. Recently, an impaired autophagy pathway was suggested to be involved in the pathogenesis of chronic CsA nephrotoxicity. However, the underlying mechanisms of CsA-induced autophagy blockade in tubular cells remain unclear. In the present study, we observed that CsA suppressed the activation and expression of transcription factor EB (TFEB) by increasing the activation of mTOR, in turn promoting lysosomal dysfunction and autophagy flux blockade in tubular epithelial cells (TECs) in vivo and in vitro. Restoration of TFEB activation by Torin1-mediated mTOR inhibition significantly improved lysosomal function and rescued autophagy pathway activity, suppressing TEC injury. In summary, targeting TFEB-mediated autophagy flux represents a potential therapeutic strategy for CsA-induced nephrotoxicity.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cyclosporine/toxicity , Epithelial Cells/pathology , Kidney Tubules/pathology , Lysosomes/pathology , TOR Serine-Threonine Kinases/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Immunosuppressive Agents/toxicity , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Male , Mice , Mice, Inbred BALB C , TOR Serine-Threonine Kinases/genetics
17.
J Cell Mol Med ; 25(5): 2703-2713, 2021 03.
Article in English | MEDLINE | ID: mdl-33605079

ABSTRACT

Acute kidney injury (AKI) is the main obstacle that limits the use of cisplatin in cancer treatment. Proton pump inhibitors (PPIs), the most commonly used class of medications for gastrointestinal complications in cancer patients, have been reported to cause adverse renal events. However, the effect of PPIs on cisplatin-induced AKI remains unclear. Herein, the effect and mechanism of lansoprazole (LPZ), one of the most frequently prescribed PPIs, on cisplatin-induced AKI were investigated in vivo and in vitro. C57BL/6 mice received a single intraperitoneal (i.p.) injection of cisplatin (18 mg/kg) to induce AKI, and LPZ (12.5 or 25 mg/kg) was administered 2 hours prior to cisplatin administration and then once daily for another 2 days via i.p. injection. The results showed that LPZ significantly aggravated the tubular damage and further increased the elevated levels of serum creatinine and blood urea nitrogen induced by cisplatin. However, LPZ did not enhance cisplatin-induced tubular apoptosis, as evidenced by a lack of significant change in mRNA and protein expression of Bax/Bcl-2 ratio and TUNEL staining. Notably, LPZ increased the number of necrotic renal tubular cells compared to that by cisplatin treatment alone, which was further confirmed by the elevated necroptosis-associated protein expression of RIPK1, p-RIPK3 and p-MLKL. Furthermore, LPZ deteriorated cisplatin-induced inflammation, as revealed by the increased mRNA expression of pro-inflammatory factors including, NLRP3, IL-1ß, TNF-α and caspase 1, as well as neutrophil infiltration. Consistently, in in vitro study, LPZ increased HK-2 cell death and enhanced inflammation, compared with cisplatin treatment alone. Collectively, our results demonstrate that LPZ aggravates cisplatin-induced AKI, and necroptosis may be involved in the exacerbation of kidney damage.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Kidney Tubular Necrosis, Acute/etiology , Kidney Tubular Necrosis, Acute/metabolism , Lansoprazole/adverse effects , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Apoptosis/drug effects , Disease Models, Animal , Drug Synergism , Kidney Tubular Necrosis, Acute/pathology , Mice
18.
Front Immunol ; 12: 792884, 2021.
Article in English | MEDLINE | ID: mdl-35003113

ABSTRACT

Systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) are two common multisystem autoimmune diseases that share, among others, many clinical manifestations and serological features. The role of long non-coding RNAs (lncRNAs) has been of particular interest in the pathogenesis of autoimmune diseases. Here, we aimed to summarize the roles of lncRNAs as emerging novel biomarkers and therapeutic targets in SLE and RA. We conducted a narrative review summarizing original articles on lncRNAs associated with SLE and RA, published until November 1, 2021. Based on the studies on lncRNA expression profiles in samples (including PBMCs, serum, and exosomes), it was noted that most of the current research is focused on investigating the regulatory mechanisms of these lncRNAs in SLE and/or RA. Several lncRNAs have been hypothesized to play key roles in these diseases. In SLE, lncRNAs such as GAS5, NEAT1, TUG1, linc0949, and linc0597 are dysregulated and may serve as emerging novel biomarkers and therapeutic targets. In RA, many validated lncRNAs, such as HOTAIR, GAS5, and HIX003209, have been identified as promising novel biomarkers for both diagnosis and treatment. The shared lncRNAs, for example, GAS5, may participate in SLE pathogenesis through the mitogen-activated protein kinase pathway and trigger the AMP-activated protein kinase pathway in RA. Here, we summarize the data on key lncRNAs that may drive the pathogenesis of SLE and RA and could potentially serve as emerging novel biomarkers and therapeutic targets in the coming future.


Subject(s)
Arthritis, Rheumatoid/genetics , Lupus Erythematosus, Systemic/genetics , RNA, Long Noncoding/genetics , Animals , Biomarkers , Humans , Molecular Targeted Therapy , Signal Transduction , Transcriptome
19.
Front Immunol ; 12: 816956, 2021.
Article in English | MEDLINE | ID: mdl-35116040

ABSTRACT

Animal models have played a crucial role in the understanding of the mechanisms and treatments of human diseases; however, owing to the large differences in genetic background and disease-specific characteristics, animal models cannot fully simulate the occurrence and progression of human diseases. Recently, humanized immune system mice, based on immunodeficient mice, have been developed that allow for the partial reconstruction of the human immune system and mimic the human in vivo microenvironment. Systemic lupus erythematosus (SLE) is a complex disease characterized by the loss of tolerance to autoantigens, overproduction of autoantibodies, and inflammation in multiple organ systems. The detailed immunological events that trigger the onset of clinical manifestations in patients with SLE are still not well known. Two methods have been adopted for the development of humanized SLE mice. They include transferring peripheral blood mononuclear cells from patients with SLE to immunodeficient mice or transferring human hematopoietic stem cells to immunodeficient mice followed by intraperitoneal injection with pristane to induce lupus. However, there are still several challenges to be overcome, such as how to improve the efficiency of reconstruction of the human B cell immune response, how to extend the lifespan and improve the survival rate of mice to extend the observation period, and how to improve the development of standardized commercialized models and use them. In summary, there are opportunities and challenges for the development of humanized mouse models of SLE, which will provide novel strategies for understanding the mechanisms and treatments of SLE.


Subject(s)
Disease Models, Animal , Lupus Erythematosus, Systemic/etiology , Lupus Erythematosus, Systemic/metabolism , Mice, Transgenic , Animals , Autoantibodies/immunology , Autoantigens/immunology , Autoimmune Diseases , Autoimmunity , Biomarkers , Disease Management , Disease Susceptibility/immunology , Humans , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/therapy , Mice
20.
Discov Med ; 30(159): 19-25, 2020.
Article in English | MEDLINE | ID: mdl-33357359

ABSTRACT

BACKGROUND: Crescent formation is a serious pathological change in the IgA nephropathy (IgAN) which is believed to be primarily mediated by a mixture of parietal epithelial cells, macrophages, and myofibroblasts. It was recommended that IgAN patients with rapid renal hypofunction with a crescent body >50% should begin treatment with corticosteroids combined with cyclophosphamide. However, for patients with partial crescent formation, whether immunosuppressive therapy is necessary is a contested topic. MATERIALS AND METHODS: Data from IgAN patients with partial crescent formation who underwent repeat renal biopsy were retrospectively analyzed. RESULTS: From the first to the second renal biopsy, the mean hemoglobin level and albumin level increased significantly (P < 0.05), and uric acid and triglyceride levels decreased significantly (P < 0.05). Also, the 24-hour urinary protein excretion decreased significantly (P < 0.001), but no differences in blood pressure, creatinine level, or estimated glomerular filtration rate. For pathological indices, there were no differences in fluorescence intensity of IgA or C3 deposition (P > 0.05), but the mesangial cell proliferation decreased significantly (P < 0.05), and the proportions of global glomerulosclerosis and tubulointerstitial fibrosis increased significantly (P < 0.05, respectively). In addition, a decreased tendency in the proportion of crescent formation was observed in the second renal biopsy. CONCLUSIONS: Immunosuppressive therapy for IgAN patients with partial crescent formation can reduce proteinuria, stabilize renal function, improve anemia, and mitigate acute kidney injury in the short term.


Subject(s)
Acute Kidney Injury/drug therapy , Glomerulonephritis, IGA/drug therapy , Immunosuppressive Agents/administration & dosage , Kidney/pathology , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Adult , Biopsy , Cyclophosphamide/administration & dosage , Disease Progression , Drug Therapy, Combination , Female , Follow-Up Studies , Glomerular Filtration Rate , Glomerulonephritis, IGA/complications , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/pathology , Glucocorticoids/administration & dosage , Humans , Kidney/drug effects , Kidney/immunology , Male , Middle Aged , Retrospective Studies , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...