Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 205
Filter
1.
Oncol Rep ; 52(2)2024 Aug.
Article in English | MEDLINE | ID: mdl-38940330

ABSTRACT

Ferroptosis, a regulated form of cell death, is intricately linked to iron­dependent lipid peroxidation. Recent evidence strongly supports the induction of ferroptosis as a promising strategy for treating cancers resistant to conventional therapies. A key player in ferroptosis regulation is ferroptosis suppressor protein 1 (FSP1), which promotes cancer cell resistance by promoting the production of the antioxidant form of coenzyme Q10. Of note, FSP1 confers resistance to ferroptosis independently of the glutathione (GSH) and glutathione peroxidase­4 pathway. Therefore, targeting FSP1 to weaken its inhibition of ferroptosis may be a viable strategy for treating refractory cancer. This review aims to clarify the molecular mechanisms underlying ferroptosis, the specific pathway by which FSP1 suppresses ferroptosis and the effect of FSP1 inhibitors on cancer cells.


Subject(s)
Ferroptosis , Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Ferroptosis/drug effects , S100 Calcium-Binding Protein A4/metabolism , S100 Calcium-Binding Protein A4/antagonists & inhibitors , Ubiquinone/analogs & derivatives , Ubiquinone/therapeutic use , Ubiquinone/pharmacology , Lipid Peroxidation/drug effects , Drug Resistance, Neoplasm/drug effects , Animals , Glutathione/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Molecular Targeted Therapy/methods
2.
Int J Biol Sci ; 20(7): 2622-2639, 2024.
Article in English | MEDLINE | ID: mdl-38725840

ABSTRACT

Sorafenib is a standard first-line drug for advanced hepatocellular carcinoma, but the serious cardiotoxic effects restrict its therapeutic applicability. Here, we show that iron-dependent ferroptosis plays a vital role in sorafenib-induced cardiotoxicity. Remarkably, our in vivo and in vitro experiments demonstrated that ferroptosis inhibitor application neutralized sorafenib-induced heart injury. By analyzing transcriptome profiles of adult human sorafenib-treated cardiomyocytes, we found that Krüppel-like transcription factor 11 (KLF11) expression significantly increased after sorafenib stimulation. Mechanistically, KLF11 promoted ferroptosis by suppressing transcription of ferroptosis suppressor protein 1 (FSP1), a seminal breakthrough due to its ferroptosis-repressing properties. Moreover, FSP1 knockdown showed equivalent results to glutathione peroxidase 4 (GPX4) knockdown, and FSP1 overexpression counteracted GPX4 inhibition-induced ferroptosis to a substantial extent. Cardiac-specific overexpression of FSP1 and silencing KLF11 by an adeno-associated virus serotype 9 markedly improved cardiac dysfunction in sorafenib-treated mice. In summary, FSP1-mediated ferroptosis is a crucial mechanism for sorafenib-provoked cardiotoxicity, and targeting ferroptosis may be a promising therapeutic strategy for alleviating sorafenib-induced cardiac damage.


Subject(s)
Cardiotoxicity , Ferroptosis , Repressor Proteins , S100 Calcium-Binding Protein A4 , Sorafenib , Animals , Humans , Male , Mice , Cardiotoxicity/metabolism , Cardiotoxicity/etiology , Ferroptosis/drug effects , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Repressor Proteins/metabolism , Repressor Proteins/genetics , S100 Calcium-Binding Protein A4/metabolism , S100 Calcium-Binding Protein A4/genetics , Sorafenib/adverse effects
4.
Adv Healthc Mater ; 13(16): e2304436, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38335308

ABSTRACT

An imbalance in reactive oxygen species (ROS) levels in tumor cells can result in the accumulation of lipid peroxide (LPO) which can induce ferroptosis. Moreover, elevated ROS levels in tumors present a chance to develop ROS-based cancer therapeutics including photodynamic therapy (PDT) and ferroptosis. However, their anticancer efficacies are compromised by insufficient oxygen levels and inherent cellular ROS regulatory mechanism. Herein, a cell membrane-targeting photosensitizer, TBzT-CNQi, which can generate 1O2, •OH, and O2 •- via type I/II process to induce a high level of LPO for potent ferroptosis and photodynamic therapy is developed. The FSP1 inhibitor (iFSP1) is incorporated with TBzT-CNQi to downregulate FSP1 expression, lower the intracellular CoQ10 content, induce a high level of LPO, and activate initial tumor immunogenic ferroptosis. In vitro and in vivo experiments demonstrate that the cell membrane-targeting type I/II PDT combination with FSP1 inhibition can evoke strong ICD and activate the immune response, which subsequently promotes the invasion of CD8+ T cells infiltration, facilitates the dendritic cell maturation, and decreases the tumor infiltration of tumor-associated macrophages. The study indicates that the combination of cell membrane-targeting type I/II PDT and FSP1 inhibition holds promise as a potential strategy for ferroptosis-enhanced photodynamic immunotherapy of hypoxia tumors.


Subject(s)
Ferroptosis , Photochemotherapy , Photosensitizing Agents , S100 Calcium-Binding Protein A4 , Ferroptosis/drug effects , Photochemotherapy/methods , Animals , Mice , Humans , S100 Calcium-Binding Protein A4/metabolism , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Cell Line, Tumor , Cell Membrane/metabolism , Immunotherapy/methods , Reactive Oxygen Species/metabolism , Female
5.
Cell Death Differ ; 31(4): 497-510, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38374229

ABSTRACT

Poly ADP-ribose polymerase inhibitors (PARPis) exhibit promising efficacy in patients with BRCA mutations or homologous repair deficiency (HRD) in ovarian cancer (OC). However, less than 40% of patients have HRD, it is vital to expand the indications for PARPis in BRCA-proficient patients. Ferroptosis suppressor protein 1 (FSP1) is a key protein in a newly identified ferroptosis-protective mechanism that occurs in parallel with the GPX4-mediated pathway and is associated with chemoresistance in several cancers. Herein, FSP1 is reported to be negatively correlated with the prognosis in OC patients. Combination therapy comprising olaparib and iFSP1 (a FSP1 inhibitor) strongly inhibited tumour proliferation in BRCA-proficient OC cell lines, patient-derived organoids (PDOs) and xenograft mouse models. Surprisingly, the synergistic killing effect could not be reversed by ferroptosis inhibitors, indicating that mechanisms other than ferroptosis were responsible for the synergistic lethality. In addition, cotreatment was shown to induce increased γH2A.X foci and to impair nonhomologous end joining (NHEJ) activity to a greater extent than did any single drug. Mass spectrometry and immunoprecipitation analyses revealed that FSP1 interacted with Ku70, a classical component recruited to and occupying the end of double-strand breaks (DSBs) in the NHEJ process. FSP1 inhibition decreased Ku70 PARylation, impaired subsequent DNA-PKcs recruitment to the Ku complex at DSB sites and was rescued by restoring PARylation. These findings unprecedentedly reveal a novel role of FSP1 in DNA damage repair and provide new insights into how to sensitize OC patients to PARPi treatment.


Subject(s)
Ferroptosis , Ovarian Neoplasms , Phthalazines , Piperazines , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Female , Phthalazines/pharmacology , Phthalazines/therapeutic use , Piperazines/pharmacology , Piperazines/therapeutic use , Animals , Mice , Ferroptosis/drug effects , Cell Line, Tumor , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Cell Proliferation/drug effects , S100 Calcium-Binding Protein A4/metabolism , S100 Calcium-Binding Protein A4/genetics
6.
Int J Biochem Cell Biol ; 169: 106551, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38360265

ABSTRACT

Benign prostatic hyperplasia (BPH) is one of the most common diseases in elderly men worldwide that may result in lower urinary tract symptoms (LUTS). At present, the specific pathophysiological mechanism for BPH/LUTS LUTS remains unclear. S100 calcium binding protein A4 (S100A4), a member of the calcium binding protein family, regulates a variety of biological processes including cell proliferation, apoptosis and fibrosis. The aim of the current study was to explore and clarify the possible role of S100A4 in BPH/LUTS. The human prostate stromal cell line (WPMY-1), rat prostate epithelial cells, human prostate tissues and two BPH rat models were employed in this study. The expression and localization of S100A4 were detected by quantitative real time PCR (qRT-PCR), immunofluorescence microscopy, Western blotting and immunohistochemistry analysis. Also, S100A4 knockdown or overexpression cell models were constructed and a BPH rat model was induced with testosterone propionate (T) or phenylephrine (PE). The BPH animals were treated with Niclosamide, a S100A4 transcription inhibitor. Results demonstrated that S100A4 was mainly localized in human prostatic stroma and rat prostatic epithelium, and showed a higher expression in BPH. Knockdown of S100A4 induced cell apoptosis, cell proliferation arrest and a reduction of tissue fibrosis markers. Overexpression of S100A4 reversed the aforementioned changes. We also demonstrated that S100A4 regulated proliferation and apoptosis mainly through the ERK pathway and modulated fibrosis via Wnt/ß-catenin signaling. In conclusion, our novel data demonstrate that S100A4 could play a crucial role in BPH development and may be explored as a new therapeutic target of BPH.


Subject(s)
Prostate , Prostatic Hyperplasia , S100 Calcium-Binding Protein A4 , Aged , Animals , Humans , Male , Rats , Apoptosis , Cell Proliferation , Fibrosis , Prostate/metabolism , Prostatic Hyperplasia/metabolism , S100 Calcium-Binding Protein A4/genetics , S100 Calcium-Binding Protein A4/metabolism
7.
Int Immunopharmacol ; 128: 111555, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38280333

ABSTRACT

S100A4 is implicated in metabolic reprogramming across various cell types and is known to propel the progression of numerous diseases including allergies. Nonetheless, the influence of S100A4 on mast cell metabolic reprogramming during allergic disorders remains unexplored. Utilizing a mast cell line (C57), cells were treated with recombinant mouse S100A4 protein, with or without a PPAR-γ agonist (ROSI) or a RAGE inhibitor (FPS-ZM1). Subsequent assessments were conducted for mast cell activation and lipid metabolism. S100A4 induced mast cell activation and the release of inflammatory mediators, concurrently altering molecules involved in lipid metabolism and glycolysis over time. Furthermore, S100A4 stimulation resulted in cellular oxidative stress and mitochondrial dysfunction. Alterations in the levels of pivotal molecules within the RAGE/Src/JAK2/STAT3/PPAR-γ and NF-κB signaling pathways were noted during this stimulation, which were partially counteracted by ROSI or FPS-ZMI. Additionally, a trend of metabolic alterations was identified in patients with allergic asthma who exhibited elevated serum S100A4 levels. Correlation analysis unveiled a positive association between serum S100A4 and serum IgE, implying an indirect association with asthma. Collectively, our findings suggest that S100A4 regulates the lipid-metabolic reprogramming of mast cells, potentially via the RAGE and PPAR-γ-involved signaling pathway, offering a novel perspective in the disease management in patients with allergic disorders.


Subject(s)
Asthma , Mast Cells , Animals , Mice , Humans , S100 Calcium-Binding Protein A4/metabolism , Mast Cells/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Lipid Metabolism , Signal Transduction , Asthma/metabolism
8.
J Biol Chem ; 300(1): 105530, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38072048

ABSTRACT

Fibroblast to myofibroblast transdifferentiation mediates numerous fibrotic disorders, such as idiopathic pulmonary fibrosis (IPF). We have previously demonstrated that non-muscle myosin II (NMII) is activated in response to fibrotic lung extracellular matrix, thereby mediating myofibroblast transdifferentiation. NMII-A is known to interact with the calcium-binding protein S100A4, but the mechanism by which S100A4 regulates fibrotic disorders is unclear. In this study, we show that fibroblast S100A4 is a calcium-dependent, mechanoeffector protein that is uniquely sensitive to pathophysiologic-range lung stiffness (8-25 kPa) and thereby mediates myofibroblast transdifferentiation. Re-expression of endogenous fibroblast S100A4 rescues the myofibroblastic phenotype in S100A4 KO fibroblasts. Analysis of NMII-A/actin dynamics reveals that S100A4 mediates the unraveling and redistribution of peripheral actomyosin to a central location, resulting in a contractile myofibroblast. Furthermore, S100A4 loss protects against murine in vivo pulmonary fibrosis, and S100A4 expression is dysregulated in IPF. Our data reveal a novel mechanosensor/effector role for endogenous fibroblast S100A4 in inducing cytoskeletal redistribution in fibrotic disorders such as IPF.


Subject(s)
Idiopathic Pulmonary Fibrosis , Mechanotransduction, Cellular , Myofibroblasts , S100 Calcium-Binding Protein A4 , Animals , Mice , Cell Transdifferentiation , Fibrosis , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , S100 Calcium-Binding Protein A4/genetics , S100 Calcium-Binding Protein A4/metabolism
9.
Arthritis Rheumatol ; 76(5): 783-795, 2024 May.
Article in English | MEDLINE | ID: mdl-38108109

ABSTRACT

OBJECTIVE: S100A4 is a DAMP protein. S100A4 is overexpressed in patients with systemic sclerosis (SSc), and levels correlate with organ involvement and disease activity. S100A4-/- mice are protected from fibrosis. The aim of this study was to assess the antifibrotic effects of anti-S100A4 monoclonal antibody (mAb) in murine models of SSc and in precision cut skin slices of patients with SSc. METHODS: The effects of anti-S100A4 mAbs were evaluated in a bleomycin-induced skin fibrosis model and in Tsk-1 mice with a therapeutic dosing regimen. In addition, the effects of anti-S100A4 mAbs on precision cut SSc skin slices were analyzed by RNA sequencing. RESULTS: Inhibition of S100A4 was effective in the treatment of pre-established bleomycin-induced skin fibrosis and in regression of pre-established fibrosis with reduced dermal thickening, myofibroblast counts, and collagen accumulation. Transcriptional profiling demonstrated targeting of multiple profibrotic and proinflammatory processes relevant to the pathogenesis of SSc on targeted S100A4 inhibition in a bleomycin-induced skin fibrosis model. Moreover, targeted S100A4 inhibition also modulated inflammation- and fibrosis-relevant gene sets in precision cut SSc skin slices in an ex vivo trial approach. Selected downstream targets of S100A4, such as AMP-activated protein kinase, calsequestrin-1, and phosphorylated STAT3, were validated on the protein level, and STAT3 inhibition was shown to prevent the profibrotic effects of S100A4 on fibroblasts in human skin. CONCLUSION: Inhibition of S100A4 confers dual targeting of inflammatory and fibrotic pathways in complementary mouse models of fibrosis and in SSc skin. These effects support the further development of anti-S100A4 mAbs as disease-modifying targeted therapies for SSc.


Subject(s)
Antibodies, Monoclonal , Bleomycin , Disease Models, Animal , Fibrosis , S100 Calcium-Binding Protein A4 , Scleroderma, Systemic , Skin , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/genetics , Animals , S100 Calcium-Binding Protein A4/genetics , S100 Calcium-Binding Protein A4/metabolism , Humans , Mice , Skin/pathology , Skin/drug effects , Skin/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , STAT3 Transcription Factor/metabolism , Female
10.
Sci Rep ; 13(1): 22540, 2023 12 18.
Article in English | MEDLINE | ID: mdl-38110482

ABSTRACT

Podocyte expression of fibroblast specific protein 1 (FSP1) is observed in various types of human glomerulonephritis. Considering that FSP1 is secreted extracellularly and has been shown to have multiple biological effects on distant cells, we postulated that secreted FSP1 from podocytes might impact renal tubules. Our RNA microarray analysis in a tubular epithelial cell line (mProx) revealed that FSP1 induced the expression of heme oxygenase 1, sequestosome 1, solute carrier family 7, member 11, and cystathionine gamma-lyase, all of which are associated with nuclear factor erythroid 2-related factor (Nrf2) activation. Therefore, FSP1 is likely to exert cytoprotective effects through Nrf2-induced antioxidant activity. Moreover, in mProx, FSP1 facilitated Nrf2 translocation to the nucleus, increased levels of reduced glutathione, inhibited the production of reactive oxygen species (ROS), and reduced cisplatin-induced cell death. FSP1 also ameliorated acute tubular injury in mice with cisplatin nephrotoxicity, which is a representative model of ROS-mediated tissue injury. Similarly, in transgenic mice that express FSP1 specifically in podocytes, tubular injury associated with cisplatin nephrotoxicity was also mitigated. Extracellular FSP1 secreted from podocytes acts on downstream tubular cells, exerting renoprotective effects through Nrf2-mediated antioxidant activity. Consequently, podocytes and tubular epithelial cells have a remote communication network to limit injury.


Subject(s)
Antioxidants , NF-E2-Related Factor 2 , Humans , Mice , Animals , Antioxidants/pharmacology , Antioxidants/metabolism , Reactive Oxygen Species/metabolism , S100 Calcium-Binding Protein A4/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Cisplatin/pharmacology , Cisplatin/metabolism , Oxidative Stress , Heme Oxygenase-1/metabolism
11.
Discov Med ; 35(176): 251-263, 2023 06.
Article in English | MEDLINE | ID: mdl-37272092

ABSTRACT

BACKGROUND: Cancer stem cells (CSCs) are characterized by an ability for unlimited proliferation and efficiency of self-renewal. The targeting of lung CSCs (LCSCs)-related signaling pathways represent a promising therapeutic strategy for treatment of lung cancer. Ferroptosis a potential strategy for LCSCs treatment, and curcumin cloud induce ferroptosis. In this study, we aimed to observe the effects of curcumin on LCSCs via ferroptosis-related pathways. METHODS: In this study, A549 cluster of differentiation (CD)133+ and A549 CD133- cells were isolated using magnetic bead-based separation. Colony formation and sphere formation assays, as well as cells injection in non-obese diabetes/severe combined immune deficiency (NOD/SCID) mice, were used to analyze the tumorigenic ability of cells differentially expressing CD133. A549 CD133+ cells were treated with different doses of curcumin (0, 10, 20, 40, 80 µM). Cell viability, glutathione peroxidase 4 (GPX4) and ferroptosis suppressor protein 1 (FSP1) expressions were measured. The 50% inhibitory concentration (IC50) of curcumin, two ferroptosis inducers, inhibitor of GPX4 (RSL3) and inhibitor of FSP1 (iFSP1), and a ferroptosis inhibitor, ferrostatin-1 (Fer-1), were used to investigate the mechanism underlying the effect of curcumin on ferroptosis in A549 CD133+ cells. RESULTS: A549 CD133+ cells had greater tumorigenic ability than A549 cells. Curcumin treatment suppressed the expressions of GPX4 (glutathione peroxidase 4) and FSP1 in A549 CD133+ cells, thereby inducing ferroptosis. RSL3 and iFSP1 respectively suppressed the GSH (glutathione)-GPX4 and FSP1 (ferroptosis suppressor protein 1)-CoQ10 (coenzyme Q10)-nicotinamide adenine dinucleotide (NADH) pathways in A549 CD133+ cells. However, the roles of curcumin were blocked by Fer-1 treatment. CONCLUSIONS: In this study, curcumin induced ferroptosis through inhibiting the GSH-GPX4 and FSP1-CoQ10-NADH pathways in A549 CD133+ cells, resulting in the inhibition of their self-renewal potential.


Subject(s)
Antineoplastic Agents , Curcumin , Ferroptosis , Lung , Neoplastic Stem Cells , Humans , Animals , Mice , A549 Cells , Mice, SCID , Mice, Inbred NOD , Curcumin/administration & dosage , Signal Transduction , Ferroptosis/drug effects , Antineoplastic Agents/administration & dosage , S100 Calcium-Binding Protein A4/metabolism , Glutathione Peroxidase/metabolism , Neoplastic Stem Cells/drug effects , Lung/cytology
12.
Histol Histopathol ; 38(12): 1391-1401, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37154201

ABSTRACT

Fibroblast specific protein 1 (FSP1)/S100A4 is a calcium binding protein which has been linked to epithelial-mesenchymal transition, tissue fibrosis, pulmonary vascular disease, metastatic tumour development, increased tumour cell motility and invasiveness. This protein is reported to be also expressed in newly formed and differentiated fibroblasts and has been used in various studies to demonstrate epithelial-mesenchymal transition (EMT). We aimed to characterize S100A4 positive cells in different human tissue compartments, with the focus on fibroblasts/myofibroblast. We found S100A4 expression in a wide range of cells. Fibroblasts/myofibroblasts showed a broad spectrum of staining intensity, ranging from negative to strong expression of S100A4, with the strongest expression in smooth muscle actin positive myofibroblasts. Cells of haematopoietic lineage, namely CD4 and CD8 positive T-lymphocytes, but not B-lymphocytes expressed S100A4. All investigated monocytes, macrophages and specialised histiocytes were positive for S100A4. Even some epithelial cells of the kidney and bladder were positive for S100A4. Expression was also found in the vasculature. Here, cells of the subendothelial space, tunica adventitia and some smooth muscle cells of the tunica media were positive for S100A4. In summary, S100A4 is expressed in various cell types of different lineage and is not, as originally believed, specific for fibroblasts (FSP). Results attained under the premise of specificity of FSP1/S100A4 for fibroblasts, like the founding research on EMT type 2 in kidney and liver, therefore need to be reinterpreted.


Subject(s)
Kidney , Neoplasms , Humans , S100 Calcium-Binding Protein A4/analysis , S100 Calcium-Binding Protein A4/metabolism , Kidney/metabolism , Calcium-Binding Proteins/analysis , Calcium-Binding Proteins/metabolism , Fibrosis , Fibroblasts/metabolism , Neoplasms/metabolism
13.
Front Immunol ; 14: 1114808, 2023.
Article in English | MEDLINE | ID: mdl-37090702

ABSTRACT

Background: Fibrosis and dystrophic calcification disrupting conduction tissue architecture are histopathological lesions characterizing cardiac manifestations of neonatal lupus (cardiac-NL) associated with maternal anti-SSA/Ro antibodies. Objectives: Increased appreciation of heterogeneity in fibroblasts encourages re-examination of existing models with the consideration of multiple fibroblast subtypes (and their unique functional differences) in mind. This study addressed fibroblast heterogeneity by examining expression of α-Smooth Muscle Actin (myofibroblasts) and of S100 Calcium-Binding Protein A4 (S100A4). Methods: Using a previously established model of rheumatic scarring/fibrosis in vitro, supported by the evaluation of cord blood from cardiac-NL neonates and their healthy (anti-SSA/Ro-exposed) counterparts, and autopsy tissue from fetuses dying with cardiac-NL, the current study was initiated to more clearly define and distinguish the S100A4-positive fibroblast in the fetal cardiac environment. Results: S100A4 immunostaining was observed in 4 cardiac-NL hearts with positional identity in the conduction system at regions of dystrophic calcification but not fibrotic zones, the latter containing only myofibroblasts. In vitro, fibroblasts cultured with supernatants of macrophages transfected with hY3 (noncoding ssRNA) differentiated into myofibroblasts or S100A4+ fibroblasts. Myofibroblasts expressed collagen while S100A4+ fibroblasts expressed pro-angiogenic cytokines and proteases that degrade collagen. Cord blood levels of S100A4 in anti-SSA/Ro-exposed neonates tracked disease severity and, in discordant twins, distinguished affected from unaffected. Conclusions: These findings position the S100A4+ fibroblast alongside the canonical myofibroblast in the pathogenesis of cardiac-NL. Neonatal S100A4 levels support a novel biomarker of poor prognosis.


Subject(s)
Calcinosis , Heart Block , Infant, Newborn , Humans , Heart Block/etiology , Heart Block/pathology , Heart , Biomarkers , Fibrosis , Fibroblasts/metabolism , S100 Calcium-Binding Protein A4/metabolism
14.
Medicine (Baltimore) ; 102(14): e33466, 2023 Apr 07.
Article in English | MEDLINE | ID: mdl-37026957

ABSTRACT

S100 protein family, which represents 25 relatively small calcium binding proteins, is involved in many intracellular and/or extracellular processes, including differentiation, apoptosis, migration/invasion, Ca2+ homeostasis, inflammation, and tissue repair. As an important member, S100A4 was reported to have an abnormal expression in several lung diseases, such as lung cancer, pulmonary hypertension, idiopathic pulmonary fibrosis (IPF), etc. For example, in lung cancer, S100A4 was demonstrated to be associated to metastatic tumor progression and epithelial to mesenchymal transition (EMT). In IPF, S100A4 was considered as a promising serum biomarker predicting disease progression. Various studies in recent years focused on the S100A4 function in lung diseases, showing researchers' interests on this protein. It is necessary to focuses on relative studies, and make a comprehensive understanding of S100A4 in common pulmonary diseases. By doing this, this paper provides a review of the evidence for S100A4 in lung cancer, chronic obstructive pulmonary disease (COPD), asthma, IPF and pulmonary hypertension.


Subject(s)
Idiopathic Pulmonary Fibrosis , Lung Diseases , Lung Neoplasms , Humans , Epithelial-Mesenchymal Transition , S100 Calcium-Binding Protein A4/metabolism , Lung Neoplasms/pathology , S100 Proteins , Idiopathic Pulmonary Fibrosis/metabolism
15.
Nat Cancer ; 4(3): 419-435, 2023 03.
Article in English | MEDLINE | ID: mdl-36973439

ABSTRACT

Most tumor cells undergo apoptosis in circulation and at the metastatic organ sites due to host immune surveillance and a hostile microenvironment. It remains to be elucidated whether dying tumor cells have a direct effect on live tumor cells during the metastatic process and what the underlying mechanisms are. Here we report that apoptotic cancer cells enhance the metastatic outgrowth of surviving cells through Padi4-mediated nuclear expulsion. Tumor cell nuclear expulsion results in an extracellular DNA-protein complex that is enriched with receptor for advanced glycation endproducts (RAGE) ligands. The chromatin-bound RAGE ligand S100a4 activates RAGE receptors in neighboring surviving tumor cells, leading to Erk activation. In addition, we identified nuclear expulsion products in human patients with breast, bladder and lung cancer and a nuclear expulsion signature correlated with poor prognosis. Collectively, our study demonstrates how apoptotic cell death can enhance the metastatic outgrowth of neighboring live tumor cells.


Subject(s)
Lung Neoplasms , S100 Calcium-Binding Protein A4 , Humans , Apoptosis , Lung Neoplasms/metabolism , Receptor for Advanced Glycation End Products/metabolism , S100 Calcium-Binding Protein A4/genetics , S100 Calcium-Binding Protein A4/metabolism , Tumor Microenvironment
16.
Immun Inflamm Dis ; 11(2): e730, 2023 02.
Article in English | MEDLINE | ID: mdl-36799806

ABSTRACT

OBJECTIVE: Asthma is a chronic respiratory disease with an increasing incidence every year. microRNAs (miRNAs) have been demonstrated to have implications for asthma. However, limited information is available regarding the effect of miR-124-3p on this disease. Therefore, this study aimed to explore the possible effects of miR-124-3p and S100A4 on inflammation and epithelial-mesenchymal transition (EMT) in asthma using mouse models. METHOD: Ovalbumin was used to induce asthmatic mouse models. Lung injury in mouse models was assessed, and the bronchoalveolar lavage fluid of mice was collected to determine the number of eosinophilic granulocytes and assess inflammation. The expression levels of miR-124-3p, S100A4, E-cadherin, N-cadherin, Snail1, vimentin, and TGF-ß1/Smad2 signaling pathway-related proteins were measured using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. In vitro experiments, cells were transfected with miR-124-3p mimics or inhibitors to test the expression of S100A4 by RT-qPCR and western blot analysis, and the mutual binding of miR-124-3p and S100A4 was validated by dual-luciferase reporter gene assay. RESULTS: Overexpression of miR-124-3p or inhibition of S100A4 expression attenuated bronchial mucus secretion and collagenous fibers and suppressed inflammatory cell infiltration. Additionally, upon miR-124-3p overexpression or S100A4 suppression, eosinophilic granulocytes were decreased, interleukin-4 (IL-4) and IL-13 expression levels were reduced in the bronchoalveolar lavage fluid, serum total IgE level was reduced, and the TGF-ß1/Smad2 signaling pathway was suppressed. Mechanically, a dual-luciferase reporter gene assay verified the binding relationship between miR-124-3p and S100A4. CONCLUSION: miR-124-3p can negatively target S100A4 to attenuate inflammation in asthmatic mouse models by suppressing the EMT process and the TGF-ß/smad2 signaling pathway.


Subject(s)
Asthma , MicroRNAs , S100 Calcium-Binding Protein A4 , Animals , Mice , Asthma/etiology , Inflammation/genetics , MicroRNAs/genetics , Ovalbumin , Transforming Growth Factor beta1/metabolism , S100 Calcium-Binding Protein A4/metabolism
17.
Cells ; 12(2)2023 01 04.
Article in English | MEDLINE | ID: mdl-36672147

ABSTRACT

BACKGROUND: The infiltration of inflammatory cells during a kidney injury stimulates myofibroblast activation leading to kidney fibrosis. Fibroblast-specific protein 1 (FSP-1) positive cells have been reported as either myofibroblasts or monocytes during tissue fibrosis. The functions of FSP-1+ cells that are associated with the development of renal fibrosis and the signaling pathways that regulate FSP-1+ cell activation have not been well defined. METHODS: In mice with unilateral ureteral obstruction (UUO), we characterized FSP-1+ cells and determined the role of the Notch signaling pathway in the activation of bone marrow-derived FSP-1+ cells during kidney fibrosis. RESULTS: In kidneys from mice with UUO, the FSP-1+ cells accumulated significantly in the tubulointerstitial area. By using immunostaining and FSP-1 reporter mice, we found that FSP-1 was co-stained with inflammatory cell markers, but not myofibroblast markers. Results from mice with bone marrow transplantations showed that FSP-1+ cells in obstructed kidneys represent a bone marrow-derived population of inflammatory cells. In cultured FSP-1+ cells, the inhibition of Notch signaling suppressed the activation and cytokine secretion of FSP-1+ cells that were induced by LPS but not by IL-4. The specific KO or blockade of Notch signaling in bone marrow-derived FSP-1+ cells suppressed UUO-induced ECM deposition, the infiltration of FSP-1+ inflammatory cells, and cytokine production. These responses ameliorated myofibroblast accumulation and renal fibrosis in obstructed kidneys. CONCLUSION: Our study reveals that most FSP-1+ cells in obstructed kidneys are activated macrophages that are derived from bone marrow and that Notch signaling activates the production of M1 cytokines in FSP-1+ monocytes/macrophages, which is important for renal inflammation and fibrosis.


Subject(s)
Kidney Diseases , Ureteral Obstruction , Animals , Mice , Bone Marrow/metabolism , Cytokines/metabolism , Fibrosis , Kidney/pathology , Kidney Diseases/pathology , S100 Calcium-Binding Protein A4/metabolism , Ureteral Obstruction/complications
18.
Angiogenesis ; 26(1): 19-36, 2023 02.
Article in English | MEDLINE | ID: mdl-35829860

ABSTRACT

Tumor-induced lymphangiogenesis promotes the formation of new lymphatic vessels, contributing to lymph nodes (LNs) metastasis of tumor cells in both mice and humans. Vessel sprouting appears to be a critical step in this process. However, how lymphatic vessels sprout during tumor lymphangiogenesis is not well-established. Here, we report that S100A4 expressed in lymphatic endothelial cells (LECs) promotes lymphatic vessel sprouting in a growing tumor by regulating glycolysis. In mice, the loss of S100A4 in a whole body (S100A4-/-), or specifically in LECs (S100A4ΔLYVE1) leads to impaired tumor lymphangiogenesis and disrupted metastasis of tumor cells to sentinel LNs. Using a 3D spheroid sprouting assay, we found that S100A4 in LECs was required for the lymphatic vessel sprouting. Further investigations revealed that S100A4 was essential for the position and motility of tip cells, where it activated AMPK-dependent glycolysis during lymphatic sprouting. In addition, the expression of S100A4 in LECs was upregulated under hypoxic conditions. These results suggest that S100A4 is a novel regulator of tumor-induced lymphangiogenesis. Targeting S100A4 in LECs may be a potential therapeutic strategy for lymphatic tumor metastasis.


Subject(s)
Endothelial Cells , Lymphatic Vessels , Mice , Humans , Animals , Endothelial Cells/metabolism , Lymphatic Vessels/metabolism , Lymphangiogenesis/physiology , Lymphatic Metastasis/pathology , S100 Calcium-Binding Protein A4/genetics , S100 Calcium-Binding Protein A4/metabolism
19.
Int J Mol Sci ; 23(23)2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36499426

ABSTRACT

Cervical cancer has a poor prognosis and is the fourth most common cancer among women. Dihydromyricetin (DHM), a flavonoid compound, exhibits several pharmacological activities, including anticancer effects; however, the effects of DHM on cervical cancer have received insufficient research attention. This study examined the antitumor activity and underlying mechanisms of DHM on human cervical cancer. Our results indicated that DHM inhibits migration and invasion in HeLa and SiHa cell lines. Mechanistically, RNA sequencing analysis revealed that DHM suppressed S100A4 mRNA expression in HeLa cells. Moreover, DHM inhibited the protein expressions of ß-catenin and GSK3ß through the regulated extracellular-signal-regulated kinase (ERK)1/2 signaling pathway. By using the ERK1/2 activator, T-BHQ, reverted ß-catenin and S100A4 protein expression and cell migration, which were reduced in response to DHM. In conclusion, our study indicated that DHM inhibited cell migration by reducing the S100A4 expression through the ERK1/2/ß-catenin pathway in human cervical cancer cell lines.


Subject(s)
Flavonols , S100 Calcium-Binding Protein A4 , Uterine Cervical Neoplasms , beta Catenin , Female , Humans , beta Catenin/metabolism , Cell Movement , HeLa Cells , MAP Kinase Signaling System , S100 Calcium-Binding Protein A4/genetics , S100 Calcium-Binding Protein A4/metabolism , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Flavonols/pharmacology
20.
Redox Biol ; 58: 102543, 2022 12.
Article in English | MEDLINE | ID: mdl-36436457

ABSTRACT

Vitamin C, a key antioxidant in the central nervous system, cycles between ascorbic acid and dehydroascorbic acid under pathophysiological conditions. Clinical evidence supports that the absence of vitamin C may be linked to depressive symptoms, but much less is known about the mechanism. Herein, we show that chronic stress disrupts the expression of ascorbic acid transporter, sodium-dependent vitamin C transport 2, and induces a deficiency in endogenous ascorbic acid in the medial prefrontal cortex, leading to depressive-like behaviors by disturbing redox-dependent DNA methylation reprogramming. Attractively, ascorbic acid (100 mg/kg-1000 mg/kg, intraperitoneal injection, as bioequivalent of an intravenous drip dose of 0.48 g-4.8 g ascorbic acid per day in humans) produces rapid-acting antidepressant effects via triggering DNA demethylation catalyzed by ten-eleven translocation dioxygenases. In particular, the mechanistic studies by both transcriptome sequencing and methylation sequencing have shown that S100 calcium binding protein A4, a potentially protective factor against oxidative stress and brain injury, mediates the antidepressant activity of ascorbic acid via activating erb-b2 receptor tyrosine kinase 4 (ErbB4)-brain derived neurotrophic factor (BDNF) signaling pathway. Overall, our findings reveal a novel nutritional mechanism that couples stress to aberrant DNA methylation underlying depressive-like behaviors. Therefore, application of vitamin C may be a potential strategy for the treatment of depression.


Subject(s)
Ascorbic Acid , Sodium-Coupled Vitamin C Transporters , Humans , Ascorbic Acid/pharmacology , Ascorbic Acid/metabolism , Biological Transport , DNA/metabolism , S100 Calcium-Binding Protein A4/metabolism , Sodium-Coupled Vitamin C Transporters/genetics , Sodium-Coupled Vitamin C Transporters/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...