Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 488
Filter
1.
J Transl Med ; 22(1): 347, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38605354

ABSTRACT

BACKGROUND: THOC7-AS1 and FSTL1 expression are frequently upregulated in cutaneous squamous cell carcinoma (cSCC). However, their molecular biological mechanisms remain elusive and their potential as therapeutic targets needs urgent exploration. METHODS: Human tissue samples were used to evaluate clinical parameters. In vitro and in vivo experiments assessed biological functions. Quantitative PCR, western blot, immunohistochemistry, immunocytochemistry, immunoprecipitation, RNA fluorescence in situ hybridization, RNA pull-down, RNA immunoprecipitation, silver staining, chromatin immunoprecipitation, dual luciferase reporter assays etc. were utilized to explore the molecular biological mechanisms. RESULTS: We found FSTL1 is an oncogene in cSCC, with high expression in tumor tissues and cells. Its elevated expression closely associates with tumor size and local tissue infiltration. In vitro and in vivo, high FSTL1 expression promotes cSCC proliferation, migration and invasion, facilitating malignant behaviors. Mechanistically, FSTL1 interacts with ZEB1 to promote epithelial-to-mesenchymal transition (EMT) in cSCC cells. Exploring upstream regulation, we found THOC7-AS1 can interact with OCT1, which binds the FSTL1 promoter region and promotes FSTL1 expression, facilitating cSCC progression. Finally, treating tumors with THOC7-AS1 antisense oligonucleotides inhibited cSCC proliferative and migratory abilities, delaying tumor progression. CONCLUSIONS: The THOC7-AS1/OCT1/FSTL1 axis regulates EMT and promotes tumor progression in cSCC. This study provides clues and ideas for cSCC targeted therapy.


Subject(s)
Carcinoma, Squamous Cell , Follistatin-Related Proteins , Skin Neoplasms , Humans , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , Gene Expression Regulation, Neoplastic , In Situ Hybridization, Fluorescence , RNA , RNA, Long Noncoding/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology
2.
J Cancer Res Clin Oncol ; 150(3): 124, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38478111

ABSTRACT

BACKGROUND: Cancer-associated fibroblasts (CAF) play a critical role in promoting tumor growth, metastasis, and immune evasion. While numerous studies have investigated CAF, there remains a paucity of research on their clinical application in colorectal cancer (CRC). METHODS: In this study, we collected differentially expressed genes between CAF and normal fibroblasts (NF) from previous CRC studies, and utilized machine learning analysis to differentiate two distinct subtypes of CAF in CRC. To enable practical application, a CAF-related genes (CAFGs) scoring system was developed based on multivariate Cox regression. We then conducted functional enrichment analysis, Kaplan-Meier plot, consensus molecular subtypes (CMS) classification, and Tumor Immune Dysfunction and Exclusion (TIDE) algorithm to investigate the relationship between the CAFGs scoring system and various biological mechanisms, prognostic value, tumor microenvironment, and response to immune checkpoint blockade (ICB) therapy. Moreover, single-cell transcriptomics and proteomics analyses have been employed to validate the significance of scoring system-related molecules in the identity and function of CAF. RESULTS: We unveiled significant distinctions in tumor immune status and prognosis not only between the CAF clusters, but also across high and low CAFGs groups. Specifically, patients in CAF cluster 2 or with high CAFGs scores exhibited higher CAF markers and were enriched for CAF-related biological pathways such as epithelial-mesenchymal transition (EMT) and angiogenesis. In addition, CAFGs score was identified as a risk index and correlated with poor overall survival (OS), progression-free survival (PFS), disease-free survival (DFS), and recurrence-free survival (RFS). High CAFGs scores were observed in patients with advanced stages, CMS4, as well as lymphatic invasion. Furthermore, elevated CAFG scores in patients signified a suppressive tumor microenvironment characterized by the upregulation of programmed death-ligand 1 (PD-L1), T-cell dysfunction, exclusion, and TIDE score. And high CAFGs scores can differentiate patients with lower response rates and poor prognosis under ICB therapy. Notably, single-cell transcriptomics and proteomics analyses identified several molecules related to CAF identity and function, such as FSTL1, IGFBP7, and FBN1. CONCLUSION: We constructed a robust CAFGs score system with clinical significance using multiple CRC cohorts. In addition, we identified several molecules related to CAF identity and function that could be potential intervention targets for CRC patients.


Subject(s)
Cancer-Associated Fibroblasts , Colorectal Neoplasms , Follistatin-Related Proteins , Humans , Multiomics , Fibroblasts , Algorithms , Colorectal Neoplasms/genetics , Tumor Microenvironment/genetics , Prognosis
3.
Sci Rep ; 14(1): 6435, 2024 03 18.
Article in English | MEDLINE | ID: mdl-38499600

ABSTRACT

Hyperparathyroidism (HPT) manifests as a complex condition with a substantial disease burden. While advances have been made in surgical interventions and non-surgical pharmacotherapy for the management of hyperparathyroidism, radical options to halt underlying disease progression remain lacking. Identifying putative genetic drivers and exploring novel drug targets that can impede HPT progression remain critical unmet needs. A Mendelian randomization (MR) analysis was performed to uncover putative therapeutic targets implicated in hyperparathyroidism pathology. Cis-expression quantitative trait loci (cis-eQTL) data serving as genetic instrumental variables were obtained from the eQTLGen Consortium and Genotype-Tissue Expression (GTEx) portal. Hyperparathyroidism summary statistics for single nucleotide polymorphism (SNP) associations were sourced from the FinnGen study (5590 cases; 361,988 controls). Colocalization analysis was performed to determine the probability of shared causal variants underlying SNP-hyperparathyroidism and SNP-eQTL links. Five drug targets (CMKLR1, FSTL1, IGSF11, PIK3C3 and SLC40A1) showed significant causation with hyperparathyroidism in both eQTLGen and GTEx cohorts by MR analysis. Specifically, phosphatidylinositol 3-kinase catalytic subunit type 3 (PIK3C3) and solute carrier family 40 member 1 (SLC40A1) showed strong evidence of colocalization with HPT. Multivariable MR and Phenome-Wide Association Study analyses indicated these two targets were not associated with other traits. Additionally, drug prediction analysis implies the potential of these two targets for future clinical applications. This study identifies PIK3C3 and SLC40A1 as potential genetically proxied druggable genes and promising therapeutic targets for hyperparathyroidism. Targeting PIK3C3 and SLC40A1 may offer effective novel pharmacotherapies for impeding hyperparathyroidism progression and reducing disease risk. These findings provide preliminary genetic insight into underlying drivers amenable to therapeutic manipulation, though further investigation is imperative to validate translational potential from preclinical models through clinical applications.


Subject(s)
Follistatin-Related Proteins , Hyperparathyroidism , Humans , Mendelian Randomization Analysis , Quantitative Trait Loci/genetics , Class III Phosphatidylinositol 3-Kinases , Cost of Illness , Genome-Wide Association Study
4.
Cell Biol Int ; 48(6): 795-807, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38436106

ABSTRACT

Mitochondrial dysfunction and myocardial remodeling have been reported to be the main underlying molecular mechanisms of doxorubicin-induced cardiotoxicity. SIRT6 is a nicotinamide adenine dinucleotide-dependent enzyme that plays a vital role in cardiac protection against various stresses. Moreover, previous studies have demonstrated that FSTL1 could alleviate doxorubicin-induced cardiotoxicity by inhibiting autophagy. The present study investigated the probable mechanisms of FSTL1 on doxorubicin-induced cardiotoxicity in vivo and in vitro. We confirmed that FSTL1 exerted a pivotal protective role on cardiac tissue in vivo and on doxorubicin-induced cell injury in vitro. Furthermore, FSTL1 can alleviate doxorubicin-induced mitochondrial dysfunction by inhibiting autophagy and apoptosis. Further studies demonstrated that FSTL1 can activate SIRT6 signaling by restoring the SIRT6 protein expression in doxorubicin-induced myocardial injury. SIRT6 activation elevated the protein expression of Nrf2 in doxorubicin-induced H9C2 injury. Treatment with the Nrf2 inhibitor ML385 partially antagonized the cardioprotective role of SIRT6 on doxorubicin-induced autophagy or apoptosis. These results suggested that the protective mechanism of FSTL1 on doxorubicin-induced cardiotoxicity may be related with the inhibition of autophagy and apoptosis, partly through the activation of SIRT6/Nrf2.


Subject(s)
Apoptosis , Autophagy , Cardiotoxicity , Doxorubicin , Follistatin-Related Proteins , NF-E2-Related Factor 2 , Signal Transduction , Sirtuins , Sirtuins/metabolism , Doxorubicin/adverse effects , Doxorubicin/toxicity , Animals , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Cardiotoxicity/metabolism , Cardiotoxicity/prevention & control , Rats , Apoptosis/drug effects , Male , Autophagy/drug effects , Follistatin-Related Proteins/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Mice , Cell Line , Mice, Inbred C57BL
5.
Front Immunol ; 15: 1332279, 2024.
Article in English | MEDLINE | ID: mdl-38390317

ABSTRACT

Background: As the leading cause of chronic kidney disease, diabetic kidney disease (DKD) is an enormous burden for all healthcare systems around the world. However, its early diagnosis has no effective methods. Methods: First, gene expression data in GEO database were extracted, and the differential genes of diabetic tubulopathy were obtained. Immune-related genesets were generated by WGCNA and immune cell infiltration analyses. Then, differentially expressed immune-related cuproptosis genes (DEICGs) were derived by the intersection of differential genes and genes related to cuproptosis and immune. To investigate the functions of DEICGs, volcano plots and GO term enrichment analysis was performed. Machine learning and protein-protein interaction (PPI) network analysis helped to finally screen out hub genes. The diagnostic efficacy of them was evaluated by GSEA analysis, receiver operating characteristic (ROC) curve, single-cell RNA sequencing and the Nephroseq website. The expression of hub genes at the animal level by STZ -induced and db/db DKD mouse models was further verified. Results: Finally, three hub genes, including FSTL1, CX3CR1 and AGR2 that were up-regulated in both the test set GSE30122 and the validation set GSE30529, were screened. The areas under the curve (AUCs) of ROC curves of hub genes were 0.911, 0.935 and 0.922, respectively, and 0.946 when taking as a whole. Correlation analysis showed that the expression level of three hub genes demonstrated their negative relationship with GFR, while those of FSTL1 displayed a positive correlation with the level of serum creatinine. GSEA was enriched in inflammatory and immune-related pathways. Single-nucleus RNA sequencing indicated the main distribution of FSTL1 in podocyte and mesangial cells, the high expression of CX3CR1 in leukocytes and the main localization of AGR2 in the loop of Henle. In mouse models, all three hub genes were increased in both STZ-induced and db/db DKD models. Conclusion: Machine learning was combined with WGCNA, immune cell infiltration and PPI analyses to identify three hub genes associated with cuproptosis, immunity and diabetic nephropathy, which all have great potential as diagnostic markers for DKD and even predict disease progression.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Follistatin-Related Proteins , Animals , Mice , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/genetics , Machine Learning , Area Under Curve , Databases, Factual
6.
Heart Vessels ; 39(6): 563-570, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38381171

ABSTRACT

Cardiovascular diseases (CVD) are the leading cause of death globally. In recent years, follistatin-like protein 1 (FSTL1) has been proposed as an emerging potential clinical biomarker of CVD, since its concentration is upregulated in heart failure. The aim of the present study was to evaluate the association of FSTL1 levels and classic biomarkers with the risk of CVD in Mexican population. A case-control study was carried out in patients with cardiovascular diseases (CVD), arterial hypertension, but not CVD (cardiovascular risk factor-CRF), and healthy controls (control group) from the Mexican Institute of Social Security. Lipid profile, homocysteine (Hcys), serum amyloid A (SAA), FSTL1 concentration, PON1 concentration and activities [Arylesterase (ARE), and Lactonase (LAC)] were evaluated. High levels of FSTL1 were found in the CRF group and a positive association of FSTL1 (OR = 4.55; 95% CI 1.29-16.04, p = 0.02) with the presence of arterial hypertension, as well as Hcys (OR, 3.09; 95% CI 1.23-7.76, p = 0.02) and SAA (OR, 1.03; 95% CI 1.01-1.05, p < 0.01) with the presence of CVD. LAC activity (OR, 0.26; 95% CI 0.07-0.94, p = 0.04) and PON1 concentration (OR, 0.17; 95% CI 0.05-0.62, p = 0.01) were associated with a decrease in OR belonging to the group with CVD. Our results suggest that FSTL1 may be a useful biomarker for monitoring cardiovascular risk in clinical settings. However, longitudinal studies are needed to evaluate how FSTL1 could influence the association of PON1 activity and Hcys with CVD.


Subject(s)
Biomarkers , Cardiovascular Diseases , Follistatin-Related Proteins , Aged , Female , Humans , Male , Middle Aged , Aryldialkylphosphatase/blood , Biomarkers/blood , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/blood , Case-Control Studies , Follistatin-Related Proteins/blood , Hypertension/epidemiology , Hypertension/blood , Hypertension/diagnosis , Mexico/epidemiology , Risk Assessment/methods , Risk Factors
7.
Gene ; 906: 148263, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38346455

ABSTRACT

Flolistatin-related protein 1 (FSTL1), a secreted glycoprotein that is involved in many physiological functions, has attracted much interest and has been implicated in a wide range of diseases, including heart diseases and inflammatory diseases. In recent years, the involvement of FSTL1 in cancer progression has been implicated and researched. FSTL1 plays a contradictory role in cancer, depending on the cancer type as well as the contents of the tumor microenvironment. As reviewed here, the structure and distribution of FSTL1 are first introduced. Subsequently, the expression and clinical significance of FSTL1 in various types of cancer as a tumor enhancer or inhibitor are addressed. Furthermore, we discuss the functional role of FSTL1 in various processes that involve tumor cell proliferation, metastasis, immune responses, stemness, cell apoptosis, and resistance to chemotherapy. FSTL1 expression is tightly controlled in cancer, and a multitude of cancer-related signaling cascades like TGF-ß/BMP/Smad signaling, AKT, NF-κB, and Wnt-ß-catenin signaling pathways are modulated by FSTL1. Finally, FSTL1 as a therapeutic target using monoclonal antibodies is stated. Herein, we review recent findings showing the double-edged characteristics and mechanisms of FSTL1 in cancer and elaborate on the current understanding of therapeutic approaches targeting FSTL1.


Subject(s)
Follistatin-Related Proteins , Neoplasms , Humans , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , NF-kappa B/metabolism , Tumor Microenvironment , Wnt Signaling Pathway , Animals
8.
Sci China Life Sci ; 67(3): 475-487, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37219765

ABSTRACT

Cardiopulmonary bypass has been speculated to elicit systemic inflammation to initiate acute lung injury (ALI), including acute respiratory distress syndrome (ARDS), in patients after cardiac surgery. We previously found that post-operative patients showed an increase in endothelial cell-derived extracellular vesicles (eEVs) with components of coagulation and acute inflammatory responses. However, the mechanism underlying the onset of ALI owing to the release of eEVs after cardiopulmonary bypass, remains unclear. Plasma plasminogen-activated inhibitor-1 (PAI-1) and eEV levels were measured in patients with cardiopulmonary bypass. Endothelial cells and mice (C57BL/6, Toll-like receptor 4 knockout (TLR4-/-) and inducible nitric oxide synthase knockout (iNOS-/-)) were challenged with eEVs isolated from PAI-1-stimulated endothelial cells. Plasma PAI-1 and eEVs were remarkably enhanced after cardiopulmonary bypass. Plasma PAI-1 elevation was positively correlated with the increase in eEVs. The increase in plasma PAI-1 and eEV levels was associated with post-operative ARDS. The eEVs derived from PAI-1-stimulated endothelial cells could recognize TLR4 to stimulate a downstream signaling cascade identified as the Janus kinase 2/3 (JAK2/3)-signal transducer and activator of transcription 3 (STAT3)-interferon regulatory factor 1 (IRF-1) pathway, along with iNOS induction, and cytokine/chemokine production in vascular endothelial cells and C57BL/6 mice, ultimately contributing to ALI. ALI could be attenuated by JAK2/3 or STAT3 inhibitors (AG490 or S3I-201, respectively), and was relieved in TLR4-/- and iNOS-/- mice. eEVs activate the TLR4/JAK3/STAT3/IRF-1 signaling pathway to induce ALI/ARDS by delivering follistatin-like protein 1 (FSTL1), and FSTL1 knockdown in eEVs alleviates eEV-induced ALI/ARDS. Our data thus demonstrate that cardiopulmonary bypass may increase plasma PAI-1 levels to induce FSTL1-enriched eEVs, which target the TLR4-mediated JAK2/3/STAT3/IRF-1 signaling cascade and form a positive feedback loop, leading to ALI/ARDS after cardiac surgery. Our findings provide new insight into the molecular mechanisms and therapeutic targets for ALI/ARDS after cardiac surgery.


Subject(s)
Acute Lung Injury , Extracellular Vesicles , Follistatin-Related Proteins , Respiratory Distress Syndrome , Animals , Humans , Mice , Acute Lung Injury/etiology , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Follistatin-Related Proteins/metabolism , Follistatin-Related Proteins/therapeutic use , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Lung/metabolism , Mice, Inbred C57BL , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 1/therapeutic use , Respiratory Distress Syndrome/etiology , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/therapeutic use
9.
Mol Cell Biochem ; 479(1): 171-181, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37138144

ABSTRACT

Intervertebral disc degeneration (IDD) causes pain in the back and neck. This study investigated the role of long non-coding RNA HLA complex group 18 (HCG18) in a cell model of IDD. An IDD model was established by stimulating nucleus pulposus (NP) cells with interleukin (IL)-1ß. MTT assay was performed to evaluate NP cell viability. The apoptosis was detected by flow cytometry. The expressions of HCG18, microRNA (miR)-495-3p, and follistatin-like protein-1 (FSTL1) were measured by RT-qPCR. The interactions of miR-495-3p with HCG18 and FSTL1 were analyzed by luciferase reporter assay. IL-1ß stimulation upregulated HCG18 and FSTL1, but downregulated miR-495-3p in NP cells. Silencing of HCG18 or FSTL1, as well as miR-495-3p overexpression in NP cells alleviated IL-1ß-induced apoptosis and inflammation of NP cells. Both HCG18 and FSTL1 had binding sites for miR-495-3p. Overexpression of FSTL1 abolished the effects of HCG18 silencing on IL-1ß-induced apoptosis and inflammation. The HCG18/miR-495-3p/FSTL1 axis is essential for IDD development. Therapeutic strategies targeting this axis may be used for IDD treatment.


Subject(s)
Follistatin-Related Proteins , Intervertebral Disc Degeneration , MicroRNAs , RNA, Long Noncoding , Humans , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Follistatin-Related Proteins/genetics , Apoptosis , Interleukin-1beta/metabolism , Inflammation/genetics
10.
Obesity (Silver Spring) ; 32(2): 352-362, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38018497

ABSTRACT

OBJECTIVE: The aim of this study was to investigate the role of the follistatin-like 1 (Fstl1) and disco-interacting protein 2 homolog A (DIP2a) axis in relation to lipid metabolism during and after endurance exercise and to elucidate the mechanisms underlying the metabolic effects of Fstl1 on adipocytes, considering its regulation by exercise and muscle mass and its link to obesity. METHODS: Twenty-nine sedentary males participated in endurance exercise, and blood samples were collected during and after the exercise. Body composition, Fstl1, glycerol, epinephrine, growth hormone, and atrial natriuretic peptide were measured. 3T3-L1 adipocytes, with or without DIP2a knockdown, were treated with Fstl1 to assess glycerol release, cyclic AMP/cyclic GMP production, and hormone sensitive lipase phosphorylation. The association between DIP2a gene expression levels in human adipose tissues and exercise-induced lipolysis was examined. RESULTS: Fstl1 levels significantly increased during endurance exercise and following recovery, correlating with lean body mass and lipolysis. In 3T3-L1 adipocytes, Fstl1 increased glycerol release, cyclic GMP production, and hormone sensitive lipase activation, but these effects were attenuated by DIP2a knockdown. DIP2a gene expression in human adipose tissues correlated with serum glycerol concentrations during endurance exercise. CONCLUSIONS: Fstl1 is a myokine facilitating lipid mobilization during and after endurance exercise through DIP2a-mediated lipolytic effects in adipocytes.


Subject(s)
Follistatin-Related Proteins , Follistatin , Humans , Male , Cyclic GMP/metabolism , Follistatin/metabolism , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , Glycerol/metabolism , Lipid Mobilization , Lipolysis/physiology , Myokines , Sterol Esterase/metabolism
11.
Clin Exp Hypertens ; 45(1): 2277654, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37963199

ABSTRACT

OBJECTIVE: Endothelial dysfunction is a critical initiating factor in the development of hypertension and related complications. Follistatin-like 1 (FSTL1) can promote endothelial cell function and stimulates revascularization in response to ischemic insult. However, it is unclear whether FSTL1 has an effect on ameliorating endothelial dysfunction in spontaneously hypertensive rats (SHRs). METHODS: Wistar Kyoto (WKY) and SHRs were treated with a tail vein injection of vehicle (1 mL/day) or recombinant FSTL1 (100 µg/kg body weight/day) for 4 weeks. Blood pressure was measured by tail-cuff plethysmograph, and vascular reactivity in mesenteric arteries was measured using wire myography. RESULTS: We found that treatment with FSTL1 reversed impaired endothelium-dependent relaxation (EDR) in mesenteric arteries and lowered blood pressure of SHRs. Decreased AMP-activated protein kinase (AMPK) phosphorylation, elevated endoplasmic reticulum (ER) stress markers, increased reactive oxygen species (ROS), and reduction of nitric oxide (NO) production in mesenteric arteries of SHRs were also reversed by FSTL1 treatment. Ex vivo treatment with FSTL1 improved the impaired EDR in mesenteric arteries from SHRs and reversed tunicamycin (ER stress inducer)-induced ER stress and the impairment of EDR in mesenteric arteries from WKY rats. The effects of FSTL1 were abolished by cotreatment of compound C (AMPK inhibitor). CONCLUSIONS: These results suggest that FSTL1 prevents endothelial dysfunction in mesenteric arteries of SHRs through inhibiting ER stress and ROS and increasing NO production via activation of AMPK signaling.


Subject(s)
Follistatin-Related Proteins , Hypertension , Rats , Animals , Rats, Inbred SHR , AMP-Activated Protein Kinases/metabolism , Follistatin/metabolism , Follistatin/pharmacology , Rats, Inbred WKY , Reactive Oxygen Species/metabolism , Follistatin-Related Proteins/metabolism , Follistatin-Related Proteins/pharmacology , Endothelium, Vascular , Mesenteric Arteries , Endoplasmic Reticulum Stress
12.
Oncol Rep ; 50(6)2023 Dec.
Article in English | MEDLINE | ID: mdl-37888756

ABSTRACT

The abnormal expression of follistatin­like protein 1 (FSTL1) in various tumors is a crucial regulator of the biological process of tumorigenesis. Nonetheless, the regulatory role of FSTL1 in cervical cancer is yet to be elucidated. Hence, the present study aimed to explore the expression, function, and molecular mechanism of FSTL1 in cervical cancer. The expression of FSTL1 in normal and cervical cancer tissues was examined using quantitative reverse transcription­polymerase chain reaction and immunohistochemistry assays. The effects of abnormal expression of FSTL1 on cervical cancer cells were assessed using colony formation, MTT, wound­healing, Transwell, apoptosis, and nude mouse tumorigenicity assays. FSTL1­related molecular mechanisms were screened using gene chip analysis. Western blotting analysis was used to verify the regulatory mechanisms of FSTL1 in cervical cancer. The results indicated that the expression of FSTL1 was downregulated in cervical cancer tissues and that its downregulation was associated with tumor differentiation, pathologic type, and infiltration depth. Moreover, FSTL1 inhibited the proliferation, migration, and invasion of cervical cancer cells as well as xenograft tumor growth and promoted cell apoptosis. In addition, the findings of gene chip analysis suggested that the differentially expressed genes of FSTL1 were predominantly enriched in multiple signaling pathways, of which the insulin­like growth factor (IGF)­1 signaling pathway was significantly activated. Western blotting suggested the involvement of FSTL1 in the regulation of the IGF­1R/PI3K/AKT/BCL­2 signaling pathway. These data establish the downregulation of FSTL1 in cervical cancer tissues. FSTL1 inhibited the proliferation, migration, and invasion of cervical cancer cells and promoted their apoptosis. Furthermore, xenograft tumor growth in nude mice was inhibited. FSTL1 may be involved in the regulation of the IGF­1R/PI3K/AKT/BCL­2 signaling pathway in cervical cancer. Therefore, FSTL1 may be employed as a novel biomarker to determine the extent of disease progression in patients with cervical cancer.


Subject(s)
Follistatin-Related Proteins , Uterine Cervical Neoplasms , Animals , Female , Mice , Humans , Proto-Oncogene Proteins c-akt/metabolism , Uterine Cervical Neoplasms/pathology , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , Cell Proliferation/genetics , Mice, Nude , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Movement/genetics
13.
Biomed Pharmacother ; 168: 115771, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37897975

ABSTRACT

Disco Interacting Protein 2 Homolog A (DIP2A) is expressed throughout the body and abundantly expressed in the brain tissue. It is activated by Follistatin-like 1 (FSTL1). Activated DIP2A interacts with several pathways, such as AMPK/mTOR and AKT pathways, to contribute to many biological processes, such as oxidative stress, transcriptional regulation, and apoptosis. Dysregulated DIP2A activation has been implicated in numerous processes in the brain. If the upstream pathways of DIP2A remain globally unexplored, many proteins, including cortactin, AMPK, and AKT, have been identified as its downstream targets in the literature. Recent studies have linked DIP2A to a variety of mechanisms in many types of brain disorders, suggesting that regulation of DIP2A could provide novel diagnostic and therapeutic approaches for brain disorders. In this review, we comprehensively summarized and discussed the current research on DIP2A in various brain disorders, such as stroke, autism spectrum disorders (ASD), Alzheimer's disease (AD), dyslexia, and glioma.


Subject(s)
Brain Diseases , Follistatin-Related Proteins , Humans , Proto-Oncogene Proteins c-akt/metabolism , AMP-Activated Protein Kinases/metabolism , Nuclear Proteins/genetics , Gene Expression Regulation , Follistatin-Related Proteins/metabolism
14.
Cardiovasc Diabetol ; 22(1): 297, 2023 10 30.
Article in English | MEDLINE | ID: mdl-37904173

ABSTRACT

BACKGROUND: The study aimed to investigate an association of increased liver fibrosis with acute myocardial infarction (AMI), and to investigate the mediating effect of serum follistatin-like protein 3 (FSTL3) on the association in patients with type 2 diabetes mellitus (T2DM). METHOD: A total of 1424 participants were included in this study, and were firstly divided into two groups: 429 T2DM patients and 995 T2DM patients with NAFLD to assess the association of NAFLD and AMI. Then 995 T2DM co-existent NAFLD patients were categorized by NAFLD fibrosis risk to explore the association between NAFLD fibrosis risk and AMI. Immunohistochemistry staining and semi-quantitative analysis of liver FSTL3 were performed in 60 patients with NAFLD. There were 323 individuals (191 without AMI and 132 with AMI) in T2DM co-existent NAFLD patients who had serum samples, and serum FSTL3 was tested and mediation effect of FSTL3 in association of NAFLD fibrosis and AMI was performed. RESULTS: First, increased NAFLD fibrosis risk was an independent risk factor for AMI in patients with T2DM and co-existent NAFLD. In addition, analysis of Gene Expression Omnibus (GEO) database and immunohistochemical staining confirmed the increased expression of FSTL3 in the liver of NAFLD patients with fibrosis. Serum FSTL3 significantly increased in patients with high NAFLD fibrosis risk and AMI, and closely associated with NAFLD fibrosis and AMI severity in T2DM patients with co-existent NAFLD. Most importantly, analysis of the level of mediation revealed that increased serum FSTL3 partially mediated the association of increased NAFLD fibrosis risk with AMI in T2DM patients with co-existent NAFLD. CONCLUSIONS: NAFLD fibrosis was closely associated with AMI in T2DM patients. FSTL3 expression was enriched in the liver of NAFLD patients with significant and advanced fibrosis, and serum FSTL3 partially mediated the association of increased liver fibrosis risk with AMI in T2DM patients.


Subject(s)
Diabetes Mellitus, Type 2 , Follistatin-Related Proteins , Myocardial Infarction , Non-alcoholic Fatty Liver Disease , Humans , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/epidemiology , Follistatin-Related Proteins/genetics , Liver Cirrhosis/diagnosis , Myocardial Infarction/diagnosis , Myocardial Infarction/epidemiology , Myocardial Infarction/complications , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/epidemiology , Non-alcoholic Fatty Liver Disease/complications
15.
Redox Biol ; 67: 102923, 2023 11.
Article in English | MEDLINE | ID: mdl-37832398

ABSTRACT

As the predominant immunosuppressive component within the tumor microenvironment (TME), cancer-associated fibroblasts (CAFs) inhibit Natural Killer cell (NK cell) activity to promote tumor progression and immune escape; however, the mechanisms of cross-talk between CAFs and NK cells in gastric cancer (GC) remain poorly understood. In this study, we demonstrate that NK cell levels are inversely correlated with CAFs abundance in human GC. CAFs impair the anti-tumor capacity of NK cells by inducing ferroptosis, a cell death process characterized by the accumulation of iron-dependent lipid peroxides. CAFs induce ferroptosis in NK cells by promoting iron overload; conversely, decreased intracellular iron levels protect NK cells against CAF-induced ferroptosis. Mechanistically, CAFs increase the labile iron pool within NK cells via iron export into the TME, which is mediated by the upregulated expression of iron regulatory genes ferroportin1 and hephaestin in CAFs. Moreover, CAF-derived follistatin like protein 1(FSTL1) upregulates NCOA4 expression in NK cells via the DIP2A-P38 pathway, and NCOA4-mediated ferritinophagy is required for CAF-induced NK cell ferroptosis. In a human patient-derived organoid model, functional targeting of CAFs using a combination of deferoxamine and FSTL1-neutralizing antibody significantly alleviate CAF-induced NK cell ferroptosis and boost the cytotoxicity of NK cells against GC. This study demonstrates a novel mechanism of suppression of NK cell activity by CAFs in the TME and presents a potential therapeutic approach to augment the immune response against GC mediated by NK cells.


Subject(s)
Antineoplastic Agents , Cancer-Associated Fibroblasts , Ferroptosis , Follistatin-Related Proteins , Stomach Neoplasms , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Follistatin-Related Proteins/metabolism , Stomach Neoplasms/metabolism , Iron/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Antineoplastic Agents/pharmacology , Tumor Microenvironment
16.
Biomolecules ; 13(9)2023 08 28.
Article in English | MEDLINE | ID: mdl-37759715

ABSTRACT

Knee osteoarthritis (KOA) is one of the most common progressive, age-dependent chronic degenerative joint diseases. KOA often develops as a result of a gradual articular cartilage loss caused by its wear and tear. Numerous studies suggest that the degradation of the knee joint involves inflammatory components. This process is also associated with body composition, particularly being overweight and muscle mass loss. The present study aimed to search for novel circulating KOA inflammatory biomarkers, taking into account body composition characteristics. To this aim, we recruited 98 patients diagnosed and radiologically confirmed with KOA and 519 healthy controls from the Arab community in Israel. A panel of soluble molecules, related to inflammatory, metabolic, and musculoskeletal disorders, was measured by ELISA in plasma samples, while several body composition parameters were assessed with bioimpedance analysis. Statistical analysis, including multivariable logistic regression, revealed a number of the factors significantly associated with KOA, independently of age and sex. The most significant independent associations [OR (95% CI)] were fat body mass/body weight index-1.56 (1.20-2.02), systemic immune-inflammation index-4.03 (2.23-7.27), circulating vaspin levels-1.39 (1.15-1.68), follistatin/FSTL1 ratio-1.32 (1.02-1.70), and activin A/FSTL1 ratio-1.33 (1.01-1.75). Further clinical studies are warranted to confirm the relevance of these KOA-associated biological factors. Hereafter, they could serve as reliable biomarkers for KOA in the general human population.


Subject(s)
Follistatin-Related Proteins , Osteoarthritis, Knee , Humans , Knee Joint , Body Composition , Biomarkers
17.
Nat Commun ; 14(1): 6047, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37770480

ABSTRACT

Inter-organ crosstalk has gained increasing attention in recent times; however, the underlying mechanisms remain unclear. In this study, we elucidate an endocrine pathway that is regulated by skeletal muscle interferon regulatory factor (IRF) 4, which manipulates liver pathology. Skeletal muscle specific IRF4 knockout (F4MKO) mice exhibited ameliorated hepatic steatosis, inflammation, and fibrosis, without changes in body weight, when put on a nonalcoholic steatohepatitis (NASH) diet. Proteomics analysis results suggested that follistatin-like protein 1 (FSTL1) may constitute a link between muscles and the liver. Dual luciferase assays showed that IRF4 can transcriptionally regulate FSTL1. Further, inducing FSTL1 expression in the muscles of F4MKO mice is sufficient to restore liver pathology. In addition, co-culture experiments confirmed that FSTL1 plays a distinct role in various liver cell types via different receptors. Finally, we observed that the serum FSTL1 level is positively correlated with NASH progression in humans. These data indicate a signaling pathway involving IRF4-FSTL1-DIP2A/CD14, that links skeletal muscle cells to the liver in the pathogenesis of NASH.


Subject(s)
Follistatin-Related Proteins , Non-alcoholic Fatty Liver Disease , Mice , Humans , Animals , Non-alcoholic Fatty Liver Disease/metabolism , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , Liver/metabolism , Signal Transduction/physiology , Muscle, Skeletal/metabolism , Liver Cirrhosis/pathology , Mice, Inbred C57BL
18.
J Mother Child ; 27(1): 119-133, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37595293

ABSTRACT

BACKGROUND: Within the endocrine-paracrine signalling network at the maternal-foetal interface, the activin-inhibin-follistatin system modulates extravillous trophoblast invasion, suggesting a potential role in preeclampsia pathogenesis. This study aimed to compile the evidence published in the last decade regarding the variation in maternal serum activins, inhibin- and follistatin-related proteins in preeclamptic pregnancies compared to healthy pregnancies, and to discuss their role in predicting and understanding the pathophysiology of preeclampsia. MATERIAL AND METHODS: A scoping review was conducted in MEDLINE, EMBASE and LILACS databases to identify studies published within the last ten years (2012-2022). RESULTS: Thirty studies were included. None of the studies addressed maternal serum changes of isoforms different from activin A, inhibin A, follistatin, and follistatin-like 3. Sixteen studies evaluated the potential of these isoforms in predicting preeclampsia through the area under the curve from a receiver operating characteristic curve. CONCLUSIONS: In preeclampsia, inhibin A is upregulated in all trimesters, whereas activin A increases exclusively in the late second and third trimesters. Serum follistatin levels are reduced in women with preeclampsia during the late second and third trimesters. However, changes in follistatin-like 3 remain inconclusive. Inhibin A and activin A can potentially serve as biomarkers of early-onset preeclampsia based on the outcomes of the receiver operating characteristic curve analysis. Further investigations are encouraged to explore the feasibility of quantifying maternal serum levels of activin A and inhibin A as a clinical tool in early preeclampsia prediction.


Subject(s)
Follistatin-Related Proteins , Pre-Eclampsia , Pregnancy , Female , Humans , Follistatin , Pre-Eclampsia/diagnosis , Inhibins , Activins
19.
FASEB J ; 37(8): e23064, 2023 08.
Article in English | MEDLINE | ID: mdl-37440271

ABSTRACT

Off target damage to vital organ systems is an unfortunate side effect of cancer chemotherapy and remains a major limitation to the use of these essential drugs in the clinic. Despite decades of research, the mechanisms conferring susceptibility to chemotherapy driven cardiotoxicity and hepatotoxicity remain unclear. In the livers of patients with a history of chemotherapy, we observed a twofold increase in expression of G protein regulator RGS7 and a corresponding decrease in fellow R7 family member RGS11. Knockdown of RGS7 via introduction of RGS7 shRNA via tail vein injection decreased doxorubicin-induced hepatic collagen and lipid deposition, glycogen accumulation, and elevations in ALT, AST, and triglycerides by approximately 50%. Surprisingly, a similar result could be achieved via introduction of RGS7 shRNA directly to the myocardium without impacting RGS7 levels in the liver directly. Indeed, doxorubicin-treated cardiomyocytes secrete the endocrine factors transforming growth factor ß1 (TGFß1) and TGFß superfamily binding protein follistatin-related protein 1 (FSTL1). Importantly, RGS7 overexpression in the heart was sufficient to recapitulate the impacts of doxorubicin on the liver and inhibition of TGFß1 signaling with the receptor blocker GW788388 ameliorated the effect of cardiac RGS7 overexpression on hepatic fibrosis, steatosis, oxidative stress, and cell death as well as the resultant elevation in liver enzymes. Together these data demonstrate that RGS7 controls both the release of TGFß1 from the heart and the profibrotic and pro-oxidant actions of TGFß1 in the liver and emphasize the functional significance of endocrine cardiokine signaling in the pathogenesis of chemotherapy drive multiorgan damage.


Subject(s)
Follistatin-Related Proteins , RGS Proteins , Humans , RGS Proteins/genetics , RGS Proteins/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Signal Transduction/physiology , Carrier Proteins/metabolism , Liver/metabolism , Doxorubicin/adverse effects , Follistatin-Related Proteins/metabolism
20.
Genomics ; 115(5): 110677, 2023 09.
Article in English | MEDLINE | ID: mdl-37406975

ABSTRACT

The matricellular protein, follistatin-like 1 (FSTL1), regulates lung development and saccular formation. Here, we employed single-cell RNA sequencing (scRNA-seq) to construct a transcriptomic atlas of 22,774 individual cells from wild-type (WT) and Fstl1-/- lung (E18.5) samples and identified 27 cell subtypes. We observed abnormal population sizes and gene expression profiles in diverse cell subtypes in Fstl1-/- lung samples. We identified Pdgfra and Tgfbi as genetic markers specifically expressed in postnatal myofibroblasts (MyoFBs). Fstl1 deletion decreased the number of MyoFB cells and downregulated their roles in ECM organization and muscle tissue/vasculature development, partly through the TGF-ß1/BMP4 signaling pathway. Our data provide a single-cell view of the cellular heterogeneity and the molecular mechanisms underlying abnormal saccular formation and atelectatic lungs in Fstl1-/- mice.


Subject(s)
Follistatin-Related Proteins , Lung , Myofibroblasts , Animals , Mice , Follistatin-Related Proteins/genetics , Follistatin-Related Proteins/metabolism , Lung/metabolism , Myofibroblasts/metabolism , Signal Transduction , Single-Cell Gene Expression Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...