Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 259
Filtrar
1.
Front Genet ; 15: 1342205, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39359477

RESUMO

Background: There are no reliable biomarkers to identify Graves' disease patients who will develop severe Graves' orbitopathy (GO). We hypothesize that integrating various omics platforms can enhance our understanding of disease mechanisms and uncover potential biomarkers. This study aimed to (1) elucidate the differential gene expression profile of orbital fibroblasts in GO during early adipogenesis to better understand disease mechanisms and (2) compare tear protein profiles from our earlier study and the transcriptome profiles of orbital fibroblasts (OFs) to identify possible biomarkers of the disease. Methods: OFs were grown from orbital adipose tissue obtained from nine GO patients (three for discovery and six for validation experiments). Total RNA was extracted from OFs on day 0 as the baseline for each sample and from differentiated OFs on days 4 and 8. Protein-protein interaction (PPI) analysis and functional enrichment analysis were also carried out. The differentially expressed genes (DEGs) from the RNA sequencing experiments were then compared to the full tear proteome profile from the author's previous study, which examined the tear protein changes of GO patients based on fold change > 1.6 or < -1.6. FDR < 0.05 was applied within all datasets. Further validation of S100 calcium-binding protein A4 (S100A4) downregulation in GO was performed via quantitative real-time PCR (qPCR). Results: The whole transcriptomic analysis revealed 9 upregulated genes and 15 downregulated genes in common between the discovery and validation experiments. From the PPI network analysis, an interaction network containing six identified DEGs (ALDH2, MAP2K6, MT2A, SOCS3, S100A4, and THBD) was observed. The functional enrichment network analysis identified a set of genes related to oxysterol production. S100A4 was found to be consistently downregulated in both our transcriptome studies and the full-tear proteome profile from the author's previous study. Conclusion: Our study identified several DEGs and potential gene pathways in GO patients, which concurred with the results of other studies. Tear S100A4 may serve as a biomarker for the propensity to develop thyroid eye disease (TED) in patients with autoimmune thyroid disease (AITD) before clinical manifestation and should be confirmed in future studies.

2.
Int Immunopharmacol ; 142(Pt A): 112849, 2024 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-39241524

RESUMO

Multiple sclerosis (MS) is a neurodegenerating autoimmune disease with no clinical cure currently. The calcium-binding protein S100A4 has been demonstrated to exert regulatory roles in inflammatory disorders including MS. However, the precise mechanisms by which S100A4 regulates neuroinflammation in MS remains unknown. To investigate the regulatory effect of S100A4 on microglial inflammation and its impact on neuroinflammation, the mouse-derived microglia cell line BV2 cells were infected with lentivirus to knockout S100A4 for in vitro studies. Wild-type (WT) and S100A4-/- mice were induced to develop experimental autoimmune encephalomyelitis (EAE), an animal model of MS, for in vivo investigation. Results indicated that the frequencies of microglia in the spinal cord and brain and the expression of S100A4 in these tissues varied kinetically along with the progression of the disease in mice with EAE. S100A4-/- mice presented ameliorated clinical scores of EAE and exhibited less severe EAE signs, including inflammatory cell infiltration in the spinal cord and brain and demyelination of the spinal cord. Moreover, these mice demonstrated overall reduced levels of inflammatory cytokines in the spinal cord and brain. Compromised systematic inflammatory responses including circulating cytokines and frequencies of immune cells in the spleen were also observed in these mice. In addition, both exogenous and endogenous S100A4 could promote the microglial inflammation, affect the polarization of microglia and enhance inflamed microglia-mediated apoptosis of neuronal cells through TLR4/NF-κB signaling pathway. Thus, S100A4 may participate in the regulation of neuroinflammation at least partly through regulating the inflammation of microglia.


Assuntos
Encefalomielite Autoimune Experimental , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia , NF-kappa B , Proteína A4 de Ligação a Cálcio da Família S100 , Transdução de Sinais , Receptor 4 Toll-Like , Animais , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Microglia/imunologia , Microglia/metabolismo , NF-kappa B/metabolismo , Camundongos , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100/genética , Medula Espinal/patologia , Medula Espinal/imunologia , Medula Espinal/metabolismo , Feminino , Citocinas/metabolismo , Esclerose Múltipla/imunologia , Linhagem Celular , Inflamação/imunologia , Encéfalo/patologia , Encéfalo/imunologia , Encéfalo/metabolismo
4.
Biochim Biophys Acta Mol Basis Dis ; 1871(1): 167498, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39243827

RESUMO

BACKGROUND: Acute myeloid leukemia (AML) is an immunosuppressive hematologic malignancy with a poor prognosis. An immunosuppressive microenvironment blunts AML therapy. However, the prognostic and therapeutic roles of the factors that mediate immunosuppression in AML remain elusive. METHODS: S100 calcium-binding protein A4 (S100A4) was identified as an immunosuppression-mediating factor by analyzing The Cancer Genome Atlas AML project (TCGA-LAML) transcriptome data and data from AML-bearing mice and AML patients. The S100A4-mediated signaling pathway in myeloid-derived suppressor cells (MDSCs) was evaluated. RESULTS: Elevated S100A4 expression was positively associated with worse survival of AML patients, MDSCs, macrophages and immune checkpoints. S100A4 silencing downregulated the expression levels of MDSC-associated CD14, CCR2 and CCL2, reduced MDSC expansion and impaired MDSC-mediated inhibition of T cell activation and proliferation. S100A4-based prognostic signature (SPS) was an independent risk factor for AML patients. The high-risk group based on SPS was not only associated with adverse survival, MDSCs and macrophages and immune checkpoints but also insensitive to 25 chemotherapy drugs. It was also found that CCAAT enhancer binding protein beta (CEBPB) mediated S100A4 transcription. CEBPB silencing downregulated the expression levels of MDSC-associated CD14, CCR2 and CCL2. Mechanistically, S100A4 activated GP130/JAK2/STAT3 signaling in MDSCs by interacting with the cytokine-binding domain of GP130. Moreover, S100A4 mediated MDSC expansion through JAK2/STAT3 signaling. CONCLUSION: This study uncovers the critical role of S100A4 in MDSC accumulation, and S100A4-based prognostic signature may guide chemotherapy sensitivity in patients with AML.

5.
Vaccine ; 42(25): 126151, 2024 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-39089961

RESUMO

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), remains one of the top three causes of death. Currently, the only licensed vaccine against TB is the bacillus Calmette-Guerin (BCG), which lacks efficacy in preventing and controlling pulmonary TB in adults. We aimed to evaluate a nasal TB vaccine formulation composed of the Mtb-specific vaccine antigen ESAT-6, an Mtb-associated protein that can trigger protective immune responses, and S100A4, a recently characterized novel mucosal adjuvant. Mice were intranasally given recombinant ESAT-6 in the presence or absence of S100A4 as an adjuvant. We have provided experimental evidence demonstrating that S100A4 admixed to ESAT-6 could induce Mtb-specific adaptive immune responses after intranasal immunization. S100A4 remarkably augmented the levels of anti-ESAT-6 IgG in serum and IgA in mucosal sites, including lung exudates, bronchoalveolar lavage fluid (BALF) and nasal lavage. Furthermore, in both lung and spleen tissues, S100A4 strongly promoted ESAT-6-specific expansion of CD4 T cells. Both CD4 and CD8 T cells from these tissues expressed increased levels of IFN-γ, TNF-α, and IL-17, cytokines critical for antimicrobial activity. Antigen-reencounter-induced T cell proliferative responses, a key vaccine performance indicator, were augmented in the spleen of S100A4-adjuvanted mice. Furthermore, CD8 T cells from the spleen and lung tissues of these mice expressed higher levels of granzyme B upon antigen re-stimulation. S100A4-adjuvanted immunization may predict good mucosal protection against TB.


Assuntos
Adjuvantes Imunológicos , Administração Intranasal , Antígenos de Bactérias , Proteínas de Bactérias , Mycobacterium tuberculosis , Proteína A4 de Ligação a Cálcio da Família S100 , Vacinas contra a Tuberculose , Animais , Vacinas contra a Tuberculose/imunologia , Vacinas contra a Tuberculose/administração & dosagem , Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Adjuvantes Imunológicos/administração & dosagem , Camundongos , Mycobacterium tuberculosis/imunologia , Feminino , Proteína A4 de Ligação a Cálcio da Família S100/imunologia , Linfócitos T CD4-Positivos/imunologia , Tuberculose/prevenção & controle , Tuberculose/imunologia , Adjuvantes de Vacinas/administração & dosagem , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Linfócitos T CD8-Positivos/imunologia , Imunidade nas Mucosas , Imunoglobulina A/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Citocinas/metabolismo , Camundongos Endogâmicos C57BL , Baço/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Camundongos Endogâmicos BALB C
6.
Cureus ; 16(7): e65615, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-39205741

RESUMO

Background Colorectal cancer (CRC) remains a major cause of morbidity and mortality worldwide. Understanding the clinical and pathological characteristics of CRC patients is essential for improving diagnosis, treatment, and prognostication. S100 proteins play a crucial role in CRC by promoting tumor growth, metastasis, and inflammation through their involvement in various cellular processes such as proliferation, migration, and immune response modulation. Elevated levels of specific S100 proteins have been associated with poor prognosis and serve as potential biomarkers for early detection and therapeutic targets in CRC. This study aims to analyze the general and medical characteristics of CRC patients, with a particular focus on the expression patterns of S100A4 and S100A14 proteins and their correlation with tumor location and various clinical parameters. Methods This cross-sectional study included 98 CRC patients aged 21 to 92 years. Clinical data were collected from Vajeen Hospital (Duhok/ Iraq), including age, gender, and presenting symptoms. Pathological data such as tumor site, tumor size, tumor, node, and metastasis (TNM) stage, tumor grade angio-lymphatic invasion, perineural invasion, and metastasis were analyzed. The expression of S100A4 and S100A14 proteins was assessed using immunohistochemistry, and their correlation with clinico-pathological features and tumor location was evaluated using statistical analysis. Results The 98 patients with a mean age of 57.27 years. The majority were over 50 years old (68, 69.39%) with a nearly equal gender distribution. The most common symptom was bleeding per rectum (36, 36.74%). TNM staging revealed 25.51% (n=25) of patients at stage I, 32.65% (n=32) at stage II, 24.49% (n=24) at stage III, and 17.35% (n=17) at stage IV. Angio-lymphatic invasion was present in 65.31% (n=64) of patients, and lymph node invasion in 38.78% (n=38). All tumors were adenocarcinomas, with 82.65% (n=81) being intermediate grade. S100A4 expression was low in early-stage tumors but significantly higher in advanced stages (P < 0.0001). High S100A4 expression was associated with vascular invasion (P = 0.0006), perineural invasion (P = 0.0002), lymph node invasion (P < 0.0001), and metastasis (P = 0.0010). S100A14 expression was inversely correlated with disease severity. Low S100A14 expression was more common in advanced stages (P < 0.0001) and was associated with higher rates of vascular invasion (P = 0.0018), lymph node invasion (P < 0.0001), and metastasis (P = 0.0001). Conclusion This study highlights significant correlations between S100A4 and S100A14 expression with various clinico-pathological features in CRC patients. High S100A4 expression is linked with tumor aggressiveness, whereas low S100A14 expression is associated with advanced disease stages and increased metastasis. However, there is no observed correlation between the expression of these proteins and the tumor site.

7.
Environ Toxicol ; 39(9): 4447-4458, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38953363

RESUMO

VPS9D1-AS1 functions as an oncogene in many cancers. However, its role and potential mechanism in the progression of endometrial cancer (EC) are not fully understood. VPS9D1-AS1 levels in EC and adjacent normal tissues were investigated using the TCGA-UCEC cohort and 24 paired clinical samples. The roles of VPS9D1-AS1 and miR-187-3p in cell cycle, proliferation, and apoptosis were evaluated by loss- and gain-of-function experiments. In addition, the effect of VPS9D1-AS1 on tumor growth was further investigated in vivo. Rescue experiments were performed to investigate the involvement of the miR-187-3p/S100A4 axis in VPS9D1-AS1 knockdown-mediated antitumor effects. VPS9D1-AS1 was highly expressed in EC tissues. VPS9D1-AS1 knockdown, similar to miR-187-3p overexpression, significantly inhibited cell proliferation, inhibited colony formation, induced cell cycle arrest, and facilitated apoptosis of KLE cells. MiR-187-3p bound directly to VPS9D1-AS1 and the 3'UTR of S100A4. Furthermore, VPS9D1-AS1 negatively regulated miR-187-3p while positively regulating S100A4 expression in EC cells. MiR-187-3p knockdown or S100A4 overexpression partially reversed the tumor suppressive function of VPS9D1-AS1 knockdown. The results suggest that VPS9D1-AS1 affects EC progression by regulating the miR-187-3p/S100A4 axis. This may provide a promising therapeutic target to help treat EC.


Assuntos
Proliferação de Células , Neoplasias do Endométrio , MicroRNAs , RNA Longo não Codificante , Proteína A4 de Ligação a Cálcio da Família S100 , Humanos , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Feminino , MicroRNAs/genética , MicroRNAs/metabolismo , Linhagem Celular Tumoral , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Proliferação de Células/genética , Proteína A4 de Ligação a Cálcio da Família S100/genética , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Animais , Apoptose/genética , Progressão da Doença , Camundongos Nus , Regulação Neoplásica da Expressão Gênica , Camundongos , Camundongos Endogâmicos BALB C
8.
Biochem Biophys Res Commun ; 727: 150309, 2024 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-38936224

RESUMO

Versican is a large chondroitin sulfate proteoglycan in the extracellular matrix. It plays a pivotal role in the formation of the provisional matrix. S100a4, previously known as fibroblast-specific protein, functions as a calcium channel-binding protein. To investigate the role of versican expressed in fibroblasts, we generated conditional knockout mice in which versican expression is deleted in cells expressing S100a4. We found that S100a4 is expressed in adipose tissues, and these mice exhibit obesity under a normal diet, which becomes apparent as early as five months. The white adipose tissues of these mice exhibited decreased expression levels of S100a4 and versican and hypertrophy of adipocytes. qRT-PCR showed a reduced level of UCP1 in their white adipose tissues, indicating that the basic energy metabolism is diminished. These results suggest that versican in adipose tissues maintains the homeostasis of adipose tissues and regulates energy metabolism.


Assuntos
Tecido Adiposo , Metabolismo Energético , Homeostase , Camundongos Knockout , Versicanas , Animais , Versicanas/metabolismo , Versicanas/genética , Camundongos , Tecido Adiposo/metabolismo , Obesidade/metabolismo , Obesidade/genética , Tecido Adiposo Branco/metabolismo , Camundongos Endogâmicos C57BL , Masculino , Adipócitos/metabolismo
9.
Front Vet Sci ; 11: 1410371, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38872805

RESUMO

Background: Gonadotrophin-releasing hormone (GnRH) administration significantly decreases the pregnancy rate of recipient ewes after embryo transfer, possibly because GnRH affects endometrial epithelial cell function. Therefore, this study investigated the effect of GnRH on endometrial epithelial cells. Methods: Transcriptome sequencing was used to determine the regulatory effect of GnRH on the ewe endometrium, and the S100A4 gene, which showed altered transcription, was screened as a candidate regulator of this effect. Endometrial epithelial cells were further isolated, the S100A4 protein was immunoprecipitated, and host proteins that interacted with S100A4 were identified by mass spectrometry. We further verified the effects of S100A4 and GNAI2 on the proliferation of endometrial epithelial cells via overexpression/knockdown experiments and subsequent CCK-8 and EdU assays. The effect of S100A4 deletion in endometrial cells on reproduction was verified in mice with S100A4 knockout. Results: Our results showed that S100A4 gene transcription in endometrial cells was significantly inhibited after GnRH administration. GNAI2 was identified as a downstream interacting protein of S100A4, and S100A4 was confirmed to activate the MAPK signaling pathway to promote cell proliferation by targeting GNAI2. Conclusion: GnRH can suppress the expression of S100A4 in the endometrium, consequently inhibiting the proliferation of endometrial cells through the S100A4/GNAI2/MAPK signaling pathway. These findings suggest a potential explanation for the limited efficacy of GnRH in promoting embryo implantation.

10.
Int J Mol Sci ; 25(12)2024 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-38928339

RESUMO

Receptors of cytokines are major regulators of the immune response. In this work, we have discovered two new ligands that can activate the TNFR1 (tumor necrosis factor receptor 1) receptor. Earlier, we found that the peptide of the Tag (PGLYRP1) protein designated 17.1 can interact with the TNFR1 receptor. Here, we have found that the Mts1 (S100A4) protein interacts with this peptide with a high affinity (Kd = 1.28 × 10-8 M), and that this complex is cytotoxic to cancer cells that have the TNFR1 receptor on their surface. This complex induces both apoptosis and necroptosis in cancer cells with the involvement of mitochondria and lysosomes in cell death signal transduction. Moreover, we have succeeded in locating the Mts1 fragment that is responsible for protein-peptide interaction, which highly specifically interacts with the Tag7 protein (Kd = 2.96 nM). The isolated Mts1 peptide M7 also forms a complex with 17.1, and this peptide-peptide complex also induces the TNFR1 receptor-dependent cell death. Molecular docking and molecular dynamics experiments show the amino acids involved in peptide binding and that may be used for peptidomimetics' development. Thus, two new cytotoxic complexes were created that were able to induce the death of tumor cells via the TNFR1 receptor. These results may be used in therapy for both cancer and autoimmune diseases.


Assuntos
Apoptose , Receptores Tipo I de Fatores de Necrose Tumoral , Humanos , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/química , Apoptose/efeitos dos fármacos , Ligação Proteica , Simulação de Acoplamento Molecular , Linhagem Celular Tumoral , Peptídeos/química , Peptídeos/farmacologia , Peptídeos/metabolismo , Simulação de Dinâmica Molecular , Transdução de Sinais/efeitos dos fármacos , Necroptose/efeitos dos fármacos , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Oligopeptídeos/metabolismo , Citocinas
11.
Cell Calcium ; 119: 102869, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38484433

RESUMO

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a pleiotropic myelopoietic growth factor and proinflammatory cytokine, clinically used for multiple indications and serving as a promising target for treatment of many disorders, including cancer, multiple sclerosis, rheumatoid arthritis, psoriasis, asthma, COVID-19. We have previously shown that dimeric Ca2+-bound forms of S100A6 and S100P proteins, members of the multifunctional S100 protein family, are specific to GM-CSF. To probe selectivity of these interactions, the affinity of recombinant human GM-CSF to dimeric Ca2+-loaded forms of 18 recombinant human S100 proteins was studied by surface plasmon resonance spectroscopy. Of them, only S100A4 protein specifically binds to GM-CSF with equilibrium dissociation constant, Kd, values of 0.3-2 µM, as confirmed by intrinsic fluorescence and chemical crosslinking data. Calcium removal prevents S100A4 binding to GM-CSF, whereas monomerization of S100A4/A6/P proteins disrupts S100A4/A6 interaction with GM-CSF and induces a slight decrease in S100P affinity for GM-CSF. Structural modelling indicates the presence in the GM-CSF molecule of a conserved S100A4/A6/P-binding site, consisting of the residues from its termini, helices I and III, some of which are involved in the interaction with GM-CSF receptors. The predicted involvement of the 'hinge' region and F89 residue of S100P in GM-CSF recognition was confirmed by mutagenesis. Examination of S100A4/A6/P ability to affect GM-CSF signaling showed that S100A4/A6 inhibit GM-CSF-induced suppression of viability of monocytic THP-1 cells. The ability of the S100 proteins to modulate GM-CSF activity is relevant to progression of various neoplasms and other diseases, according to bioinformatics analysis. The direct regulation of GM-CSF signaling by extracellular forms of the S100 proteins should be taken into account in the clinical use of GM-CSF and development of the therapeutic interventions targeting GM-CSF or its receptors.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos , Proteínas S100 , Humanos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Proteínas S100/metabolismo , Proteínas Recombinantes/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/química , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Ligação Proteica , Sítios de Ligação
12.
Int J Biochem Cell Biol ; 169: 106551, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38360265

RESUMO

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.


Assuntos
Próstata , Hiperplasia Prostática , Proteína A4 de Ligação a Cálcio da Família S100 , Idoso , Animais , Humanos , Masculino , Ratos , Apoptose , Proliferação de Células , Fibrose , Próstata/metabolismo , Hiperplasia Prostática/metabolismo , Proteína A4 de Ligação a Cálcio da Família S100/genética , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo
13.
Emerg Microbes Infect ; 13(1): 2300466, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38164719

RESUMO

During its global epidemic, Zika virus (ZIKV) attracted widespread attention due to its link with various severe neurological symptoms and potential harm to male fertility. However, the understanding of how ZIKV invades and persists in the male reproductive system is limited due to the lack of immunocompetent small animal models. In this study, immunocompetent murine models were generated by using anti-IFNAR antibody blocked C57BL/6 male mice and human STAT2 (hSTAT2) knock in (KI) male mice. After infection, viral RNA could persist in the testes even after the disappearance of viremia. We also found a population of ZIKV-susceptible S100A4+ monocytes/macrophages that were recruited into testes from peripheral blood and played a crucial role for ZIKV infection in the testis. By using single-cell RNA sequencing, we also proved that S100A4+ monocytes/macrophages had a great impact on the microenvironment of ZIKV-infected testes, thus promoting ZIKV-induced testicular lesions. In conclusion, this study proposed a novel mechanism of long-term ZIKV infection in the male reproductive system.


Assuntos
Infecção por Zika virus , Zika virus , Humanos , Masculino , Camundongos , Animais , Zika virus/genética , Testículo , Monócitos , Camundongos Endogâmicos C57BL , Macrófagos , Modelos Animais de Doenças , Proteína A4 de Ligação a Cálcio da Família S100
14.
Cell Commun Signal ; 22(1): 27, 2024 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-38200591

RESUMO

BACKGROUND: Claudin-18.2 (CLDN18.2) has emerged as an alluring therapeutic target against gastrointestinal tumors in recent years. However, a thorough understanding of its regulatory mechanism in gastric cancer remains elusive. METHODS: We presented a comprehensive study comprising 185 gastric cancer patients, which included 112 cases with high CLDN18.2 expression and 73 cases with low CLDN18.2 expression as determined by immunohistochemistry. After overdressed CLDN18.2 in AGS and NUGC4 cell lines, we elucidated the functions of CLDN18.2 in connecting gastric cancer cells and cancer-associated fibroblasts (CAFs) through an in vitro adhesion models and in vivo lung colonization models. The molecular mechanism underlying CLDN18.2-mediated interaction between gastric cancer cells and CAFs was identified through RNA sequencing and protein-proximity labeling techniques in vivo. RESULTS: In our own cohort, a correlation was observed between high levels of CLDN18.2 expression and advanced cancer stage, poor prognosis, and heightened infiltration of CAFs. We elucidated a pivotal role of CLDN18.2 in mediating adhesion between gastric cancer cells and CAFs, which leads to the adhesion of cancer cells to stroma tissue and facilitates the clustering of cancer cells and CAFs into embolus, enhancing gastric cancer's metastatic progression and the risk of embolic death. Mechanistically, it was discovered that CAFs can activate adhesion and metastasis-related signaling pathways in CLDN18.2-positive gastric cancer cells. Furthermore, using an in vivo protein-proximity labeling approach, we identified S100 calcium binding protein A4 (S100A4) as a distinctive marker of CAFs that interacts with CLDN18.2 to enhance gastric cancer progression. CONCLUSIONS: Our findings illuminated the role of the CLDN18.2-mediated interaction between cancer cells and CAFs in promoting gastric cancer progression and embolism, thereby providing insight into potential therapeutic avenues for CLDN18.2 positive cancers. Video Abstract.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias Gástricas , Humanos , Moléculas de Adesão Celular , Linhagem Celular , Claudinas
15.
Int Immunopharmacol ; 128: 111555, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38280333

RESUMO

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.


Assuntos
Asma , Mastócitos , Animais , Camundongos , Humanos , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Mastócitos/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Metabolismo dos Lipídeos , Transdução de Sinais , Asma/metabolismo
16.
Matrix Biol ; 127: 1-7, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38219976

RESUMO

Fibrosis regardless of aetiology is characterised by persistently activated myofibroblasts that are contractile and secrete excessive amounts of extracellular matrix molecules that leads to loss of organ function. Damage-Associated Molecular Patterns (DAMPs) are endogenous host-derived molecules that are released from cells dying or under stress that can be triggered by a variety of insults, either chemical or physical, leading to an inflammatory response. Among these DAMPs is S100A4, part of the S100 family of calcium binding proteins that participate in a variety of cellular processes. S100A4 was first described in context of cancer as a pro-metastatic factor. It is now appreciated that aside from its role in cancer promotion, S100A4 is intimately involved in tissue fibrosis. The extracellular form of S100A4 exerts its effects through multiple receptors including Toll-Like Receptor 4 and RAGE to evoke signalling cascades involving downstream mediators facilitating extracellular matrix deposition and myofibroblast generation and can play a role in persistent activation of myofibroblasts. S100A4 may be best understood as an amplifier of inflammatory and fibrotic processes. S100A4 appears critical in systemic sclerosis pathogenesis and blocking the extracellular form of S100A4 in vivo in various animal models of disease mitigates fibrosis and may even reverse established disease. This review appraises S100A4's position as a DAMP and its role in fibrotic conditions and highlight therapeutically targeting this protein to halt fibrosis, suggesting that it is a tractable target.


Assuntos
Neoplasias , Escleroderma Sistêmico , Animais , Fibrose , Miofibroblastos , Transdução de Sinais , Humanos
17.
Int J Biol Sci ; 20(1): 29-46, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38164183

RESUMO

Background: Thoracic aortic dissection (TAD) is one of the cardiovascular diseases with high incidence and fatality rates. Vascular smooth muscle cells (VSMCs) play a vital role in TAD formation. Recent studies have shown that extracellular S100A4 may participate in VSMCs regulation. However, the mechanism(s) underlying this association remains elusive. Consequently, this study investigated the role of S100A4 in VSMCs regulation and TAD formation. Methods: Hub genes were screened based on the transcriptome data of aortic dissection in the Gene Expression Synthesis database. Three-week-old male S100A4 overexpression (AAV9- S100A4 OE) and S100A4 knockdown (AAV9- S100A4 KD) mice were exposed to ß-aminopropionitrile monofumarate through drinking water for 28 days to create the murine TAD model. Results: S100A4 was observed to be the hub gene in aortic dissection. Furthermore, overexpression of S100A4 was exacerbated, whereas inhibition of S100A4 significantly improved TAD progression. In the TAD model, the S100A4 was observed to aggravate the phenotypic transition of VSMCs. Additionally, lysyl oxidase (LOX) was an important target of S100A4 in TAD. S100A4 interacted with LOX in VSMCs, reduced mature LOX (m-LOX), and decreased elastic fiber deposition, thereby disrupting extracellular matrix homeostasis and promoting TAD development. Elastic fiber deposition in human aortic tissues was negatively correlated with the expression of S100A4, which in turn, was negatively correlated with LOX. Conclusions: Our data showed that S100A4 modulates TADprogression, induces lysosomal degradation of m-LOX, and reduces the deposition of elastic fibers by interacting with LOX, thus contributing to the disruption of extracellular matrix homeostasis in TAD. These findings suggest that S100A4 may be a new target for the prevention and treatment of TAD.


Assuntos
Dissecção Aórtica , Dissecção da Aorta Torácica , Masculino , Humanos , Camundongos , Animais , Dissecção Aórtica/genética , Aorta , Matriz Extracelular , Proteína A4 de Ligação a Cálcio da Família S100/genética
18.
J Histochem Cytochem ; 72(2): 79-94, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38264898

RESUMO

Abdominal hernia is a protruding weakness in the abdominal wall. It affects abdominal strength and life quality and can lead to complications due to intestinal entrapment. Autologous full-thickness skin graft (FTSG) has recently become an alternative material for reinforcement in the surgical repair of large abdominal hernias instead of synthetic mesh. FTSG eventually integrates with the abdominal wall, but the long-term fate of the graft itself is not fully understood. This has implications as to how these grafts should be optimally used and handled intraoperatively. This study investigates the remodeling of FTSG in either the onlay or the intraperitoneal position 8 weeks after FTSG transplantation in an experimental mouse model. There was a significant presence of fibroblasts, indicated by vimentin and S100A4 staining, but there were significant variations among animals as to how much of the graft had been remodeled into dense connective tissue. This correlated significantly with the proportion of vimentin-positive cells in the dense connective tissue. We also found that collagen hybridizing peptide staining intensity, a marker of active remodeling, was significantly associated with the proportion of S100A4-positive cells in the dense connective tissue of the FTSG.


Assuntos
Parede Abdominal , Transplante de Pele , Animais , Camundongos , Vimentina , Parede Abdominal/cirurgia , Modelos Animais de Doenças , Matriz Extracelular
19.
J Biol Chem ; 300(1): 105530, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38072048

RESUMO

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.


Assuntos
Fibrose Pulmonar Idiopática , Mecanotransdução Celular , Miofibroblastos , Proteína A4 de Ligação a Cálcio da Família S100 , Animais , Camundongos , Transdiferenciação Celular , Fibrose , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Proteína A4 de Ligação a Cálcio da Família S100/genética , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo
20.
Mol Neurobiol ; 61(4): 2228-2240, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37872355

RESUMO

Retinal ischemia exists in various ischemic retinopathies including glaucoma, contributing to the death of retinal neurons. Calcium binding protein S100A4 is important in tumors, and our previous study found that S100A4 protects retinal ganglion cells (RGCs) against retinal ischemia-reperfusion (I/R) injury. This study was aimed to further discuss the neuroprotection and mechanisms of S100A4 in retinal I/R of mice. The rAAV-EF1α-s100a4-EGFP-WPRE or rAAV-EF1α-EGFP-WPRE-Pa was injected intravitreally 4 weeks before I/R. S100A4, molecules in TLR4 signaling pathway and endoplasmic reticulum (ER) stress branches, inflammatory molecules, and surviving RGCs and cholinergic amacrine (ChAT) cells were determined by quantitative PCR, western blot, or immunofluorescent staining. The apoptosis and necrosis of retinal neurons induced by I/R were inhibited by overexpressed S100A4. RGCs, ChAT cells, and the retinal function were preserved by S100A4 overexpressing 7 days after I/R. Mechanistically, the beneficial effects of S100A4 may be mediated by inhibiting the activation of TLR4 signaling pathway and alleviating ER stress, leading to the attenuation of inflammatory response of the retina after I/R. Our findings indicated that S100A4 has neuroprotective effect against retinal I/R injury, and promoting S100A4 expression may be an effective strategy to inhibit retinal neurons from degeneration in ischemic retinopathy.


Assuntos
Traumatismo por Reperfusão , Doenças Retinianas , Animais , Camundongos , Estresse do Retículo Endoplasmático , Isquemia/metabolismo , NF-kappa B/metabolismo , Traumatismo por Reperfusão/metabolismo , Retina/metabolismo , Doenças Retinianas/metabolismo , Receptor 4 Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...