Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 797
Filtrar
1.
J Neuroimmune Pharmacol ; 19(1): 17, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38717643

RESUMEN

In our previous study, we concluded that sirtuin 5 (SIRT5) was highly expressed in microglia following ischaemic stroke, which induced excessive neuroinflammation and neuronal injury. Therefore, SIRT5-targeting interventions should reduce neuroinflammation and protect against ischaemic brain injury. Here, we showed that treatment with a specific SIRT5 inhibitor, MC3482, alleviated microglia-induced neuroinflammation and improved long-term neurological function in a mouse model of stroke. The mice were administrated with either vehicle or 2 mg/kg MC3482 daily for 7 days via lateral ventricular injection following the onset of middle cerebral artery occlusion. The outcome was assessed by a panel of tests, including a neurological outcome score, declarative memory, sensorimotor tests, anxiety-like behavior and a series of inflammatory factors. We observed a significant reduction of infarct size and inflammatory factors, and the improvement of long-term neurological function in the early stages during ischaemic stroke when the mice were treated with MC3482. Mechanistically, the administration of MC3482 suppressed the desuccinylation of annexin-A1, thereby promoting its membrane recruitment and extracellular secretion, which in turn alleviated neuroinflammation during ischaemic stroke. Based on our findings, MC3482 offers promise as an anti-ischaemic stroke treatment that targets directly the disease's underlying factors.


Asunto(s)
Anexina A1 , Accidente Cerebrovascular Isquémico , Ratones Endogámicos C57BL , Microglía , Enfermedades Neuroinflamatorias , Regulación hacia Arriba , Animales , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Masculino , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Anexina A1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Sirtuinas/metabolismo , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo
2.
Commun Biol ; 7(1): 514, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710749

RESUMEN

Acute lung injury (ALI) is characterized by respiratory failure resulting from the disruption of the epithelial and endothelial barriers as well as immune system. In this study, we evaluated the therapeutic potential of airway epithelial cell-derived extracellular vesicles (EVs) in maintaining lung homeostasis. We isolated human bronchial epithelial cell-derived EVs (HBEC-EVs), which endogenously express various immune-related surface markers and investigated their immunomodulatory potential in ALI. In ALI cellular models, HBEC-EVs demonstrated immunosuppressive effects by reducing the secretion of proinflammatory cytokines in both THP-1 macrophages and HBECs. Mechanistically, these effects were partially ascribed to nine of the top 10 miRNAs enriched in HBEC-EVs, governing toll-like receptor-NF-κB signaling pathways. Proteomic analysis revealed the presence of proteins in HBEC-EVs involved in WNT and NF-κB signaling pathways, pivotal in inflammation regulation. ANXA1, a constituent of HBEC-EVs, interacts with formyl peptide receptor (FPR)2, eliciting anti-inflammatory responses by suppressing NF-κB signaling in inflamed epithelium, including type II alveolar epithelial cells. In a mouse model of ALI, intratracheal administration of HBEC-EVs reduced lung injury, inflammatory cell infiltration, and cytokine levels. Collectively, these findings suggest the therapeutic potential of HBEC-EVs, through their miRNAs and ANXA1 cargo, in mitigating lung injury and inflammation in ALI patients.


Asunto(s)
Lesión Pulmonar Aguda , Anexina A1 , Células Epiteliales , Vesículas Extracelulares , Receptores de Formil Péptido , Receptores de Lipoxina , Transducción de Señal , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/patología , Humanos , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/trasplante , Anexina A1/metabolismo , Anexina A1/genética , Animales , Ratones , Receptores de Formil Péptido/metabolismo , Receptores de Formil Péptido/genética , Células Epiteliales/metabolismo , Bronquios/metabolismo , Bronquios/citología , Masculino , Ratones Endogámicos C57BL , MicroARNs/metabolismo , MicroARNs/genética , FN-kappa B/metabolismo , Citocinas/metabolismo , Células THP-1
3.
Front Biosci (Landmark Ed) ; 29(4): 158, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38682206

RESUMEN

BACKGROUND: Immunogenic cell death (ICD) is a crucial mechanism for triggering the adaptive immune response in cancer patients. Damage-associated molecular patterns (DAMPs) are critical factors in the detection of ICD. Chemotherapeutic drugs can cause ICD and the release of DAMPs. The aim of this study was to assess the potential for paclitaxel and platinum-based chemotherapy regimens to induce ICD in squamous cell carcinoma (SCC) cell lines. In addition, we examined the immunostimulatory effects of clinically relevant chemotherapeutic regimens utilized in the treatment of SCC. METHODS: We screened for differentially expressed ICD markers in the supernatants of three SCC cell lines following treatment with various chemotherapeutic agents. The ICD markers included Adenosine Triphosphate (ATP), Calreticulin (CRT), Annexin A1 (ANXA 1), High Mobility Group Protein B1 (HMGB1), and Heat Shock Protein 70 (HSP70). A vaccination assay was also employed in C57BL/6J mice to validate our in vitro findings. Lastly, the levels of CRT and HMGB1 were evaluated in Serum samples from SCC patients. RESULTS: Addition of the chemotherapy drugs cisplatin (DDP), carboplatin (CBP), nedaplatin (NDP), oxaliplatin (OXA) and docetaxel (DOC) increased the release of ICD markers in two of the SCC cell lines. Furthermore, mice that received vaccinations with cervical cancer cells treated with DDP, CBP, NDP, OXA, or DOC remained tumor-free. Although CBP induced the release of ICD-associated molecules in vitro, it did not prevent tumor growth at the vaccination site in 40% of mice. In addition, both in vitro and in vivo results showed that paclitaxel (TAX) and LBP did not induce ICD in SCC cells. CONCLUSION: The present findings suggest that chemotherapeutic agents can induce an adjuvant effect leading to the extracellular release of DAMPs. Of the agents tested here, DDP, CBP, NDP, OXA and DOC had the ability to act as inducers of ICD.


Asunto(s)
Antineoplásicos , Calreticulina , Carcinoma de Células Escamosas , Cisplatino , Proteína HMGB1 , Muerte Celular Inmunogénica , Ratones Endogámicos C57BL , Compuestos Organoplatinos , Paclitaxel , Animales , Muerte Celular Inmunogénica/efectos de los fármacos , Humanos , Línea Celular Tumoral , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Proteína HMGB1/metabolismo , Calreticulina/metabolismo , Cisplatino/farmacología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Compuestos Organoplatinos/farmacología , Oxaliplatino/farmacología , Ratones , Carboplatino/farmacología , Docetaxel/farmacología , Docetaxel/uso terapéutico , Femenino , Adenosina Trifosfato/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Anexina A1/metabolismo
4.
Int Immunopharmacol ; 131: 111854, 2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38479155

RESUMEN

Annexin A1 (ANXA1) is widely expressed in a variety of body tissues and cells and is also involved in tumor development through multiple pathways. The invasion, metastasis, and immune escape of tumor cells depend on the interaction between tumor cells and their surrounding environment. Research shows that ANXA1 can act on a variety of cells in the tumor microenvironment (TME), and subsequently affect the proliferation, invasion and metastasis of tumors. This article describes the role of ANXA1 in the various components of the tumor microenvironment and its mechanism of action, as well as the existing clinical treatment measures related to ANXA1. These findings provide insight for the further design of strategies targeting ANXA1 for the diagnosis and treatment of malignant tumors.


Asunto(s)
Anexina A1 , Microambiente Tumoral , Anexina A1/genética , Anexina A1/metabolismo , Línea Celular Tumoral , Humanos , Animales
5.
Int J Oncol ; 64(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38516766

RESUMEN

Although annexin A1 (ANXA1), a 37 kDa phospholipid­binding anti­inflammatory protein expressed in various tissues and cell types, has been investigated extensively for its regulatory role in cancer biology, studies have mainly focused on its intracellular role. However, cancer cells and stromal cells expressing ANXA1 have the ability to transmit signals within the tumor microenvironment (TME) through autocrine, juxtacrine, or paracrine signaling. This bidirectional crosstalk between cancer cells and their environment is also crucial for cancer progression, contributing to uncontrolled tumor proliferation, invasion, metastasis and resistance to therapy. The present review explored the important role of ANXA1 in regulating the cell­specific crosstalk between various compartments of the TME and analyzed the guiding significance of the crosstalk effects in promotion or suppressing cancer progression in the development of cancer treatments. The literature shows that ANXA1 is critical for the regulation of the TME, indicating that ANXA1 signaling between cancer cells and the TME is a potential therapeutic target for the development of novel therapeutic approaches for impeding cancer development.


Asunto(s)
Anexina A1 , Microambiente Tumoral , Humanos , Anexina A1/genética , Anexina A1/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Transducción de Señal , Microambiente Tumoral/genética
6.
Neuroreport ; 35(7): 466-475, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38526918

RESUMEN

Diffuse axonal injury (DAI) is a critical pathological facet of traumatic brain injury (TBI). Oxidative stress plays a significant role in the progress of DAI. Annexin A1 (AnxA1) has been demonstrated to benefit from recovery of neurofunctional outcomes after TBI. However, whether AnxA1 exhibits neuronal protective function by modulating oxidative stress in DAI remains unknown. Expression of AnxA1 was evaluated via real-time PCR and western blotting in rat brainstem after DAI. The neurological effect of AnxA1 following DAI through quantification of modified neurologic severity score (mNSS) was compared between wild-type and AnxA1-knockout rats. Brain edema and neuronal apoptosis, as well as expression of oxidative factors and inflammatory cytokines, were analyzed between wild-type and AnxA1 deficiency rats after DAI. Furthermore, mNSS, oxidative and inflammatory cytokines were assayed after timely administration of recombinant AnxA1 for DAI rats. In the brainstem of DAI, the expression of AnxA1 remarkably increased. Ablation of AnxA1 increased the mNSS score and brain water content of rats after DAI. Neuron apoptosis in the brainstem after DAI was exaggerated by AnxA1 deficiency. In addition, AnxA1 deficiency significantly upregulated the level of oxidative and inflammatory factors in the brainstem of DAI rats. Moreover, mNSS decreased by AnxA1 treatment in rats following DAI. Expression of oxidative and inflammatory molecules in rat brainstem subjected to DAI inhibited by AnxA1 administration. AnxA1 exhibited neuronal protective function in the progression of DAI mainly dependent on suppressing oxidative stress and inflammation.


Asunto(s)
Anexina A1 , Lesiones Traumáticas del Encéfalo , Lesión Axonal Difusa , Animales , Ratas , Anexina A1/genética , Anexina A1/metabolismo , Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Citocinas/metabolismo , Lesión Axonal Difusa/patología , Inflamación/metabolismo
7.
FASEB J ; 38(3): e23457, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38318648

RESUMEN

Aging is associated with chronic, low-level inflammation which may contribute to cardiovascular pathologies such as hypertension and atherosclerosis. This chronic inflammation may be opposed by endogenous mechanisms to limit inflammation, for example, by the actions of annexin A1 (ANXA1), an endogenous glucocorticoid-regulated protein that has anti-inflammatory and pro-resolving activity. We hypothesized the pro-resolving mediator ANXA1 protects against age-induced changes in blood pressure (BP), cardiovascular structure and function, and cardiac senescence. BP was measured monthly in conscious mature (4-month) and middle-aged (12-month) ANXA1-deficient (ANXA1-/- ) and wild-type C57BL/6 mice. Body composition was measured using EchoMRI, and both cardiac and vascular function using ultrasound imaging. Cardiac hypertrophy, fibrosis and senescence, vascular fibrosis, elastin, and calcification were assessed histologically. Gene expression relevant to structural remodeling, inflammation, and cardiomyocyte senescence were also quantified. In C57BL/6 mice, progression from 4 to 12 months of age did not affect the majority of cardiovascular parameters measured, with the exception of mild cardiac hypertrophy, vascular calcium, and collagen deposition. Interestingly, ANXA1-/- mice exhibited higher BP, regardless of age. Additionally, age progression had a marked impact in ANXA1-/- mice, with markedly augmented vascular remodeling, impaired vascular distensibility, and body composition. Consistent with vascular dysfunction, cardiac dysfunction, and hypertrophy were also evident, together with markers of senescence and inflammation. These findings suggest that endogenous ANXA1 plays a critical role in regulating BP, cardiovascular function, and remodeling and delays cardiac senescence. Our findings support the development of novel ANXA1-based therapies to prevent age-related cardiovascular pathologies.


Asunto(s)
Anexina A1 , Presión Sanguínea , Remodelación Vascular , Animales , Ratones , Anexina A1/genética , Anexina A1/metabolismo , Cardiomegalia , Fibrosis , Inflamación/patología , Ratones Endogámicos C57BL , Ratones Noqueados
8.
Biomed Pharmacother ; 172: 116254, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38340398

RESUMEN

Leishmaniases, a group of diseases caused by the species of the protozoan parasite Leishmania, remains a significant public health concern worldwide. Host immune responses play a crucial role in the outcome of Leishmania infections, and several mediators that regulate inflammatory responses are potential targets for therapeutic approaches. Annexin A1 (AnxA1), an endogenous protein endowed with anti-inflammatory and pro-resolving properties, has emerged as a potential player. We have shown that during L. braziliensis infection, deficiency of AnxA1 exacerbates inflammatory responses but does not affect parasite burden. Here, we have investigated the role of AnxA1 in L. amazonensis infection, given the non-healing and progressive lesions characteristic of this infectious model. Infection of AnxA1 KO BALB/c mice resulted in increased lesion size and tissue damage associated with higher parasite burdens and enhanced inflammatory response. Notably, therapeutic application of the AnxA1 peptidomimetic Ac2-26 improves control of parasite replication and increases IL-10 production in vivo and in vitro, in both WT and AnxA1 KO mice. Conversely, administration of WRW4, an inhibitor of FPR2/3, resulted in larger lesions and decreased production of IL-10, suggesting that the effects of AnxA1 during L. amazonensis infection are associated with the engagement of these receptors. Our study illuminates the role of AnxA1 in L. amazonensis infection, demonstrating its impact on the susceptibility phenotype of BALB/c mice. Furthermore, our results indicate that targeting the AnxA1 pathway by using the Ac2-26 peptide could represent a promising alternative for new treatments for leishmaniasis.


Asunto(s)
Anexina A1 , Leishmania , Leishmaniasis , Péptidos , Animales , Ratones , Anexina A1/administración & dosificación , Anexina A1/metabolismo , Inmunidad , Interleucina-10/metabolismo , Leishmaniasis/tratamiento farmacológico , Ratones Endogámicos BALB C , Péptidos/administración & dosificación
9.
Oncogene ; 43(8): 608-614, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38200229

RESUMEN

In this study we conducted the first investigation to assess the efficacy of a novel therapeutic antibody developed to target annexin-A1 (ANXA1). ANXA1 is an immunomodulatory protein which has been shown to be overexpressed in, and promote the development and progression of, several cancer types. In particular, high ANXA1 expression levels correlate with poorer overall survival in pancreatic and triple-negative breast cancers, two cancers with considerable unmet clinical need. MDX-124 is a humanised IgG1 monoclonal antibody which specifically binds to ANXA1 disrupting its interaction with formyl peptide receptors 1 and 2 (FPR1/2). Here we show that MDX-124 significantly reduced proliferation (p < 0.013) in a dose-dependent manner across a panel of human cancer cell lines expressing ANXA1. The anti-proliferative effect of MDX-124 is instigated by arresting cell cycle progression with cancer cells accumulating in the G1 phase of the cell cycle. Furthermore, MDX-124 significantly inhibited tumour growth in both the 4T1-luc triple-negative breast and Pan02 pancreatic cancer syngeneic mouse models (p < 0.0001). These findings suggest ANXA1-targeted therapy is a viable and innovative approach to treat tumours which overexpress ANXA1.


Asunto(s)
Anexina A1 , Neoplasias , Animales , Humanos , Ratones , Anexina A1/antagonistas & inhibidores , Anexina A1/metabolismo , Línea Celular
10.
Cell Mol Immunol ; 21(1): 47-59, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38049523

RESUMEN

A highly immunosuppressive tumor microenvironment (TME) and the presence of the blood‒brain barrier are the two major obstacles to eliciting an effective immune response in patients with high-grade glioma (HGG). Here, we tried to enhance the local innate immune response in relapsed HGG by intracranially injecting poly(I:C) to establish a robust antitumor immune response in this registered clinical trial (NCT03392545). During the follow-up, 12/27 (44.4%) patients who achieved tumor control concomitant with survival benefit were regarded as responders in our study. We found that the T-cell receptor (TCR) repertoire in the TME was reshaped after poly(I:C) treatment. Based on the RNA-seq analysis of tumor samples, the expression of annexin A1 (ANXA1) was significantly upregulated in the tumor cells of nonresponders, which was further validated at the protein level. In vitro and in vivo experiments showed that ANXA1 could induce the production of M2-like macrophages and microglia via its surface receptor formyl peptide receptor 1 (FPR1) to establish a Treg cell-driven immunosuppressive TME and suppress the antitumor immune response facilitated by poly(I:C). The ANXA1/FPR1 signaling axis can inhibit the innate immune response of glioma patients by promoting an anti-inflammatory and Treg-driven TME. Moreover, ANXA1 could serve as a reliable predictor of response to poly(I:C), with a notable predictive accuracy rate of 92.3%. In light of these notable findings, this study unveils a new perspective of immunotherapy for gliomas.


Asunto(s)
Anexina A1 , Glioma , Humanos , Anexina A1/metabolismo , Antiinflamatorios , Inmunidad , Receptor Toll-Like 3/metabolismo , Microambiente Tumoral
11.
Cell Calcium ; 117: 102823, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37976974

RESUMEN

There have been in the last three decades repeated publications indicating that the inositol 1,4,5-trisphosphate receptor (IP3R) is regulated not only by cytosolic Ca2+ but also by intraluminal Ca2+. Although most studies indicated that a decreasing intraluminal Ca2+ level led to an inhibition of the IP3R, a number of publications reported exactly the opposite effect, i.e. an inhibition of the IP3R by high intraluminal Ca2+ levels. Although intraluminal Ca2+-binding sites on the IP3Rs were reported, a regulatory role for them was not demonstrated. It is also well known that the IP3R is regulated by a vast array of associated proteins, but only relatively recently proteins were identified that can be linked to the regulation of the IP3R by intraluminal Ca2+. The first to be reported was annexin A1 that is proposed to associate with the second intraluminal loop of the IP3R at high intraluminal Ca2+ levels and to inhibit the IP3R. More recently, ERdj5/PDIA19 reductase was described to reduce an intraluminal disulfide bridge of IP3R1 only at low intraluminal Ca2+ levels and thereby to inhibit the IP3R. Annexin A1 and ERdj5/PDIA19 can therefore explain most of the experimental results on the regulation of the IP3R by intraluminal Ca2+. Further studies are needed to provide a fuller understanding of the regulation of the IP3R from the intraluminal side. These findings underscore the importance of the state of the endoplasmic reticulum in the control of IP3R activity.


Asunto(s)
Anexina A1 , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Anexina A1/metabolismo , Señalización del Calcio , Sitios de Unión , Oxidación-Reducción , Calcio/metabolismo
12.
J Biomol Struct Dyn ; 42(6): 2825-2833, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37194290

RESUMEN

Annexin A1 (A1) has been shown to form a tetrameric complex (A1t) with S100A11 which is implicated in calcium homeostasis and EGFR pathways. In this work, a full-length model of the A1t was generated for the first time. Multiple molecular dynamics simulations were performed on the complete A1t model for several hundred nanoseconds each to assess the structure and dynamics of A1t. These simulations yielded three structures for the A1 N-terminus (ND) which were identified via principal component analysis. The orientations and interactions of the first 11 A1-ND residues for all three structures were conserved, and their binding modes were strikingly similar to those of the Annexin A2 N-terminus in the Annexin A2-p11 tetramer. In this study, we provided detailed atomistic information for the A1t. Strong interactions were identified within the A1t between the A1-ND and both S100A11 monomers. Residues M3, V4, S5, E6, L8, K9, W12, E15, and E18 of A1 were the strongest interactions between A1 and the S100A11 dimer. The different conformations of the A1t were attributed to the interaction between W12 of the A1-ND with M63 of S100A11 which caused a kink in the A1-ND. Cross-correlation analysis revealed strong correlated motion throughout the A1t. Strong positive correlation was observed between the ND and S100A11 in all simulations regardless of conformation. This work suggests that the stable binding of the first 11 residues of A1-ND to S100A11 is potentially a theme for Annexin-S100 complexes and that the flexibility of the A1-ND allows for multiple conformations of the A1t.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Anexina A1 , Anexina A2 , Sulfonamidas , Anexina A1/química , Anexina A1/metabolismo , Anexina A2/química , Anexina A2/metabolismo , Simulación de Dinámica Molecular , Proteínas S100/química , Proteínas S100/metabolismo
13.
Nephrology (Carlton) ; 29(2): 76-85, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37927194

RESUMEN

BACKGROUND: Annexin A1 is a membrane-associated calcium-binding protein that participates in the progression of many diseases by facilitating vesicle aggregation. It has been documented that reducing vesicle formation alleviates podocyte injury and albuminuria in idiopathic membranous nephropathy (IMN). However, the role of Annexin A1 (ANXA1) in IMN is unknown. METHODS: Electron microscopy was used to observe the numbers of vesicles in podocytes. The expression of ANXA1 in IMN was investigated by bioinformatics analysis. We validated the hub genes with the Nephroseq V5 online tool and microarray data from the GEO. Immunohistochemical staining and qPCR were performed to measure gene and protein expression. RESULTS: The numbers of vesicles in IMN podocytes were significantly increased. Bioinformatics analysis showed that ANXA1, one of the differentially expressed genes, was upregulated in glomeruli from IMN patients. In the validation database and dataset, we confirmed that ANXA1 expression was upregulated in the glomeruli of IMN patients. We revealed that the increased expression of ANXA1 was negatively correlated with the glomerular filtration rate (GFR) and proteinuria. Moreover, ANXA1 was enriched in the biological process of vesicle fusion, in which the expression of SNAREs and the SNARE complex was increased. Finally, the expression of ANXA1 and genes related to SNAREs and the SNARE complex was upregulated in glomeruli from IMN patients according to immunohistochemical staining and qPCR. CONCLUSION: We conclude that ANXA1 may mediate endocytic vesicle fusion and transport by promoting SNARE assembly, contributing to the morphological changes in podocytes and massive proteinuria in IMN.


Asunto(s)
Anexina A1 , Glomerulonefritis Membranosa , Podocitos , Humanos , Anexina A1/genética , Anexina A1/metabolismo , Glomerulonefritis Membranosa/genética , Glomerulonefritis Membranosa/metabolismo , Podocitos/metabolismo , Proteinuria , Proteínas SNARE/metabolismo , Vesículas Transportadoras/metabolismo
14.
J Physiol Pharmacol ; 74(5)2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38085519

RESUMEN

This study aimed to observe the differential expression of Annexin-A1 in esophageal squamous cell carcinoma (ESCC) and explored the effect of small interfering ribonucleic acid (RNAi)-Annexin-A1 on the biological behavior of CE81T-0 cells. An immunohistochemical approach was used to detect the expression of Annexin-A1 in 86 pairs of ESCC samples. Quantitative reverse transcription polymerase chain reaction was used to detect the expression of Annexin-A1 in CE81T-0 and CE81T-4 cells, and the expression of Annexin-A1 in CE81T-0 cells was knocked out by RNAi. A methyl-thiazolyl-tetrazolium assay was used to observe the effect of Annexin-A1 on cell proliferation, and flow cytometry was conducted to analyze its effect on cell cycles and apoptosis. A scratch assay and a Transwell chamber were used to detect changes in cell migration and invasion. From the results, compared with the Annexin-A1 expression rate of 59.3% in para-carcinoma tissues, the expression of Annexin-A1 in cancer was reduced to only 32.6% in ESCC cells. Annexin-A1 was strongly expressed in highly differentiated ESCC cells without lymphatic metastasis and highly expressed in the CE81T-0 cell group with low metastasis. Annexin-A1 gene silencing promoted cell proliferation and inhibited apoptosis, blocked cells in the S-phase, and increased cell migration, leading to an increase in the number of invaded cells. Above all, Annexin-A1 could reflect the differentiation degree and lymph node metastasis of ESCC cells to some extent and was involved in the invasion, metastasis, proliferation, and other biological behaviors of ESCC cells, indicating an experimental basis for Annexin-A1 as a molecular marker in the early diagnosis of ESCC and the prediction of cell metastasis, invasion, and differentiation degree.


Asunto(s)
Anexina A1 , Carcinoma de Células Escamosas , Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Humanos , Anexina A1/genética , Anexina A1/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Regulación Neoplásica de la Expresión Génica , Metástasis Linfática , Invasividad Neoplásica/genética
15.
Theranostics ; 13(15): 5561-5583, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37908731

RESUMEN

Rationale: Recent studies indicate that microglial activation and the resulting inflammatory response could be potential targets of adjuvant therapy for ischemic stroke. Many studies have emphasized a well-established function of Annexin-A1 (ANXA1) in the immune system, including the regulation of microglial activation. Nevertheless, few therapeutic interventions targeting ANXA1 in microglia for ischemic stroke have been conducted. In the present study, Tat-NTS, a small peptide developed to prevent ANXA1 from entering the nucleus, was utilized. We discovered the underlying mechanism that Tat-NTS peptide targets microglial ANXA1 to protect against ischemic brain injury. Methods: Preclinical studies of ischemic stroke were performed using an oxygen-glucose deprivation and reperfusion (OGD/R) cell model in vitro and the middle cerebral artery occlusion (MCAO) animal model of ischemic stroke in vivo. Confocal imaging and 3D reconstruction analyses for detecting the protein expression and subcellular localization of microglia in vivo. Co-immunoprecipitation (Co-IP), immunoblotting, ELISA, quantitative real-time PCR (qRT-PCR), Luciferase reporter assay for determining the precise molecular mechanism. Measurement on the cytotoxicity of Tat-NTS peptide for microglia was assessed by CCK-8 and LDH assay. TUNEL staining was used to detect the microglia conditioned medium-mediated neuronal apoptosis. Adeno-associated viruses (AAVs) were injected into the cerebral cortex, striatum and hippocampal CA1 region of adult male Cx3cr1-Cre mice, to further verify the neurofunctional outcome and mechanism of Tat-NTS peptide by TTC staining, the modified Neurological Severity Score (mNSS) test, the open field test (OFT), the novel object recognition task (NORT), the Morris water maze (MWM) test, the long-term potentiation (LTP) and the Transmission electron microscopy (TEM). Results: It was observed that administration of Tat-NTS led to a shift of subcellular localization of ANXA1 in microglia from the nucleus to the cytoplasm in response to ischemic injury. Notably, this shift was accompanied by an increase in ANXA1 SUMOylation in microglia and a transformation of microglia towards an anti-inflammatory phenotype. We confirmed that Tat-NTS-induced ANXA1 SUMOylation in microglia mediated IKKα degradation via NBR1-dependent selective autophagy, then blocking the activation of the NF-κB pathway. As a result, the expression and release of the pro-inflammatory factors IL-1ß and TNF-α were reduced in both in vitro and in vivo experiments. Furthermore, we found that Tat-NTS peptide's protective effect on microglia relieved ischemic neuron apoptosis. Finally, we demonstrated that Tat-NTS peptide administration, through induction of ANXA1 SUMOylation in microglia, reduced infarct volume, improved neurological function and facilitated behavioral recovery in MCAO mice. Conclusions: Our study provides evidence for a novel mechanism of Tat-NTS peptide in regulating microglial ANXA1 function and its substantial neuroprotective effect on neurons with ischemic injuries. These findings suggest that Tat-NTS peptides have a high potential for clinical application and may be a promising therapeutic candidate for treating cerebral ischemia.


Asunto(s)
Anexina A1 , Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Daño por Reperfusión , Ratones , Animales , Masculino , Microglía/metabolismo , Anexina A1/metabolismo , Sumoilación , Isquemia Encefálica/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Péptidos/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Daño por Reperfusión/metabolismo , Neuronas/metabolismo
16.
J Biosci Bioeng ; 136(6): 423-429, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37805288

RESUMEN

Several fusion tags have been developed for non-chromatographic fusion protein purification. Previously, we identified that human annexin A1 as a novel N-terminal purification tag was used for purifying the fusion proteins produced in Escherichia coli through precipitation in 10 mM Ca2+ buffer, and redissolution of the precipitate in 15 mM EDTA buffer. In this work, we selected four metal-dependent enzymes including E. coli 5-aminolevulinate dehydratase, yeast 3-hydroxyanthranilate 3,4-dioxygenase, maize serine racemase and copper amine oxidase for investigating the annexin A1 tag applicability. Fusion of the His6-tag or the enzyme changed the behavior of precipitation-redissolution. The relatively high recovery yields of three tagged enzymes with the improved purities were obtained through two rounds of purification, whereas low recovery yield of the annexin A1 tagged maize amine oxidase was prepared. The added EDTA displayed different abilities to redissolve the fusion proteins precipitates in two precipitation-redissolution cycles. It inactivated three enzymes and obviously inhibited the activity of the fused maize serine racemase. Based on current findings, we believe that four enzymes could be applied for evaluating applicability of the proteins or peptides as affinity tags for chromatographic purification in a calcium dependent manner.


Asunto(s)
Anexina A1 , Humanos , Anexina A1/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Ácido Edético/metabolismo , Cromatografía de Afinidad/métodos , Péptidos/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
17.
Biomed Pharmacother ; 165: 115230, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37531784

RESUMEN

Diabetes mellitus (DM) is characterized by metabolic alterations that involve defects in the secretion and/or action of insulin, being responsible for several complications, such as impaired healing. Studies from our research group have shown that annexin A1 protein (AnxA1) is involved in the regulation of inflammation and cell proliferation. In light of these findings, we have developed a new technology and evaluated its effect on a wound healing in vivo model using type 1 diabetes (T1DM)-induced mice. We formulated a hydrogel containing AnxA12-26 using defined parameters such as organoleptic characteristics, pH, UV-vis spectroscopy and cytotoxicity assay. UV-vis spectroscopy confirmed the presence of the associated AnxA12-26 peptide in the three-dimensional hydrogel matrix, while the in vitro cytotoxicity assay showed excellent biocompatibility. Mice showed increased blood glucose levels, confirming the efficacy of streptozotocin (STZ) to induce T1DM. Treatment with AnxA12-26 hydrogel showed to improve diabetic wound healing, defined as complete re-epithelialization and tissue remodeling, with reduction of inflammatory infiltrate in diabetic animals. We envisage that the AnxA12-26 hydrogel, with its innovative composition and formulation be efficient on improving diabetic healing and contributing on the expansion of the therapeutic arsenal to treat diabetic wounds, at a viable cost.


Asunto(s)
Anexina A1 , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Enfermedades de la Piel , Ratones , Animales , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Hidrogeles/farmacología , Hidrogeles/química , Anexina A1/farmacología , Anexina A1/metabolismo , Diabetes Mellitus Experimental/metabolismo , Cicatrización de Heridas
18.
Am J Pathol ; 193(11): 1817-1832, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37423551

RESUMEN

Annexin A1 (AnxA1) is the primary mediator of the anti-inflammatory actions of glucocorticoids. AnxA1 functions as a pro-resolving mediator in cultured rat conjunctival goblet cells to ensure tissue homeostasis through stimulation of intracellular [Ca2+] ([Ca2+]i) and mucin secretion. AnxA1 has several N-terminal peptides with anti-inflammatory properties of their own, including Ac2-26, Ac2-12, and Ac9-25. The increase in [Ca2+]i caused by AnxA1 and its N-terminal peptides in goblet cells was measured to determine the formyl peptide receptors used by the compounds and the action of the peptides on histamine stimulation. Changes in [Ca2+]i were determined by using a fluorescent Ca2+ indicator. AnxA1 and its peptides each activated formyl peptide receptors in goblet cells. AnxA1 and Ac2-26 at 10-12 mol/L and Ac2-12 at 10-9 mol/L inhibited the histamine-stimulated increase in [Ca2+]i, as did resolvin D1 and lipoxin A4 at 10-12 mol/L, whereas Ac9-25 did not. AnxA1 and Ac2-26 counter-regulated the H1 receptor through the p42/p44 mitogen-activated protein kinase/extracellular regulated kinase 1/2, ß-adrenergic receptor kinase, and protein kinase C pathways, whereas Ac2-12 counter-regulated only through ß-adrenergic receptor kinase. In conclusion, current data show that the N-terminal peptides Ac2-26 and Ac2-12, but not Ac9-25, share multiple functions with the full-length AnxA1 in goblet cells, including inhibition of histamine-stimulated increase in [Ca2+]i and counter-regulation of the H1 receptor. These actions suggest a potential pharmaceutical application of the AnxA1 N-terminal peptides Ac2-26 and Ac2-12 in homeostasis and ocular inflammatory diseases.


Asunto(s)
Anexina A1 , Ratas , Animales , Anexina A1/farmacología , Anexina A1/química , Anexina A1/metabolismo , Células Caliciformes/metabolismo , Receptores de Formil Péptido/metabolismo , Histamina/farmacología , Péptidos/farmacología , Antiinflamatorios/farmacología , Quinasas de Receptores Adrenérgicos beta/metabolismo
19.
Int J Mol Sci ; 24(12)2023 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-37373303

RESUMEN

The functions of annexin A1 (ANXA1), which is expressed on membranes and in cytoplasmic granules, have been fully described. Nonetheless, the role of this protein in protecting against DNA damage in the nucleus is still emerging and requires further investigation. Here, we investigated the involvement of ANXA1 in the DNA damage response in placental cells. Placenta was collected from ANXA1 knockout mice (AnxA1-/-) and pregnant women with gestational diabetes mellitus (GDM). The placental morphology and ANXA1 expression, which are related to the modulation of cellular response markers in the presence of DNA damage, were analyzed. The total area of AnxA1-/- placenta was smaller due to a reduced labyrinth zone, enhanced DNA damage, and impaired base excision repair (BER) enzymes, which resulted in the induction of apoptosis in the labyrinthine and junctional layers. The placentas of pregnant women with GDM showed reduced expression of AnxA1 in the villous compartment, increased DNA damage, apoptosis, and a reduction of enzymes involved in the BER pathway. Our translational data provide valuable insights into the possible involvement of ANXA1 in the response of placental cells to oxidative DNA damage and represent an advancement in investigations into the mechanisms involved in placental biology.


Asunto(s)
Anexina A1 , Diabetes Gestacional , Ratones , Animales , Embarazo , Humanos , Femenino , Placenta/metabolismo , Diabetes Gestacional/genética , Diabetes Gestacional/metabolismo , Anexina A1/metabolismo , Procesamiento Proteico-Postraduccional , Daño del ADN
20.
Cells ; 12(8)2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37190040

RESUMEN

Dysregulated inflammatory responses are often correlated with disease severity during viral infections. Annexin A1 (AnxA1) is an endogenous pro-resolving protein that timely regulates inflammation by activating signaling pathways that culminate with the termination of response, clearance of pathogen and restoration of tissue homeostasis. Harnessing the pro-resolution actions of AnxA1 holds promise as a therapeutic strategy to control the severity of the clinical presentation of viral infections. In contrast, AnxA1 signaling might also be hijacked by viruses to promote pathogen survival and replication. Therefore, the role of AnxA1 during viral infections is complex and dynamic. In this review, we provide an in-depth view of the role of AnxA1 during viral infections, from pre-clinical to clinical studies. In addition, this review discusses the therapeutic potential for AnxA1 and AnxA1 mimetics in treating viral infections.


Asunto(s)
Anexina A1 , Virosis , Humanos , Anexina A1/metabolismo , Inflamación/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA