Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 629
1.
CNS Neurosci Ther ; 30(5): e14749, 2024 05.
Article En | MEDLINE | ID: mdl-38739004

AIMS: A bone-invasive pituitary adenoma exhibits aggressive behavior, leading to a worse prognosis. We have found that TNF-α promotes bone invasion by facilitating the differentiation of osteoclasts, however, before bone-invasive pituitary adenoma invades bone tissue, it needs to penetrate the dura mater, and this mechanism is not yet clear. METHODS: We performed transcriptome microarrays on specimens of bone-invasive pituitary adenomas (BIPAs) and noninvasive pituitary adenomas (NIPAs) and conducted differential expressed gene analysis and enrichment analysis. We altered the expression of TNF-α through plasmids, then validated the effects of TNF-α on GH3 cells and verified the efficacy of the TNF-α inhibitor SPD304. Finally, the effects of TNF-α were validated in in vivo experiments. RESULTS: Pathway act work showed that the MAPK pathway was significantly implicated in the pathway network. The expression of TNF-α, MMP9, and p-p38 is higher in BIPAs than in NIPAs. Overexpression of TNF-α elevated the expression of MAPK pathway proteins and MMP9 in GH3 cells, as well as promoted proliferation, migration, and invasion of GH3 cells. Flow cytometry indicated that TNF-α overexpression increased the G2 phase ratio in GH3 cells and inhibited apoptosis. The expression of MMP9 was reduced after blocking the P38 MAPK pathway; overexpression of MMP9 promoted invasion of GH3 cells. In vivo experiments confirm that the TNF-α overexpression group has larger tumor volumes. SPD304 was able to suppress the effects caused by TNF-α overexpression. CONCLUSION: Bone-invasive pituitary adenoma secretes higher levels of TNF-α, which then acts on itself in an autocrine manner, activating the MAPK pathway and promoting the expression of MMP9, thereby accelerating the membrane invasion process. SPD304 significantly inhibits the effect of TNF-α and may be applied in the clinical treatment of bone-invasive pituitary adenoma.


Adenoma , MAP Kinase Signaling System , Matrix Metalloproteinase 9 , Neoplasm Invasiveness , Pituitary Neoplasms , Tumor Necrosis Factor-alpha , Tumor Necrosis Factor-alpha/metabolism , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Humans , Adenoma/pathology , Adenoma/metabolism , Animals , Matrix Metalloproteinase 9/metabolism , MAP Kinase Signaling System/physiology , MAP Kinase Signaling System/drug effects , Male , Cell Line, Tumor , Female , Mice , Mice, Nude , Autocrine Communication/physiology , Autocrine Communication/drug effects , Middle Aged , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Adult , Rats , Cell Movement/drug effects , Cell Movement/physiology , Signal Transduction/physiology , Signal Transduction/drug effects
2.
Nat Commun ; 13(1): 4130, 2022 07 15.
Article En | MEDLINE | ID: mdl-35840551

Fate determination and maintenance of fetal testes in most mammals occur cell autonomously as a result of the action of key transcription factors in Sertoli cells. However, the cases of freemartin, where an XX twin develops testis structures under the influence of an XY twin, imply that hormonal factor(s) from the XY embryo contribute to sex reversal of the XX twin. Here we show that in mouse XY embryos, Sertoli cell-derived anti-Mullerian hormone (AMH) and activin B together maintain Sertoli cell identity. Sertoli cells in the gonadal poles of XY embryos lacking both AMH and activin B transdifferentiate into their female counterpart granulosa cells, leading to ovotestis formation. The ovotestes remain to adulthood and produce both sperm and oocytes, although there are few of the former and the latter fail to mature. Finally, the ability of XY mice to masculinize ovaries is lost in the absence of these two factors. These results provide insight into fate maintenance of fetal testes through the action of putative freemartin factors.


Activins , Anti-Mullerian Hormone , Cell Differentiation , Testis , Activins/metabolism , Activins/pharmacology , Animals , Anti-Mullerian Hormone/metabolism , Anti-Mullerian Hormone/pharmacology , Autocrine Communication/drug effects , Autocrine Communication/physiology , Cell Differentiation/physiology , Female , Male , Mammals , Mice , Paracrine Communication/physiology , Semen , Sertoli Cells , Testis/metabolism
3.
Int J Mol Sci ; 22(22)2021 Nov 18.
Article En | MEDLINE | ID: mdl-34830320

Emerging evidence suggests that breast cancer stem cells (BCSCs), and epithelial-mesenchymal transition (EMT) may be involved in resistance to doxorubicin. However, it is unlear whether the doxorubicin-induced EMT and expansion of BCSCs is related to cancer dormancy, or outgrowing cancer cells with maintaining resistance to doxorubicin, or whether the phenotypes can be transferred to other doxorubicin-sensitive cells. Here, we characterized the phenotype of doxorubicin-resistant TNBC cells while monitoring the EMT process and expansion of CSCs during the establishment of doxorubicin-resistant MDA-MB-231 human breast cancer cells (DRM cells). In addition, we assessed the potential signaling associated with the EMT process and expansion of CSCs in doxorubicin-resistance of DRM cells. DRM cells exhibited morphological changes from spindle-shaped MDA-MB-231 cells into round-shaped giant cells. They exhibited highly proliferative, EMT, adhesive, and invasive phenotypes. Molecularly, they showed up-regulation of Cyclin D1, mesenchymal markers (ß-catenin, and N-cadherin), MMP-2, MMP-9, ICAM-1 and down-regulation of E-cadherin. As the molecular mechanisms responsible for the resistance to doxorubicin, up-regulation of EGFR and its downstream signaling, were suggested. AKT and ERK1/2 expression were also increased in DRM cells with the advancement of resistance to doxorubicin. Furthermore, doxorubicin resistance of DRM cells can be transferred by autocrine signaling. In conclusion, DRM cells harbored EMT features with CSC properties possessing increased proliferation, invasion, migration, and adhesion ability. The doxorubicin resistance, and doxorubicin-induced EMT and CSC properties of DRM cells, can be transferred to parental cells through autocrine signaling. Lastly, this feature of DRM cells might be associated with the up-regulation of EGFR.


Antibiotics, Antineoplastic/pharmacology , Autocrine Communication/drug effects , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Neoplastic Stem Cells/drug effects , Antigens, CD/genetics , Antigens, CD/metabolism , Autocrine Communication/genetics , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , beta Catenin/genetics , beta Catenin/metabolism
4.
Cell Rep ; 36(7): 109559, 2021 08 17.
Article En | MEDLINE | ID: mdl-34407399

Acute myeloid leukemia (AML) is a rapidly progressing cancer, for which chemotherapy remains standard treatment and additional therapeutic targets are requisite. Here, we show that AML cells secrete the stem cell growth factor R-spondin 2 (RSPO2) to promote their self-renewal and prevent cell differentiation. Although RSPO2 is a well-known WNT agonist, we reveal that it maintains AML self-renewal WNT independently, by inhibiting BMP receptor signaling. Autocrine RSPO2 signaling is also required to prevent differentiation and to promote self-renewal in normal hematopoietic stem cells as well as primary AML cells. Comprehensive datamining reveals that RSPO2 expression is elevated in patients with AML of poor prognosis. Consistently, inhibiting RSPO2 prolongs survival in AML mouse xenograft models. Our study indicates that in AML, RSPO2 acts as an autocrine BMP antagonist to promote cancer cell renewal and may serve as a marker for poor prognosis.


Bone Morphogenetic Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Leukemia, Myeloid, Acute/metabolism , Signal Transduction , Animals , Autocrine Communication/drug effects , Cell Differentiation/drug effects , Cell Self Renewal/drug effects , Cytarabine/pharmacology , HEK293 Cells , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis , Risk Factors , Signal Transduction/drug effects , Survival Analysis , Xenograft Model Antitumor Assays
5.
Biomed Res Int ; 2021: 6695663, 2021.
Article En | MEDLINE | ID: mdl-33937411

BACKGROUND: When vascular endothelial cells are subjected to external stimuli, paracrine hormones and cytokines act on adjacent cells. The regulation of the biological behaviour of cells is closely related to the maintenance of organ function and the occurrence and development of disease. However, it is unclear whether vascular endothelial cells affect the biological behaviour of cells involved in wound repair through autocrine and paracrine mechanisms and ultimately play a role in wound healing. We aimed to verify the effect of the autocrine and paracrine functions of vascular endothelial cells on wound healing. MATERIALS AND METHODS: ELISA was used to detect platelet-derived growth factor, basic fibroblast growth factor, epidermal growth factor, and vascular endothelial growth factor in human umbilical vascular endothelial cell-conditioned medium (HUVEC-CM). Different concentrations of HUVEC-CM were used to treat different stem cells. CCK-8 and scratch assays were used to detect the proliferation and migration ability of each cell. A full-thickness dorsal skin defect model was established in mice, and skin wound healing was observed after the local injection of HUVEC-CM, endothelial cell medium (ECM), or normal saline. H&E staining and immunofluorescence were used to observe the gross morphology of the wound tissue, the epithelial cell migration distance, and the expression of CD3 and CD31. RESULTS: HUVEC-CM promotes the proliferation and migration of epidermal stem cells, skin fibroblasts, bone marrow mesenchymal stem cells, and HUVECs themselves. Furthermore, HUVEC-CM can promote angiogenesis in mouse skin wounds and granulation tissue formation and can accelerate wound surface epithelialization and collagen synthesis, thereby promoting wound healing. CONCLUSION: Our results clearly suggest that it is practicable and effective to promote wound healing with cytokines secreted by vascular endothelial cells in a mouse model.


Autocrine Communication , Human Umbilical Vein Endothelial Cells/metabolism , Paracrine Communication , Skin/pathology , Wound Healing , Antigens, CD/metabolism , Autocrine Communication/drug effects , Biomarkers/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Cytokines/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Paracrine Communication/drug effects , Wound Healing/drug effects
6.
Am J Physiol Endocrinol Metab ; 320(4): E822-E834, 2021 04 01.
Article En | MEDLINE | ID: mdl-33615874

Fibroblast growth factor-21 (FGF21) is a hormonal regulator of metabolism; it promotes glucose oxidation and the thermogenic capacity of adipose tissues. The levels of ß-klotho (KLB), the co-receptor required for FGF21 action, are decreased in brown (BAT) and white (WAT) adipose tissues during obesity, diabetes, and lipodystrophy. Reduced ß-klotho levels have been proposed to account for FGF21 resistance in these conditions. In this study, we explored whether downregulation of ß-klotho affects metabolic regulation and the thermogenic responsiveness of adipose tissues using mice with total (KLB-KO) or partial (KLB-heterozygotes) ablation of ß-klotho. We herein show that KLB gene dosage was inversely associated with adiposity in mice. Upon cold exposure, impaired browning of subcutaneous WAT and milder alterations in BAT were associated with reduced KLB gene dosage in mice. Cultured brown and beige adipocytes from mice with total or partial ablation of the KLB gene showed reduced thermogenic responsiveness to ß3-adrenergic activation by treatment with CL316,243, indicating that these effects were cell-autonomous. Deficiency in FGF21 mimicked the KLB-reduction-induced impairment of thermogenic responsiveness in brown and beige adipocytes. These results indicate that the levels of KLB in adipose tissues determine their thermogenic capacity to respond to cold and/or adrenergic stimuli. Moreover, an autocrine action of FGF21 in brown and beige adipocytes may account for the ability of the KLB level to influence thermogenic responsiveness.NEW & NOTEWORTHY Reduced levels of KLB (the obligatory FGF21 co-receptor), as occurring in obesity and type 2 diabetes, reduce the thermogenic responsiveness of adipose tissues in cold-exposed mice. Impaired response to ß3-adrenergic activation in brown and beige adipocytes with reduced KLB occurs in a cell-autonomous manner involving an autocrine action of FGF21.


Adipose Tissue/metabolism , Fibroblast Growth Factors/physiology , Membrane Proteins/physiology , Thermogenesis/genetics , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adiposity/genetics , Animals , Autocrine Communication/drug effects , Autocrine Communication/genetics , Cells, Cultured , Fibroblast Growth Factors/pharmacology , Gene Dosage/physiology , Klotho Proteins , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Thermogenesis/drug effects
7.
Nat Commun ; 12(1): 66, 2021 01 04.
Article En | MEDLINE | ID: mdl-33397952

IL11 is important for fibrosis in non-alcoholic steatohepatitis (NASH) but its role beyond the stroma in liver disease is unclear. Here, we investigate the role of IL11 in hepatocyte lipotoxicity. Hepatocytes highly express IL11RA and secrete IL11 in response to lipid loading. Autocrine IL11 activity causes hepatocyte death through NOX4-derived ROS, activation of ERK, JNK and caspase-3, impaired mitochondrial function and reduced fatty acid oxidation. Paracrine IL11 activity stimulates hepatic stellate cells and causes fibrosis. In mouse models of NASH, hepatocyte-specific deletion of Il11ra1 protects against liver steatosis, fibrosis and inflammation while reducing serum glucose, cholesterol and triglyceride levels and limiting obesity. In mice deleted for Il11ra1, restoration of IL11 cis-signaling in hepatocytes reconstitutes steatosis and inflammation but not fibrosis. We found no evidence for the existence of IL6 or IL11 trans-signaling in hepatocytes or NASH. These data show that IL11 modulates hepatocyte metabolism and suggests a mechanism for NAFLD to NASH transition.


Hepatocytes/metabolism , Interleukin-11/metabolism , Lipids/toxicity , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction , Adult , Animals , Autocrine Communication/drug effects , Cells, Cultured , Disease Models, Animal , Feeding Behavior , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Interleukin-11 Receptor alpha Subunit/metabolism , Interleukin-6/metabolism , Mice, Knockout , Models, Biological , Paracrine Communication/drug effects , Phenotype , Signal Transduction/drug effects
8.
Int J Mol Sci ; 22(2)2021 Jan 19.
Article En | MEDLINE | ID: mdl-33478130

Autocrine signaling is defined as the production and secretion of an extracellular mediator by a cell followed by the binding of that mediator to receptors on the same cell to initiate signaling. Autocrine stimulation often operates in autocrine loops, a type of interaction, in which a cell produces a mediator, for which it has receptors, that upon activation promotes expression of the same mediator, allowing the cell to repeatedly autostimulate itself (positive feedback) or balance its expression via regulation of a second factor that provides negative feedback. Autocrine signaling loops with positive or negative feedback are an important feature in cancer, where they enable context-dependent cell signaling in the regulation of growth, survival, and cell motility. A growth factor that is intimately involved in tumor development and progression and often produced by the cancer cells in an autocrine manner is transforming growth factor-ß (TGF-ß). This review surveys the many observations of autocrine TGF-ß signaling in tumor biology, including data from cell culture and animal models as well as from patients. We also provide the reader with a critical discussion on the various experimental approaches employed to identify and prove the involvement of autocrine TGF-ß in a given cellular response.


Autocrine Communication , Neoplasms/metabolism , Transforming Growth Factor beta/physiology , Animals , Autocrine Communication/drug effects , Autocrine Communication/physiology , Biomedical Research/methods , Biomedical Research/standards , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Neoplasms/pathology , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Research Design , Transforming Growth Factor beta/pharmacology
9.
Am J Physiol Endocrinol Metab ; 320(1): E78-E86, 2021 01 01.
Article En | MEDLINE | ID: mdl-33103455

Insulin secretion by ß-cells is largely controlled by circulating nutrients, hormones, and neurotransmitters. However, recent years have witnessed the multiplication of studies investigating whether local regulation also takes place within pancreatic islets, in which ß-cells cohabit with several other cell types. The cell composition and architectural organization of human islets differ from those of rodent islets and are particularly favorable to cellular interactions. An impressive number of hormonal (glucagon, glucagon-like peptide-1, somatostatin, etc.) and nonhormonal products (ATP, acetylcholine, γ-aminobutyric acid, dopamine, etc.) are released by islet cells and have been implicated in a local control of insulin secretion. This review analyzes reports directly testing paracrine and autocrine control of insulin secretion in isolated human islets. Many of these studies were designed on background information collected in rodent islets. However, the perspective of the review is not to highlight species similarities or specificities but to contrast established and speculative mechanisms in human islets. It will be shown that the current evidence is convincing only for a minority of candidates for a paracrine function whereas arguments supporting a physiological role of others do not stand up to scrutiny. Several pending questions await further investigation.


Autocrine Communication/physiology , Insulin Secretion/physiology , Islets of Langerhans/metabolism , Paracrine Communication/physiology , Autocrine Communication/drug effects , Hormones/pharmacology , Humans , Insulin Secretion/drug effects , Islets of Langerhans/drug effects , Paracrine Communication/drug effects
10.
J Exp Med ; 218(2)2021 02 01.
Article En | MEDLINE | ID: mdl-33125053

Macrophages help defend the host against Mycobacterium tuberculosis (Mtb), the major cause of tuberculosis (TB). Once phagocytized, Mtb resists killing by macrophages, replicates inside them, and leads to their death, releasing Mtb that can infect other cells. We found that the death of Mtb-infected mouse macrophages in vitro does not appear to proceed by a currently known pathway. Through genome-wide CRISPR-Cas9 screening, we identified a critical role for autocrine or paracrine signaling by macrophage-derived type I IFNs in the death of Mtb-infected macrophages in vitro, and blockade of type I IFN signaling augmented the effect of rifampin, a first-line TB drug, in Mtb-infected mice. Further definition of the pathway of type I IFN-mediated macrophage death may allow for host-directed therapy of TB that is more selective than systemic blockade of type I IFN signaling.


Cell Death/physiology , Interferon Type I/metabolism , Macrophages/metabolism , Signal Transduction/physiology , Tuberculosis/metabolism , Animals , Autocrine Communication/drug effects , Autocrine Communication/physiology , CRISPR-Cas Systems/drug effects , CRISPR-Cas Systems/physiology , Cell Death/drug effects , Cell Line , HEK293 Cells , Humans , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/drug effects , Paracrine Communication/drug effects , Paracrine Communication/physiology , RAW 264.7 Cells , Rifampin/pharmacology , Signal Transduction/drug effects , Tuberculosis/drug therapy , Tuberculosis/microbiology
11.
Cell Death Dis ; 11(12): 1035, 2020 12 05.
Article En | MEDLINE | ID: mdl-33279931

Medulloblastoma (MB) is a high-grade pediatric brain malignancy that originates from neuronal precursors located in the posterior cranial fossa. In this study, we evaluated the role of STAT3 and IL-6 in a tumor microenvironment mediated drug resistance in human MBs. We established that the Group 3 MB cell line, Med8A, is chemosensitive (hence Med8A-S), and this is correlated with a basal low phosphorylated state of STAT3, while treatment with IL-6 induced robust increases in pY705-STAT3. Via incremental selection with vincristine, we derived the stably chemoresistant variant, Med8A-R, that exhibited multi-drug resistance, enhanced IL-6 induced pY705-STAT3 levels, and increased IL6R expression. Consequently, abrogation of STAT3 or IL6R expression in Med8A-R led to restored chemosensitivity to vincristine, highlighting a prominent role for canonical IL-6/STAT3 signaling in acquired drug resistance. Furthermore, Med8A-S subjected to conditioning exposure with IL-6, termed Med8A-IL6+ cells, exhibited enhanced vincristine resistance, increased expression of pY705-STAT3 and IL6R, and increased secretion of IL-6. When cocultured with Med8A-IL6+ cells, Med8A-S cells exhibited increased pY705-STAT3 and increased IL-6 secretion, suggesting a cytokine feedback loop responsible for amplifying STAT3 activity. Similar IL-6 induced phenomena were also observed in the Group 3 MB cell lines, D283 and D341, including increased pY705-STAT3, drug resistance, IL-6 secretion and IL6R expression. Our study unveiled autocrine IL-6 as a promoter of STAT3 signaling in development of drug resistance, and suggests therapeutic benefits for targeting the IL-6/STAT3 signaling axis in Group 3 MBs.


Autocrine Communication , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Drug Resistance, Neoplasm , Interleukin-6/metabolism , Medulloblastoma/drug therapy , Medulloblastoma/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Autocrine Communication/drug effects , Autocrine Communication/genetics , Cell Line, Tumor , Cisplatin/pharmacology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , Niclosamide/pharmacology , Niclosamide/therapeutic use , Receptors, Interleukin-6/metabolism , Signal Transduction/drug effects , Vincristine/pharmacology , Vincristine/therapeutic use
12.
Cell Commun Signal ; 18(1): 185, 2020 11 23.
Article En | MEDLINE | ID: mdl-33228717

BACKGROUND: The matrix metalloproteinase-9 (MMP-9) is up-regulated by several proinflammatory mediators in the central nervous system (CNS) diseases. Increasing reports show that MMP-9 expression is an inflammatory biomarker of several CNS disorders, including the CNS inflammation and neurodegeneration. Bradykinin (BK) is a common proinflammatory mediator and elevated in several brain injury and inflammatory disorders. The raised BK may be detrimental effects on the CNS that may aggravate brain inflammation through MMP-9 up-regulation or cyclooxygenase-2 (COX-2)-derived prostaglandin E2 (PGE2) production in brain astrocytes. However, the relationship between BK-induced MMP-9 expression and COX-2-derived PGE2 release in brain astrocytes remains unclear. METHODS: Herein we used rat brain astrocytes (RBA) to investigate the role of the COX-2/PGE2 system in BK-induced MMP-9 expression. We used zymographic, RT-PCR, EIA, and Western blotting analyses to confirm that BK induces MMP-9 expression via a COX-2/PGE2-dependent pathway. RESULTS: Our results show activation of native COX-2 by BK led to PGE2 production and release. Subsequently, PGE2 induced MMP-9 expression via PGE2 receptor (EP)-mediated c-Src, Jak2, ERK1/2, and then activated signal transducer and activator of transcription 3 (STAT3) signaling pathway. Finally, up-regulation of MMP-9 by BK via the pathway may promote astrocytic migration. CONCLUSION: These results demonstrated that a novel autocrine pathway for BK-induced MMP-9 protein expression is mediated through activation of STAT3 by native COX-2/PGE2-mediated c-Src/Jak2/ERK cascades in brain astrocytes. Video Abstract.


Astrocytes/cytology , Astrocytes/enzymology , Autocrine Communication , Bradykinin/pharmacology , Cell Movement/drug effects , Dinoprostone/metabolism , Matrix Metalloproteinase 9/metabolism , STAT3 Transcription Factor/metabolism , Animals , Astrocytes/drug effects , Autocrine Communication/drug effects , Celecoxib/pharmacology , Cell Line , Janus Kinase 2/metabolism , MAP Kinase Signaling System/drug effects , Rats , Receptors, Prostaglandin E/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , src-Family Kinases/metabolism
13.
Basic Res Cardiol ; 115(6): 65, 2020 11 10.
Article En | MEDLINE | ID: mdl-33169229

Proprotein convertase subtilisin kexin type 9 (PCSK9) is in the focus of cardiovascular research due to its role in hepatic low density lipoprotein (LDL) clearance. However, extrahepatic expression of PCSK9 such as in cardiomyocytes and its regulation by oxidized LDL (oxLDL) put notion on extrahepatic effects of PCSK9 as well. This study was aimed to reveal the role of PCSK9 in oxLDL-dependent regulation of cardiomyocyte function. Adult rat and mouse ventricular cardiomyocytes and isolated perfused hearts were used. OxLDL was applied to increase PCSK9 expression in cardiomyocytes. Cell function was analyzed by load-free cell shortening as well as left ventricular developed pressure of isolated hearts. OxLDL decreased shortening in wild-type-derived mouse cardiomyocytes but not in those isolated from PCSK9 knockout mice. Overexpression of human PCSK9 in rat cardiomyocytes reduced shortening in the absence of oxLDL. Addition of recombinant PCSK9 mimicked these effects. In cardiomyocytes, oxLDL induced PCSK9 release into the supernatant. Inhibition of PCSK9 by Pep 2-8 or alirocumab attenuated the oxLDL-induced loss of cardiomyocyte shortening. Cardiomyocytes express surfeit locus protein 4 (SURF-4), a protein required for PCSK9 secretion in human embryonic kidney cells (HEK 293 T), and silencing of SURF-4 reduced the oxLDL effects on cardiomyocytes. In isolated perfused rat hearts PCSK9 inhibition by alirocumab improved the function. In addition, left ventricular function of isolated hearts from PCSK9 knockout mice was increased under basal conditions as well as at 10 min and 120 min of reperfusion following 45 min of ischemia. Collectively, the data show that cardiomyocytes express and release PCSK9 that acts in an autocrine way on cardiomyocytes and impairs their function.


Autocrine Communication , Myocytes, Cardiac/enzymology , Proprotein Convertase 9/metabolism , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Autocrine Communication/drug effects , Hep G2 Cells , Humans , Isolated Heart Preparation , Lipoproteins, LDL/pharmacology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocytes, Cardiac/drug effects , PCSK9 Inhibitors , Proprotein Convertase 9/genetics , Rats, Wistar , Serine Proteinase Inhibitors/pharmacology , Signal Transduction , Ventricular Function, Left , Ventricular Pressure
14.
Anticancer Res ; 40(11): 6075-6081, 2020 Nov.
Article En | MEDLINE | ID: mdl-33109545

BACKGROUND: One of the most prevalent causes of cancer fatalities is hepatocellular carcinoma (HCC), which has been linked to metabolic syndrome. Circulating levels of the saturated fatty acid palmitate are elevated in metabolic syndrome and lead to cellular stress. MATERIALS AND METHODS: Using enzyme-linked immunosorbent assay, flow cytometry, and migration assays, we characterized the response of rat hepatoma cells to palmitate treatment. RESULTS: We detected a 60% increase in secretion of C-X-C motif ligand 1 (CXCL1) which was dose-dependent and coincided with apoptosis. We measured expression of C-X-C motif chemokine receptor 2 (CXCR2) and observed a 4.5-fold increase on apoptotic hepatoma cells. Furthermore, we assayed migration of hepatoma cells and saw a 2-fold increase in the number of migrating cells towards CXCL1. CONCLUSION: These findings suggest that HCC cells secrete CXCL1 in response to metabolic syndrome signals and may promote the progression of cancer through apoptosis recovery or metastasis.


Autocrine Communication , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Chemokine CXCL1/metabolism , Disease Progression , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Animals , Apoptosis/drug effects , Autocrine Communication/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Glucose/pharmacology , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Mice , Palmitic Acid/toxicity , Rats , Receptors, Interleukin-8B/metabolism
15.
Oncogene ; 39(42): 6606-6618, 2020 10.
Article En | MEDLINE | ID: mdl-32929152

A major clinical challenge of ovarian cancer is the development of malignant ascites accompanied by widespread peritoneal metastasis. In ovarian clear cell carcinoma (OCCC), a challenging subtype of ovarian cancer, this problem is compounded by near-universal primary chemoresistance; patients with advanced stage OCCC thus lack effective therapies and face extremely poor survival rates. Here we show that tumor-cell-expressed serine protease inhibitor Kazal type 1 (SPINK1) is a key driver of OCCC progression and metastasis. Using cell culture models of human OCCC, we find that shRNA silencing of SPINK1 sensitizes tumor cells to anoikis and inhibits proliferation. Knockdown of SPINK1 in OCCC cells also profoundly suppresses peritoneal metastasis in mouse implantation models of human OCCC. We next identify a novel autocrine signaling axis in OCCC cells whereby tumor-cell-produced interleukin-6 (IL-6) regulates SPINK1 expression to stimulate a common protumorigenic gene expression pattern leading to anoikis resistance and proliferation of OCCC cells. We further demonstrate that this signaling pathway can be successfully interrupted with the IL-6Rα inhibitor tocilizumab, sensitizing cells to anoikis in vitro and reducing metastasis in vivo. These results suggest that clinical trials of IL-6 pathway inhibitors in OCCC may be warranted, and that SPINK1 might offer a candidate predictive biomarker in this population.


Adenocarcinoma, Clear Cell/prevention & control , Antibodies, Monoclonal, Humanized/therapeutic use , Interleukin-6/antagonists & inhibitors , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/prevention & control , Trypsin Inhibitor, Kazal Pancreatic/metabolism , Adenocarcinoma, Clear Cell/mortality , Adenocarcinoma, Clear Cell/secondary , Animals , Anoikis/drug effects , Antibodies, Monoclonal, Humanized/pharmacology , Autocrine Communication/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/mortality , Ovary/pathology , Peritoneal Neoplasms/secondary , Prognosis , Signal Transduction/drug effects , Trypsin Inhibitor, Kazal Pancreatic/genetics , Xenograft Model Antitumor Assays
16.
Mech Ageing Dev ; 192: 111355, 2020 12.
Article En | MEDLINE | ID: mdl-32941937

Senescent cells are deeply involved in the induction of tissue damage and aging-related diseases. The identification of factors that eliminate senescent cells or inhibit the senescence-associated secretory phenotype (SASP) in these cells is necessary. Here, we report an avenanthramice C (Avn C) extracted from oat as a new SASP modulator. Treatment with Avn C led to a significant reduction in the levels of markers of senescent cells, with no toxicity observed. The SASP was also inhibited by Avn C treatment, similar to non-senescent cells, and the suppression of cell division by autocrine signals associated with SASP was restored. To investigate the mechanism underlying SASP inhibition by Avn C, we analyzed the effect of Avn C in lipopolysaccharide (LPS)-induced inflammation in non-senescent cells. Avn C inhibited nuclear factor κB (NF-κB) activity and the secretion of inflammatory cytokines before or after LPS treatment. Although the activity of MAP kinases, which are NF-κB upstream signals, was inhibited by Avn C in LPS-induced inflammation, only p38 activity was specifically inhibited in senescent cells. Interestingly, the inhibition of p38 in senescent cells was observed through Avn C-induced 5'-adenosine monophosphate-activated protein kinase (AMPK) activity. Avn C-induced inhibition of the SASP is triggered by senescence-related stress.


Cellular Senescence , Inflammation , MAP Kinase Signaling System/drug effects , NF-kappa B/metabolism , Protein Kinases/metabolism , ortho-Aminobenzoates/pharmacology , AMP-Activated Protein Kinase Kinases , Autocrine Communication/drug effects , Cell Division/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Cellular Senescence/physiology , Cytokines/biosynthesis , Enzyme Inhibitors/pharmacology , Fibroblasts , Humans , Inflammation/drug therapy , Inflammation/metabolism
17.
Eur J Pharmacol ; 885: 173504, 2020 Oct 15.
Article En | MEDLINE | ID: mdl-32858046

Leishmania infected macrophages have conditions to produce adenosine. Despite its known immunosuppressive effects, no studies have yet established whether adenosine alter Leishmania parasitic burden upon macrophage infection. This work aimed at investigating whether endogenous adenosine exerts an autocrine modulation of macrophage response towards Leishmania infection, identifying its origin and potential pharmacological targets for visceral leishmaniasis (VL), using THP-1 differentiated macrophages. Adenosine deaminase treatment of infected THP-1 cells reduced the parasitic burden (29.1 ± 2.2%, P < 0.05). Adenosine A2A and A2B receptor subtypes expression was confirmed by RT-qPCR and by immunocytochemistry and their blockade with selective adenosine A2A and A2B antagonists reduced the parasitic burden [14.5 ± 3.1% (P < 0.05) and 12.3 ± 3.1% (P < 0.05), respectively; and 24.9 ± 2.8% (P < 0.05), by the combination of the two antagonists)], suggesting that adenosine A2 receptors are tonically activated in infected THP-1 differentiated macrophages. The tonic activation of adenosine A2 receptors was dependent on the release of intracellular adenosine through equilibrative nucleoside transporters (ENT1/ENT2): NBTI or dipyridamole reduced (~25%) whereas, when ENTs were blocked, adenosine A2 receptor antagonists failed to reduce and A2 agonists increase parasitic burden. Effects of adenosine A2 receptors antagonists and ENT1/2 inhibitor were prevented by L-NAME, indicating that nitric oxide production inhibition prevents adenosine from increasing parasitic burden. Results suggest that intracellular adenosine, released through ENTs, elicits an autocrine increase in parasitic burden in THP-1 macrophages, through adenosine A2 receptors activation. These observations open the possibility to use well-established ENT inhibitors or adenosine A2 receptor antagonists as new therapeutic approaches in VL.


Adenosine/metabolism , Autocrine Communication/drug effects , Leishmania infantum/drug effects , Leishmaniasis, Visceral/drug therapy , Macrophages/drug effects , Macrophages/parasitology , Receptor, Adenosine A2A/drug effects , Receptor, Adenosine A2B/drug effects , THP-1 Cells/drug effects , Adenosine A2 Receptor Antagonists/pharmacology , Body Burden , Equilibrative Nucleoside Transporter 1/drug effects , Equilibrative-Nucleoside Transporter 2/drug effects , Humans , Leishmaniasis, Visceral/parasitology , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors
18.
Am J Physiol Cell Physiol ; 319(4): C630-C640, 2020 10 01.
Article En | MEDLINE | ID: mdl-32726160

The neurohypophyseal hormone oxytocin (OT) plays critical roles in lactation and parturition, while its function in male reproduction system is largely unknown. This study aims to investigate the effect of OT on regulating transepithelial ion transport in rat cauda epididymal epithelium. With the use of RT-PCR, Western blot, and immunohistochemical analysis, we found that OT receptor (OTR) was expressed and localized at the basal membrane of rat cauda epididymal epithelium. The short-circuit current (Isc) measurement showed that basolateral application of OT to the primary cultured rat cauda epididymal epithelial cells elicited an increase in Isc, which was abrogated by pretreating the epithelial cells with CFTRinh-172, a blocker of cystic fibrosis transmembrane conductance regulator (CFTR). Pretreatment with the prostaglandin H synthase inhibitors indomethacin and piroxicam, or the nonselective antagonists of prostaglandin E2 (PGE2) receptor EP2 or EP4, AH-6809, and AH-23848, significantly attenuated OT-stimulated Isc response. Furthermore, the generation of PGE2 was measured using enzyme-linked immunosorbent assay, demonstrating that OT induced a substantial increase in PGE2 release from primary cultured rat cauda epididymal epithelial cells. In conclusion, activation of OTR by OT triggered PGE2 release, resulting in CFTR-dependent Cl- secretion through paracrine/autocrine pathways in rat cauda epididymal epithelium.


Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Dinoprostone/genetics , Oxytocin/genetics , Receptors, Oxytocin/genetics , Animals , Autocrine Communication/drug effects , Biphenyl Compounds/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Epididymis/drug effects , Epididymis/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Gene Expression Regulation/drug effects , Lactation/genetics , Male , Paracrine Communication/drug effects , Primary Cell Culture , Rats
19.
Sci Rep ; 10(1): 12409, 2020 07 24.
Article En | MEDLINE | ID: mdl-32710087

Vascular endothelial growth factor-A (VEGF) is the angiogenic factor promoting the pathological neovascularization in age-related macular degeneration (AMD) or diabetic macular edema (DME). Evidences have suggested a neurotrophic and neuroprotective role of VEGF, albeit in retina, cellular mechanisms underlying the VEGF neuroprotection remain elusive. Using purified adult retinal ganglion cells (RGCs) in culture, we demonstrated here that VEGF is released by RGCs themselves to promote their own survival, while VEGF neutralization by specific antibodies or traps drastically reduced the RGC survival. These results indicate an autocrine VEGF neuroprotection on RGCs. In parallel, VEGF produced by mixed retinal cells or by mesenchymal stem cells exerted a paracrine neuroprotection on RGCs. Such neuroprotective effect was obtained using the recombinant VEGF-B, suggesting the involvement of VEGF-R1 pathway in VEGF-elicited RGC survival. Finally, glaucomatous patients injected with VEGF traps (ranibizumab or aflibercept) due to either AMD or DME comorbidity, showed a significant reduction of RGC axon fiber layer thickness, consistent with the plausible reduction of the VEGF autocrine stimulation of RGCs. Our results provide evidence of the autocrine neuroprotective function of VEGF on RGCs is crucially involved to preserve injured RGCs such as in glaucomatous patients.


Glaucoma/drug therapy , Retinal Ganglion Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Aged , Aged, 80 and over , Animals , Autocrine Communication/drug effects , Cell Survival/drug effects , Cells, Cultured , Diabetic Retinopathy/complications , Diabetic Retinopathy/drug therapy , Female , Glaucoma/etiology , Glaucoma/pathology , Humans , Intravitreal Injections , Macular Degeneration/complications , Macular Degeneration/drug therapy , Macular Edema/complications , Macular Edema/drug therapy , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Paracrine Communication/drug effects , Primary Cell Culture , Prospective Studies , Ranibizumab/administration & dosage , Rats , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor B/genetics , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-1/metabolism
20.
Circ Res ; 127(7): e166-e183, 2020 09 11.
Article En | MEDLINE | ID: mdl-32588751

RATIONALE: Ca2+ signaling is a key and ubiquitous actor of cell organization and its modulation controls many cellular responses. SERCAs (sarco-endoplasmic reticulum Ca2+-ATPases) pump Ca2+ into internal stores that play a major role in the cytosolic Ca2+ concentration rise upon cell activation. Platelets exhibit 2 types of SERCAs, SERCA2b and SERCA3 (SERCA3 deficient mice), which may exert specific roles, yet ill-defined. We have recently shown that Ca2+ mobilization from SERCA3-dependent stores was required for full platelet activation in weak stimulation conditions. OBJECTIVE: To uncover the signaling mechanisms associated with Ca2+ mobilization from SERCA3-dependent stores leading to ADP secretion. METHODS AND RESULTS: Using platelets from wild-type or Serca3-deficient mice, we demonstrated that an early (within 5-10 s following stimulation) secretion of ADP specifically dependent on SERCA3 stored Ca2+ is exclusively mobilized by nicotinic acid adenosine dinucleotide-phosphate (NAADP): both Ca2+ mobilization from SERCA3-dependent stores and primary ADP secretion are blocked by the NAADP receptor antagonist Ned-19, and reciprocally both are stimulated by permeant NAADP. In contrast, Ca2+ mobilization from SERCA3-dependent stores and primary ADP secretion were unaffected by inhibition of the production of IP3 (inositol-1,4,5-trisphosphate) by phospholipase-C and accordingly were not stimulated by permeant IP3. CONCLUSIONS: Upon activation, an NAADP/SERCA3 Ca2+ mobilization pathway initiates an early ADP secretion, potentiating platelet activation, and a secondary wave of ADP secretion driven by both an IP3/SERCA2b-dependent Ca2+ stores pathway and the NAADP/SERCA3 pathway. This does not exclude that Ca2+ mobilized from SERCA3 stores may also enhance platelet global reactivity to agonists. Because of its modulating effect on platelet activation, this NAADP-SERCA3 pathway may be a relevant target for anti-thrombotic therapy. Graphic Abstract: A graphic abstract is available for this article.


Adenosine Diphosphate/blood , Autocrine Communication , Blood Platelets/enzymology , Calcium Signaling , NADP/analogs & derivatives , Platelet Activation , Sarcoplasmic Reticulum Calcium-Transporting ATPases/blood , Animals , Autocrine Communication/drug effects , Blood Platelets/drug effects , Calcium Signaling/drug effects , Humans , Inositol 1,4,5-Trisphosphate/blood , Mice, Inbred C57BL , Mice, Knockout , NADP/blood , Platelet Activation/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Secretory Pathway , Thrombin/pharmacology , Thromboxane A2/blood , Time Factors
...