Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 903
Filter
1.
CNS Neurosci Ther ; 30(5): e14749, 2024 05.
Article in English | MEDLINE | ID: mdl-38739004

ABSTRACT

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.


Subject(s)
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 in English | MEDLINE | ID: mdl-35840551

ABSTRACT

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.


Subject(s)
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.
Development ; 149(3)2022 02 01.
Article in English | MEDLINE | ID: mdl-35112129

ABSTRACT

The tracheal epithelium is a primary target for pulmonary diseases as it provides a conduit for air flow between the environment and the lung lobes. The cellular and molecular mechanisms underlying airway epithelial cell proliferation and differentiation remain poorly understood. Hedgehog (HH) signaling orchestrates communication between epithelial and mesenchymal cells in the lung, where it modulates stromal cell proliferation, differentiation and signaling back to the epithelium. Here, we reveal a previously unreported autocrine function of HH signaling in airway epithelial cells. Epithelial cell depletion of the ligand sonic hedgehog (SHH) or its effector smoothened (SMO) causes defects in both epithelial cell proliferation and differentiation. In cultured primary human airway epithelial cells, HH signaling inhibition also hampers cell proliferation and differentiation. Epithelial HH function is mediated, at least in part, through transcriptional activation, as HH signaling inhibition leads to downregulation of cell type-specific transcription factor genes in both the mouse trachea and human airway epithelial cells. These results provide new insights into the role of HH signaling in epithelial cell proliferation and differentiation during airway development.


Subject(s)
Autocrine Communication/physiology , Cell Differentiation , Cell Proliferation , Hedgehog Proteins/metabolism , Signal Transduction/genetics , Animals , Cells, Cultured , Down-Regulation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Humans , Lung/growth & development , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Smoothened Receptor/deficiency , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Trachea/cytology , Trachea/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Purinergic Signal ; 17(3): 345-370, 2021 09.
Article in English | MEDLINE | ID: mdl-33982134

ABSTRACT

Cancer comprises a collection of diseases that occur in almost any tissue and it is characterized by an abnormal and uncontrolled cell growth that results in tumor formation and propagation to other tissues, causing tissue and organ malfunction and death. Despite the undeniable improvement in cancer diagnostics and therapy, there is an urgent need for new therapeutic and preventive strategies with improved efficacy and fewer side effects. In this context, purinergic signaling emerges as an interesting candidate as a cancer biomarker or therapeutic target. There is abundant evidence that tumor cells have significant changes in the expression of purinergic receptors, which comprise the G-protein coupled P2Y and AdoR families of receptors and the ligand-gated ion channel P2X receptors. Tumor cells also exhibit changes in the expression of nucleotidases and other enzymes involved in nucleotide metabolism, and the concentrations of extracellular nucleotides are significantly higher than those observed in normal cells. In this review, we will focus on the potential role of purinergic signaling in the ten most lethal cancers (lung, breast, colorectal, liver, stomach, prostate, cervical, esophagus, pancreas, and ovary), which together are responsible for more than 5 million annual deaths.


Subject(s)
Adenosine Triphosphate/metabolism , Autocrine Communication/physiology , Neoplasms/metabolism , Paracrine Communication/physiology , Receptors, Purinergic/metabolism , Adenosine Triphosphate/genetics , Animals , Humans , Neoplasms/genetics , Neoplasms/mortality , Receptors, Purinergic/genetics , Signal Transduction/physiology
5.
Cancer Res ; 81(7): 1802-1812, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33547160

ABSTRACT

Signaling between cancer and nonmalignant (stromal) cells in the tumor microenvironment (TME) is a key to tumor progression. Here, we deconvoluted bulk tumor transcriptomes to infer cross-talk between ligands and receptors on cancer and stromal cells in the TME of 20 solid tumor types. This approach recovered known transcriptional hallmarks of cancer and stromal cells and was concordant with single-cell, in situ hybridization and IHC data. Inferred autocrine cancer cell interactions varied between tissues but often converged on Ephrin, BMP, and FGFR-signaling pathways. Analysis of immune checkpoints nominated interactions with high levels of cancer-to-immune cross-talk across distinct tumor types. Strikingly, PD-L1 was found to be highly expressed in stromal rather than cancer cells. Overall, our study presents a new resource for hypothesis generation and exploration of cross-talk in the TME. SIGNIFICANCE: This study provides deconvoluted bulk tumor transcriptomes across multiple cancer types to infer cross-talk in the tumor microenvironment.


Subject(s)
Neoplasms , Receptor Cross-Talk/physiology , Tumor Microenvironment , Autocrine Communication/physiology , Cell Communication/genetics , Computational Biology , Datasets as Topic , Female , Genomics/methods , Humans , Ligands , Male , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/physiology , Tumor Microenvironment/genetics , Exome Sequencing
6.
J Neurosci ; 41(8): 1650-1664, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33452226

ABSTRACT

Promoting oligodendrocyte (OL) differentiation represents a promising option for remyelination therapy for treating the demyelinating disease multiple sclerosis (MS). The Wnt effector transcription factor 7-like 2 (TCF7l2) was upregulated in MS lesions and had been proposed to inhibit OL differentiation. Recent data suggest the opposite yet underlying mechanisms remain elusive. Here, we unravel a previously unappreciated function of TCF7l2 in controlling autocrine bone morphogenetic protein (BMP)4-mediated signaling. Disrupting TCF7l2 in mice of both sexes results in oligodendroglial-specific BMP4 upregulation and canonical BMP4 signaling activation in vivo Mechanistically, TCF7l2 binds to Bmp4 gene regulatory element and directly represses its transcriptional activity. Functionally, enforced TCF7l2 expression promotes OL differentiation by reducing autocrine BMP4 secretion and dampening BMP4 signaling. Importantly, compound genetic disruption demonstrates that oligodendroglial-specific BMP4 deletion rescues arrested OL differentiation elicited by TCF7l2 disruption in vivo Collectively, our study reveals a novel connection between TCF7l2 and BMP4 in oligodendroglial lineage and provides new insights into augmenting TCF7l2 for promoting remyelination in demyelinating disorders such as MS.SIGNIFICANCE STATEMENT Incomplete or failed myelin repairs, primarily resulting from the arrested differentiation of myelin-forming oligodendrocytes (OLs) from oligodendroglial progenitor cells, is one of the major reasons for neurologic progression in people affected by multiple sclerosis (MS). Using in vitro culture systems and in vivo animal models, this study unraveled a previously unrecognized autocrine regulation of bone morphogenetic protein (BMP)4-mediated signaling by the Wnt effector transcription factor 7-like 2 (TCF7l2). We showed for the first time that TCF7l2 promotes oligodendroglial differentiation by repressing BMP4-mediated activity, which is dysregulated in MS lesions. Our study suggests that elevating TCF7l2 expression may be possible in overcoming arrested oligodendroglial differentiation as observed in MS patients.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Gene Expression Regulation/physiology , Neurogenesis/physiology , Oligodendroglia/cytology , Transcription Factor 7-Like 2 Protein/metabolism , Animals , Autocrine Communication/physiology , Brain/cytology , Brain/metabolism , Cell Differentiation/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism
7.
Theranostics ; 11(6): 2722-2741, 2021.
Article in English | MEDLINE | ID: mdl-33456569

ABSTRACT

Rationale: Little is known about the roles of proteoglycans in esophageal cancer. This study aims to investigate the roles and mechanisms of serglycin (SRGN) proteoglycan in promoting metastasis of esophageal squamous cell carcinoma (ESCC). Methods: Reverse phase protein array analysis was used to identify activated signaling pathways in SRGN-overexpressing cells. Chemokine array was used to identify differentially secreted factors from SRGN-overexpressing cells. Binding between SRGN and potential interacting partners was evaluated using proximity ligation assay and co-immunoprecipitation. The glycosaminoglycan (GAG) chains of SRGN were characterized using fluorophore-assisted carbohydrate electrophoresis. Tissue microarray and serum samples were used to determine the correlation of SRGN expression with clinicopathological parameters and patient survival. Results: In vitro and in vivo experiments showed that SRGN promoted invasion and metastasis in ESCC via activating ERK pathway, stabilizing c-Myc and upregulating the secretion of matrix metalloproteinases. SRGN-knockdown suppressed tumorigenic hallmarks. These SRGN-elicited functions were carried out in an autocrine manner by inducing the secretion of midkine (MDK), which was further identified as a novel binding partner of SRGN for the formation of a SRGN/MDK/CD44 complex. In addition, SRGN interacted with MDK and matrix metalloproteinase 2 in ESCC via its GAG chains, which were mainly decorated with chondroitin sulfate comprising of ∆di-4S and ∆di-6S CS. Clinically, high expression of serum SRGN in serum of patients with ESCC was an independent prognostic marker for poor survival. Conclusions: This study provides the first evidence that elevated serum SRGN has prognostic significance in patients with ESCC, and sheds light on the molecular mechanism by which elevated circulating SRGN in cancer patients might promote cancer progression.


Subject(s)
Autocrine Communication/physiology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Proteoglycans/metabolism , Vesicular Transport Proteins/metabolism , Animals , Biomarkers, Tumor/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Movement/physiology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , Hyaluronan Receptors/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Midkine/metabolism , Prognosis , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/physiology , Up-Regulation/physiology
8.
J Am Heart Assoc ; 10(3): e019169, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33470124

ABSTRACT

The myocardium consists of different cell types, of which endothelial cells, cardiomyocytes, and fibroblasts are the most abundant. Communication between these different cell types, also called paracrine signaling, is essential for normal cardiac function, but also important in cardiac remodeling and heart failure. Systematic studies on the expression of ligands and their corresponding receptors in different cell types showed that for 60% of the expressed ligands in a particular cell, the receptor is also expressed. The fact that many ligand-receptor pairs are present in most cells, including the major cell types in the heart, indicates that autocrine signaling is a widespread phenomenon. Autocrine signaling in cardiac remodeling and heart failure is involved in all pathophysiological mechanisms generally observed: hypertrophy, fibrosis, angiogenesis, cell survival, and inflammation. Herein, we review ligand-receptor pairs present in the major cardiac cell types based on RNA-sequencing expression databases, and we review current literature on extracellular signaling proteins with an autocrine function in the heart; these include C-type natriuretic peptide, fibroblast growth factors 2, F21, and 23, macrophage migration inhibitory factor, heparin binding-epidermal growth factor, angiopoietin-like protein 2, leptin, adiponectin, follistatin-like 1, apelin, neuregulin 1, vascular endothelial growth factor, transforming growth factor ß, wingless-type integration site family, member 1-induced secreted protein-1, interleukin 11, connective tissue growth factor/cellular communication network factor, and calcitonin gene‒related peptide. The large number of autocrine signaling factors that have been studied in the literature supports the concept that autocrine signaling is an essential part of myocardial biology and disease.


Subject(s)
Autocrine Communication/physiology , Heart Failure/therapy , Myocytes, Cardiac/metabolism , Ventricular Remodeling/physiology , Animals , Cells, Cultured , Heart Failure/metabolism , Heart Failure/pathology , Humans , Myocytes, Cardiac/pathology , Signal Transduction
9.
Int J Mol Sci ; 22(2)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33478130

ABSTRACT

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.


Subject(s)
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
10.
Am J Physiol Endocrinol Metab ; 320(1): E78-E86, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33103455

ABSTRACT

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.


Subject(s)
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
11.
J Neurosci ; 41(2): 251-268, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33208471

ABSTRACT

The developing CNS is exposed to physiological hypoxia, under which hypoxia-inducible factor α (HIFα) is stabilized and plays a crucial role in regulating neural development. The cellular and molecular mechanisms of HIFα in developmental myelination remain incompletely understood. A previous concept proposes that HIFα regulates CNS developmental myelination by activating the autocrine Wnt/ß-catenin signaling in oligodendrocyte progenitor cells (OPCs). Here, by analyzing a battery of genetic mice of both sexes, we presented in vivo evidence supporting an alternative understanding of oligodendroglial HIFα-regulated developmental myelination. At the cellular level, we found that HIFα was required for developmental myelination by transiently controlling upstream OPC differentiation but not downstream oligodendrocyte maturation and that HIFα dysregulation in OPCs but not oligodendrocytes disturbed normal developmental myelination. We demonstrated that HIFα played a minor, if any, role in regulating canonical Wnt signaling in the oligodendroglial lineage or in the CNS. At the molecular level, blocking autocrine Wnt signaling did not affect HIFα-regulated OPC differentiation and myelination. We further identified HIFα-Sox9 regulatory axis as an underlying molecular mechanism in HIFα-regulated OPC differentiation. Our findings support a concept shift in our mechanistic understanding of HIFα-regulated CNS myelination from the previous Wnt-dependent view to a Wnt-independent one and unveil a previously unappreciated HIFα-Sox9 pathway in regulating OPC differentiation.SIGNIFICANCE STATEMENT Promoting disturbed developmental myelination is a promising option in treating diffuse white matter injury, previously called periventricular leukomalacia, a major form of brain injury affecting premature infants. In the developing CNS, hypoxia-inducible factor α (HIFα) is a key regulator that adapts neural cells to physiological and pathologic hypoxic cues. The role and mechanism of HIFα in oligodendroglial myelination, which is severely disturbed in preterm infants affected with diffuse white matter injury, is incompletely understood. Our findings presented here represent a concept shift in our mechanistic understanding of HIFα-regulated developmental myelination and suggest the potential of intervening with an oligodendroglial HIFα-mediated signaling pathway to mitigate disturbed myelination in premature white matter injury.


Subject(s)
Autocrine Communication/physiology , Central Nervous System/growth & development , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Myelin Sheath/physiology , Wnt Signaling Pathway/physiology , Animals , Cell Differentiation , Estrogen Antagonists/pharmacology , Female , Humans , Hypoxia-Ischemia, Brain/pathology , Male , Mice , Mice, Transgenic , Neural Stem Cells/physiology , Oligodendroglia/physiology , SOX9 Transcription Factor/genetics , Tamoxifen/pharmacology , White Matter/pathology
12.
J Exp Med ; 218(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33125053

ABSTRACT

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.


Subject(s)
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
13.
Mol Metab ; 42: 101103, 2020 12.
Article in English | MEDLINE | ID: mdl-33091626

ABSTRACT

OBJECTIVES: Long-chain fatty acids (LCFAs) released from adipocytes inhibit lipolysis through an unclear mechanism. We hypothesized that the LCFA receptor, FFAR4 (GPR120), which is highly expressed in adipocytes, may be involved in this feedback regulation. METHODS AND RESULTS: Liquid chromatography mass spectrometry (LC-MS) analysis of conditioned media from isoproterenol-stimulated primary cultures of murine and human adipocytes demonstrated that most of the released non-esterified free fatty acids (NEFAs) are known agonists for FFAR4. In agreement with this, conditioned medium from isoproterenol-treated adipocytes stimulated signaling strongly in FFAR4 transfected COS-7 cells as opposed to non-transfected control cells. In transfected 3T3-L1 cells, FFAR4 agonism stimulated Gi- and Go-mini G protein binding more strongly than Gq, effects which were blocked by the selective FFAR4 antagonist AH7614. In primary cultures of murine white adipocytes, the synthetic, selective FFAR4 agonist CpdA inhibited isoproterenol-induced intracellular cAMP accumulation in a manner similar to the antilipolytic control agent nicotinic acid acting through another receptor, HCAR2. In vivo, oral gavage with the synthetic, specific FFAR4 agonist CpdB decreased the level of circulating NEFAs in fasting lean mice to a similar degree as nicotinic acid. In agreement with the identified anti-lipolytic effect of FFAR4, plasma NEFAs and glycerol were increased in FFAR4-deficient mice as compared to littermate controls despite having elevated insulin levels, and cAMP accumulation in primary adipocyte cultures was augmented by treatment with the FFAR4 antagonist conceivably by blocking the stimulatory tone of endogenous NEFAs on FFAR4. CONCLUSIONS: In white adipocytes, FFAR4 functions as an NEFA-activated, autocrine, negative feedback regulator of lipolysis by decreasing cAMP though Gi-mediated signaling.


Subject(s)
Fatty Acids, Nonesterified/metabolism , Receptors, G-Protein-Coupled/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes, White/metabolism , Adipose Tissue/metabolism , Adipose Tissue, White/metabolism , Animals , Autocrine Communication/physiology , Chromatography, Liquid/methods , Culture Media, Conditioned/pharmacology , Fatty Acids/metabolism , Feedback, Physiological/physiology , Female , Humans , Lipolysis/physiology , Male , Mass Spectrometry/methods , Mice , Mice, Inbred C57BL , Receptors, G-Protein-Coupled/physiology
14.
Diabetologia ; 63(10): 2057-2063, 2020 10.
Article in English | MEDLINE | ID: mdl-32894316

ABSTRACT

Pancreatic beta cells are the only cell type in our body capable of producing and secreting insulin to instruct the insulin-sensitive cells and tissues of our bodies to absorb nutrients after a meal. Accurate control of insulin release is of critical importance; too little insulin leads to diabetes, while an excess of insulin can cause potentially fatal hypoglycaemia. Yet, the pancreas of most people will control insulin secretion safely and effectively over decades and in response to glucose excursions driven by tens of thousands of meals. Because we only become aware of the important contributions of the pancreas when it fails to maintain glucose homeostasis, it is easy to forget just how well insulin release from a healthy pancreas is matched to insulin need to ensure stable blood glucose levels. Beta cells achieve this feat by extensive crosstalk with the rest of the endocrine cell types in the islet, notably the glucagon-producing alpha cells and somatostatin-producing delta cells. Here I will review the important paracrine contributions that each of these cells makes to the stimulation and subsequent inhibition of insulin release in response to a transient nutrient stimulation, and make the case that a breakdown of this local crosstalk contributes to the pathophysiology of diabetes. Graphical abstract.


Subject(s)
Glucagon-Secreting Cells/metabolism , Insulin Secretion/physiology , Insulin-Secreting Cells/metabolism , Paracrine Communication/physiology , Somatostatin-Secreting Cells/metabolism , Acetylcholine/metabolism , Autocrine Communication/physiology , Corticotropin-Releasing Hormone/metabolism , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Serotonin/metabolism , Somatostatin/metabolism , Urocortins/metabolism , gamma-Aminobutyric Acid/metabolism
15.
Int J Biochem Cell Biol ; 127: 105838, 2020 10.
Article in English | MEDLINE | ID: mdl-32858191

ABSTRACT

Epidermal growth factor receptor is a cancer driver whose nuclear localization has been associated with the progression of prostate cancer to the castration-resistant phenotype. Previous reports indicated a functional interaction between this receptor and the protein Annexin A1, which has also been associated with aggressive tumors. The molecular pathogenesis of castration-resistant prostate cancer remains largely unresolved, and herein we have demonstrated the correlation between the expression levels and localization of the epidermal growth factor receptor and Annexin A1 in prostate cancer samples and cell lines. Interestingly, a higher expression of both proteins was detected in castration-resistant prostate cancer cell lines and the strongest correlation was seen at the nuclear level. We verified that Annexin A1 interacts with the epidermal growth factor receptor, and by using prostate cancer cell lines knocked down for Annexin A1, we succeeded in demonstrating that Annexin A1 promotes the nuclear localization of epidermal growth factor receptor. Finally, we showed that Annexin A1 activates an autocrine signaling in castration-resistant prostate cells through the formyl peptide receptor 1. The inhibition of such signaling by Cyclosporin H inhibits the nuclear localization of epidermal growth factor receptor and its downstream signaling. The present work sheds light on the functional interaction between nuclear epidermal growth factor receptor and nuclear Annexin A1 in castration-resistant prostate cancer. Therefore, strategies to inhibit the nuclear localization of epidermal growth factor receptor through the suppression of the Annexin A1 autocrine loop could represent an important intervention strategy for castration-resistant prostate cancer.


Subject(s)
Annexin A1/metabolism , Cell Nucleus/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Aged , Annexin A1/genetics , Autocrine Communication/physiology , Cell Line, Tumor , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Male , Middle Aged , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/genetics , Signal Transduction
16.
Cell Death Dis ; 11(8): 650, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32811814

ABSTRACT

Metastasis is the main cause of failure of cancer treatment. Metastatic colonization is regarded the most rate-limiting step of metastasis and is subjected to regulation by a plethora of biological factors and processes. On one hand, regulation of metastatic colonization by autophagy appears to be stage- and context-dependent, whereas mechanistic characterization remains elusive. On the other hand, interactions between the tumor cells and their microenvironment in metastasis have long been appreciated, whether the secretome of tumor cells can effectively reshape the tumor microenvironment has not been elucidated mechanistically. In the present study, we have identified "SEC23A-S1008-BECLIN1-autophagy axis" in the autophagic regulation of metastatic colonization step, a mechanism that tumor cells can exploit autophagy to exert self-restrain for clonogenic proliferation before the favorable tumor microenvironment is established. Specifically, we employed a paired lung-derived oligometastatic cell line (OL) and the homologous polymetastatic cell line (POL) from human melanoma cell line M14 that differ in colonization efficiency. We show that S100A8 transported by SEC23A inhibits metastatic colonization via autocrine activation of autophagy. Furthermore, we verified the clinical relevance of our experimental findings by bioinformatics analysis of the expression of Sec23a and S100A8 and the clinical-pathological associations. We demonstrate that higher Sec23a and Atg5 expression levels appear to be protective factors and favorable diagnostic (TNM staging) and prognostic (overall survival) markers for skin cutaneous melanoma (SKCM) and colon adenocarcinoma (COAD) patients. And we confirm the bioinformatics analysis results with SKCM biopsy samples.


Subject(s)
Calgranulin A/metabolism , Neoplasm Metastasis/pathology , Vesicular Transport Proteins/metabolism , Animals , Autocrine Communication/physiology , Autophagy/physiology , Beclin-1/metabolism , Calgranulin A/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Humans , Male , Melanoma/pathology , Mice, SCID , Prognosis , RNA, Small Interfering/metabolism , Skin Neoplasms/pathology , Tumor Microenvironment/physiology , Vesicular Transport Proteins/physiology
17.
Cells ; 9(8)2020 08 11.
Article in English | MEDLINE | ID: mdl-32796542

ABSTRACT

Protein kinase type C-ε (PKCε) plays important roles in the sensitization of primary afferent nociceptors, such as ion channel phosphorylation, that in turn promotes mechanical hyperalgesia and pain chronification. In these neurons, PKCε is modulated through the local release of mediators by the surrounding Schwann cells (SCs). The progesterone metabolite allopregnanolone (ALLO) is endogenously synthesized by SCs, whereas it has proven to be a crucial mediator of neuron-glia interaction in peripheral nerve fibers. Biomolecular and pharmacological studies on rat primary SCs and dorsal root ganglia (DRG) neuronal cultures were aimed at investigating the hypothesis that ALLO modulates neuronal PKCε, playing a role in peripheral nociception. We found that SCs tonically release ALLO, which, in turn, autocrinally upregulated the synthesis of the growth factor brain-derived neurotrophic factor (BDNF). Subsequently, glial BDNF paracrinally activates PKCε via trkB in DRG sensory neurons. Herein, we report a novel mechanism of SCs-neuron cross-talk in the peripheral nervous system, highlighting a key role of ALLO and BDNF in nociceptor sensitization. These findings emphasize promising targets for inhibiting the development and chronification of neuropathic pain.


Subject(s)
Autocrine Communication/physiology , Brain-Derived Neurotrophic Factor/metabolism , Neuralgia/metabolism , Paracrine Communication/physiology , Pregnanolone/metabolism , Schwann Cells/metabolism , Animals , Autocrine Communication/genetics , Blotting, Western , Cells, Cultured , Chromatography, Liquid , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Ganglia, Spinal/metabolism , Humans , Hyperalgesia/metabolism , Paracrine Communication/genetics , Rats, Sprague-Dawley , Sensory Receptor Cells/metabolism , Tandem Mass Spectrometry
18.
Philos Trans R Soc Lond B Biol Sci ; 375(1807): 20190386, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32713299

ABSTRACT

Epithelial branching morphogenesis drives the development of organs such as the lung, salivary gland, kidney and the mammary gland. It involves cell proliferation, cell differentiation and cell migration. An elaborate network of chemical and mechanical signals between the epithelium and the surrounding mesenchymal tissues regulates the formation and growth of branching organs. Surprisingly, when cultured in isolation from mesenchymal tissues, many epithelial tissues retain the ability to exhibit branching morphogenesis even in the absence of proliferation. In this work, we propose a simple, experimentally plausible mechanism that can drive branching morphogenesis in the absence of proliferation and cross-talk with the surrounding mesenchymal tissue. The assumptions of our mathematical model derive from in vitro observations of the behaviour of mammary epithelial cells. These data show that autocrine secretion of the growth factor TGF[Formula: see text]1 inhibits the formation of cell protrusions, leading to curvature-dependent inhibition of sprouting. Our hybrid cellular Potts and partial-differential equation model correctly reproduces the experimentally observed tissue-geometry-dependent determination of the sites of branching, and it suffices for the formation of self-avoiding branching structures in the absence and also in the presence of cell proliferation. This article is part of the theme issue 'Multi-scale analysis and modelling of collective migration in biological systems'.


Subject(s)
Autocrine Communication/physiology , Cell Movement/physiology , Cell Proliferation , Morphogenesis , Animals , Models, Biological , Stochastic Processes
19.
Reprod Domest Anim ; 55 Suppl 2: 66-73, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32347633

ABSTRACT

Dog reproductive cycle is unique among other mammals in that females experience long and variable periods of ovarian inactivity. Neuroendocrine controls of the reproductive cycle have been thoroughly studied in the dog. However, there is little information regarding endocrine, paracrine and autocrine controls of dog ovarian folliculogenesis. Advancements in the understanding of mechanisms regulating dog ovarian follicle development will be helpful in the establishment of an approach to control cyclicity in this species. Furthermore, such information will likely be useful for the establishment of an in vitro follicle culture system to preserve fertility of genetically valuable disease models or endangered canids. This review highlights current knowledge on dog folliculogenesis with emphasis on endocrine, paracrine and autocrine controls of follicular development.


Subject(s)
Dogs/physiology , Ovarian Follicle/growth & development , Animals , Autocrine Communication/physiology , Endocrine Glands/physiology , Female , Ovarian Follicle/physiology , Paracrine Communication/physiology , Reproduction/physiology
20.
Am J Physiol Regul Integr Comp Physiol ; 318(3): R590-R604, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31913658

ABSTRACT

Lymphatic vessels play a critical role in mounting a proper immune response by trafficking peripheral immune cells to draining lymph nodes. Mast cells (MCs) are well known for their roles in type I hypersensitivity reactions, but little is known about their secretory regulation in the lymphatic niche. MCs, as innate sensor and effector cells, reside close to mesenteric lymphatic vessels (MLVs), and their activation and ability to release histamine influences the lymphatic microenvironment in a histamine-NF-κB-dependent manner. Using an established experimental protocol involving surgical isolation of rat mesenteric tissue segments, including MLVs and surrounding perilymphatic tissues, we tested the hypothesis that perilymphatic mesenteric MCs possess histamine receptors (HRs) that bind and respond to the histamine released from these same MCs. Under various experimental conditions, including inflammatory stimulation by LPS, we measured histamine in mesenteric perilymphatic tissues, evaluated expression of histidine decarboxylase in MCs along with the degree of MC degranulation, assessed the functional status of HRs in MCs, and evaluated the ability of histamine itself to induce MC activation. Finally, we evaluated the importance of MCs and HR1 and -2 for MLV-directed trafficking of CD11b/c-positive cells during acute tissue inflammation. Our data indicate the existence of a functionally potent MC-histamine autocrine regulatory loop, the elements of which are crucially important for acute inflammation-induced trafficking of the CD11b/c-positive cells toward MLVs. This MC-histamine loop serves as a first-line cellular servo control system, playing a key role in the innate and adaptive immune response as well as NF-κB-mediated maintenance of body homeostasis.


Subject(s)
Autocrine Communication/physiology , Inflammation/metabolism , Mast Cells/metabolism , Mesentery/metabolism , Animals , Histamine/pharmacology , Homeostasis/physiology , Inflammation/physiopathology , Lymphatic Vessels/metabolism , Male , NF-kappa B/metabolism , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...