Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.471
Filter
1.
Arch Dermatol Res ; 316(7): 455, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38967656

ABSTRACT

Tirbanibulin 1% ointment is a synthetic antiproliferative agent approved in 2021 by the European Union for treating actinic keratoses (AK). Topical tirbanibulin has clinically resolved HPV-57 ( +) squamous cell carcinoma (SCC), HPV-16 ( +) vulvar high-grade squamous intraepithelial lesion, epidermodysplasia verruciformis, and condyloma. We examined how tirbanibulin might affect HPV oncoprotein expression and affect other cellular pathways involved in cell proliferation and transformation. We treated the HeLa cell line, containing integrated HPV-18, with increasing doses of tirbanibulin to determine the effects on cell proliferation. Immunoblotting was performed with antibodies against the Src canonical pathway, HPV 18 E6 and E7 transcription regulation, apoptosis, and invasion and metastasis pathways. Cell proliferation assays with tirbanibulin determined the half-maximal inhibitory concentration (IC50) of HeLa cells to be 31.49 nmol/L. Increasing concentrations of tirbanibulin downregulates the protein expression of Src (p < 0.001), phospho-Src (p < 0.001), Ras (p < 0.01), c-Raf (p < 0.001), ERK1 (p < 0.001), phospho-ERK1 (p < 0.001), phospho-ERK2 (p < 0.01), phospho-Mnk1 (p < 0.001), eIF4E (p < 0.01), phospho-eIF4E (p < 0.001), E6 (p < 0.01), E7 (p < 0.01), Rb (p < 0.01), phospho-Rb (p < 0.001), MDM2 (p < 0.01), E2F1 (p < 0.001), phospho-FAK (p < 0.001), phospho-p130 Cas (p < 0.001), Mcl-1 (p < 0.01), and Bcl-2 (p < 0.001), but upregulates cPARP (p < 0.001), and cPARP/fPARP (p < 0.001). These results demonstrate that tirbanibulin may impact expression of HPV oncoproteins via the Src- MEK- pathway. Tirbanibulin significantly downregulates oncogenic proteins related to cell cycle regulation and cell proliferation while upregulating apoptosis pathways.


Tirbanibulin is Promising Novel Therapy for Human Papillomavirus (HPV)-associated Diseases.Tirbanibulin 1% ointment is an approved synthetic topical ointment for treating actinic keratoses (AK), a precancer of skin cancer. Topical tirbanibulin has previously been reported to clinically resolve human papillomavirus (HPV)-( +) diseases.In this study, we examine how tirbanibulin may affect the HPV and pathways associated with cancer.We treated the HeLa cell line to determine the effects on HPV cell proliferation. Increasing the concentration of tirbanibulin statistically significantly affected numerous cellular pathways often associated with cancer.These results demonstrate that tirbanibulin may impact expression of HPV oncoproteins and thereby kill cancer cells.


Subject(s)
Cell Proliferation , Down-Regulation , Human papillomavirus 18 , Oncogene Proteins, Viral , Humans , HeLa Cells , Cell Proliferation/drug effects , Oncogene Proteins, Viral/metabolism , Down-Regulation/drug effects , Papillomavirus Infections/virology , Papillomavirus Infections/drug therapy , Papillomavirus E7 Proteins/metabolism , Apoptosis/drug effects , Repressor Proteins/metabolism , Repressor Proteins/genetics , Signal Transduction/drug effects , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/metabolism , src-Family Kinases/metabolism , src-Family Kinases/antagonists & inhibitors , Female , Human Papillomavirus Viruses , DNA-Binding Proteins
2.
Cell Death Dis ; 15(7): 486, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38977663

ABSTRACT

Accumulating evidence suggests that caspase-3 plays critical roles beyond apoptosis, serving pro-survival functions in malignant transformation and tumorigenesis. However, the mechanism of non-apoptotic action of caspase-3 in oncogenic transformation remains unclear. In the present study, we show that caspase-3 is consistently activated in malignant transformation induced by exogenous expression of oncogenic cocktail (c-Myc, p53DD, Oct-4, and H-Ras) in vitro as well as in the mouse mammary tumor virus-polyomavirus middle T antigen (MMTV-PyMT) mouse model of breast cancer. Genetic ablation of caspase-3 significantly attenuated oncogene-induced transformation of mammalian cells and delayed breast cancer progression in MMTV-PyMT transgenic mice. Mechanistically, active caspase-3 triggers the translocation of endonuclease G (EndoG) from mitochondria, which migrates to the nucleus, thereby induces phosphorylation of Src-STAT3 signaling pathway to facilitate oncogenic transformation. Taken together, our data suggest that caspase-3 plays pivotal role in facilitating rather than suppressing oncogene-induced malignant transformation of mammalian cells.


Subject(s)
Caspase 3 , Cell Transformation, Neoplastic , Oncogenes , STAT3 Transcription Factor , Animals , STAT3 Transcription Factor/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Phosphorylation , Caspase 3/metabolism , Mice , Humans , Female , Oncogenes/genetics , src-Family Kinases/metabolism , src-Family Kinases/genetics , Mice, Transgenic , Signal Transduction , Mitochondria/metabolism
3.
Front Immunol ; 15: 1395427, 2024.
Article in English | MEDLINE | ID: mdl-39007135

ABSTRACT

Systemic lupus erythematosus (SLE, lupus) is a debilitating, multisystem autoimmune disease that can affect any organ in the body. The disease is characterized by circulating autoantibodies that accumulate in organs and tissues, which triggers an inflammatory response that can cause permanent damage leading to significant morbidity and mortality. Lyn, a member of the Src family of non-receptor protein tyrosine kinases, is highly implicated in SLE as remarkably both mice lacking Lyn or expressing a gain-of-function mutation in Lyn develop spontaneous lupus-like disease due to altered signaling in B lymphocytes and myeloid cells, suggesting its expression or activation state plays a critical role in maintaining tolerance. The past 30 years of research has begun to elucidate the role of Lyn in a duplicitous signaling network of activating and inhibitory immunoreceptors and related targets, including interactions with the interferon regulatory factor family in the toll-like receptor pathway. Gain-of-function mutations in Lyn have now been identified in human cases and like mouse models, cause severe systemic autoinflammation. Studies of Lyn in SLE patients have presented mixed findings, which may reflect the heterogeneity of disease processes in SLE, with impairment or enhancement in Lyn function affecting subsets of SLE patients that may be a means of stratification. In this review, we present an overview of the phosphorylation and protein-binding targets of Lyn in B lymphocytes and myeloid cells, highlighting the structural domains of the protein that are involved in its function, and provide an update on studies of Lyn in SLE patients.


Subject(s)
Lupus Erythematosus, Systemic , Signal Transduction , src-Family Kinases , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/genetics , src-Family Kinases/metabolism , src-Family Kinases/genetics , Humans , Animals , B-Lymphocytes/immunology , Mice
4.
Cells ; 13(12)2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38920637

ABSTRACT

Excessive levels of glutamate activity could potentially damage and kill neurons. Glutamate excitotoxicity is thought to play a critical role in many CNS and retinal diseases. Accordingly, glutamate excitotoxicity has been used as a model to study neuronal diseases. Immune proteins, such as major histocompatibility complex (MHC) class I molecules and their receptors, play important roles in many neuronal diseases, while T-cell receptors (TCR) are the primary receptors of MHCI. We previously showed that a critical component of TCR, CD3ζ, is expressed by mouse retinal ganglion cells (RGCs). The mutation of CD3ζ or MHCI molecules compromises the development of RGC structure and function. In this study, we investigated whether CD3ζ-mediated molecular signaling regulates RGC death in glutamate excitotoxicity. We show that mutation of CD3ζ significantly increased RGC survival in NMDA-induced excitotoxicity. In addition, we found that several downstream molecules of TCR, including Src (proto-oncogene tyrosine-protein kinase) family kinases (SFKs) and spleen tyrosine kinase (Syk), are expressed by RGCs. Selective inhibition of an SFK member, Hck, or Syk members, Syk or Zap70, significantly increased RGC survival in NMDA-induced excitotoxicity. These results provide direct evidence to reveal the underlying molecular mechanisms that control RGC death under disease conditions.


Subject(s)
CD3 Complex , Glutamic Acid , Retinal Ganglion Cells , Signal Transduction , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Animals , Glutamic Acid/metabolism , Signal Transduction/drug effects , CD3 Complex/metabolism , Mice , Mice, Inbred C57BL , N-Methylaspartate/toxicity , Cell Survival/drug effects , Retina/metabolism , Retina/pathology , src-Family Kinases/metabolism , Syk Kinase/metabolism
5.
Cell Commun Signal ; 22(1): 307, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831315

ABSTRACT

BACKGROUND: Interleukin 24 (IL-24) has been implicated in the nociceptive signaling. However, direct evidence and the precise molecular mechanism underlying IL-24's role in peripheral nociception remain unclear. METHODS: Using patch clamp recording, molecular biological analysis, immunofluorescence labeling, siRNA-mediated knockdown approach and behavior tests, we elucidated the effects of IL-24 on sensory neuronal excitability and peripheral pain sensitivity mediated by T-type Ca2+ channels (T-type channels). RESULTS: IL-24 enhances T-type channel currents (T-currents) in trigeminal ganglion (TG) neurons in a reversible and dose-dependent manner, primarily by activating the interleukin-22 receptor 1 (IL-22R1). Furthermore, we found that the IL-24-induced T-type channel response is mediated through tyrosine-protein kinase Lyn, but not its common downstream target JAK1. IL-24 application significantly activated protein kinase A; this effect was independent of cAMP and prevented by Lyn antagonism. Inhibition of PKA prevented the IL-24-induced T-current response, whereas inhibition of protein kinase C or MAPK kinases had no effect. Functionally, IL-24 increased TG neuronal excitability and enhanced pain sensitivity to mechanical stimuli in mice, both of which were suppressed by blocking T-type channels. In a trigeminal neuropathic pain model induced by chronic constriction injury of the infraorbital nerve, inhibiting IL-22R1 signaling alleviated mechanical allodynia, which was reversed by blocking T-type channels or knocking down Cav3.2. CONCLUSION: Our findings reveal that IL-24 enhances T-currents by stimulating IL-22R1 coupled to Lyn-dependent PKA signaling, leading to TG neuronal hyperexcitability and pain hypersensitivity. Understanding the mechanism of IL-24/IL-22R1 signaling in sensory neurons may pave the way for innovative therapeutic strategies in pain management.


Subject(s)
Calcium Channels, T-Type , Cyclic AMP-Dependent Protein Kinases , Receptors, Interleukin , Sensory Receptor Cells , Signal Transduction , Trigeminal Ganglion , src-Family Kinases , Animals , Calcium Channels, T-Type/metabolism , Calcium Channels, T-Type/genetics , src-Family Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Trigeminal Ganglion/metabolism , Male , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Receptors, Interleukin/metabolism , Mice , Mice, Inbred C57BL , Interleukins/metabolism
6.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38892466

ABSTRACT

Glioblastoma (GBM) is the most common primary malignant brain tumor in adults, with few effective treatments. EGFR alterations, including expression of the truncated variant EGFRvIII, are among the most frequent genomic changes in these tumors. EGFRvIII is known to preferentially signal through STAT5 for oncogenic activation in GBM, yet targeting EGFRvIII has yielded limited clinical success to date. In this study, we employed patient-derived xenograft (PDX) models expressing EGFRvIII to determine the key points of therapeutic vulnerability within the EGFRvIII-STAT5 signaling axis in GBM. Our findings reveal that exogenous expression of paralogs STAT5A and STAT5B augments cell proliferation and that inhibition of STAT5 phosphorylation in vivo improves overall survival in combination with temozolomide (TMZ). STAT5 phosphorylation is independent of JAK1 and JAK2 signaling, instead requiring Src family kinase (SFK) activity. Saracatinib, an SFK inhibitor, attenuates phosphorylation of STAT5 and preferentially sensitizes EGFRvIII+ GBM cells to undergo apoptotic cell death relative to wild-type EGFR. Constitutively active STAT5A or STAT5B mitigates saracatinib sensitivity in EGFRvIII+ cells. In vivo, saracatinib treatment decreased survival in mice bearing EGFR WT tumors compared to the control, yet in EGFRvIII+ tumors, treatment with saracatinib in combination with TMZ preferentially improves survival.


Subject(s)
Benzodioxoles , Cell Proliferation , ErbB Receptors , Glioblastoma , Quinazolines , STAT5 Transcription Factor , Temozolomide , STAT5 Transcription Factor/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Humans , Animals , Quinazolines/pharmacology , Quinazolines/therapeutic use , Benzodioxoles/pharmacology , Benzodioxoles/therapeutic use , Mice , ErbB Receptors/metabolism , Phosphorylation/drug effects , Cell Line, Tumor , Temozolomide/pharmacology , Cell Proliferation/drug effects , Xenograft Model Antitumor Assays , Signal Transduction/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Apoptosis/drug effects , src-Family Kinases/metabolism , Tumor Suppressor Proteins
7.
Philos Trans R Soc Lond B Biol Sci ; 379(1906): 20230236, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-38853562

ABSTRACT

Alternative splicing of Grin1 exon 5 regulates induction of long-term potentiation (LTP) at Schaffer collateral-CA1 synapses: LTP in mice lacking the GluN1 exon 5-encoded N1 cassette (GluN1a mice) is significantly increased compared with that in mice compulsorily expressing this exon (GluN1b mice). The mechanism underlying this difference is unknown. Here, we report that blocking the non-receptor tyrosine kinase Src prevents induction of LTP in GluN1a mice but not in GluN1b. We find that activating Src enhances pharmacologically isolated synaptic N-methyl-d-aspartate receptor (NMDAR) currents in GluN1a mice but not in GluN1b. Moreover, we observe that Src activation increases the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor component of Schaffer collateral-evoked excitatory post-synaptic potentials in GluN1a mice, but this increase is prevented by blocking NMDARs. We conclude that at these synapses, NMDARs in GluN1a mice are subject to upregulation by Src that mediates induction of LTP, whereas NMDARs in GluN1b mice are not regulated by Src, leading to Src-resistance of LTP. Thus, we have uncovered that a key regulatory mechanism for synaptic potentiation is gated by differential splicing of exon 5 of Grin1. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.


Subject(s)
Alternative Splicing , Exons , Long-Term Potentiation , Nerve Tissue Proteins , Receptors, N-Methyl-D-Aspartate , src-Family Kinases , Animals , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Mice , src-Family Kinases/metabolism , src-Family Kinases/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Male , Synapses/physiology , Synapses/metabolism , Mice, Inbred C57BL
8.
Biomed Pharmacother ; 176: 116841, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38834004

ABSTRACT

Metastasis is the leading cause of cancer-related deaths, making the development of novel, more effective therapies imperative to alleviate patient suffering. Metabolic switching is a hallmark of cancer cells that facilitates metastasis. Cancer cells obtain most of their energy and intermediate metabolites, which are required to proliferate and metastasize, through aerobic glycolysis. Previous work from our laboratory has shown that Caveolin-1 (CAV1) expression in cancer cells promotes glycolysis and metastasis. Here, we sought to determine if limiting glycolysis reduced CAV1-enhanced metastasis and to identify the mechanism(s) involved. We evaluated the effects of the glycolysis inhibitor 2-deoxy-D-glucose (2-DG) in metastatic melanoma and breast cancer cell lines expressing or not CAV1. Non-cytotoxic concentrations of 2-DG (1 mM) inhibited the migration of B16-F10 melanoma and MDA-MB-231 breast cancer cells. CAV1-mediated activation of Src/Akt signaling was required for CAV1-enhanced migration and was blocked in the presence of 2-DG. Moreover, inhibition of Akt reduced CAV1-enhanced lung metastasis of B16-F10 cells. Collectively, these findings highlight the importance of CAV1-induced metabolic reprogramming for metastasis and point towards possible therapeutic approaches to prevent metastatic disease by inhibiting glycolysis and Src/Akt signaling.


Subject(s)
Caveolin 1 , Cell Movement , Deoxyglucose , Glycolysis , Proto-Oncogene Proteins c-akt , Signal Transduction , src-Family Kinases , Caveolin 1/metabolism , Glycolysis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Animals , Signal Transduction/drug effects , src-Family Kinases/metabolism , src-Family Kinases/antagonists & inhibitors , Humans , Cell Line, Tumor , Mice , Cell Movement/drug effects , Deoxyglucose/pharmacology , Female , Neoplasm Metastasis , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Lung Neoplasms/secondary , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/drug therapy , Mice, Inbred C57BL
9.
J Cell Sci ; 137(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38881365

ABSTRACT

Endothelial cells lining the blood vessel wall communicate intricately with the surrounding extracellular matrix, translating mechanical cues into biochemical signals. Moreover, vessels require the capability to enzymatically degrade the matrix surrounding them, to facilitate vascular expansion. c-Src plays a key role in blood vessel growth, with its loss in the endothelium reducing vessel sprouting and focal adhesion signalling. Here, we show that constitutive activation of c-Src in endothelial cells results in rapid vascular expansion, operating independently of growth factor stimulation or fluid shear stress forces. This is driven by an increase in focal adhesion signalling and size, with enhancement of localised secretion of matrix metalloproteinases responsible for extracellular matrix remodelling. Inhibition of matrix metalloproteinase activity results in a robust rescue of the vascular expansion elicited by heightened c-Src activity. This supports the premise that moderating focal adhesion-related events and matrix degradation can counteract abnormal vascular expansion, with implications for pathologies driven by unusual vascular morphologies.


Subject(s)
Extracellular Matrix , Focal Adhesions , src-Family Kinases , Focal Adhesions/metabolism , Extracellular Matrix/metabolism , Humans , src-Family Kinases/metabolism , src-Family Kinases/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Animals , CSK Tyrosine-Protein Kinase/metabolism , Signal Transduction , Endothelial Cells/metabolism , Endothelial Cells/pathology , Matrix Metalloproteinases/metabolism
10.
Elife ; 132024 May 23.
Article in English | MEDLINE | ID: mdl-38780416

ABSTRACT

Protein phosphorylation is one of the major molecular mechanisms regulating protein activity and function throughout the cell. Pannexin 1 (PANX1) is a large-pore channel permeable to ATP and other cellular metabolites. Its tyrosine phosphorylation and subsequent activation have been found to play critical roles in diverse cellular conditions, including neuronal cell death, acute inflammation, and smooth muscle contraction. Specifically, the non-receptor kinase Src has been reported to phosphorylate Tyr198 and Tyr308 of mouse PANX1 (equivalent to Tyr199 and Tyr309 of human PANX1), resulting in channel opening and ATP release. Although the Src-dependent PANX1 activation mechanism has been widely discussed in the literature, independent validation of the tyrosine phosphorylation of PANX1 has been lacking. Here, we show that commercially available antibodies against the two phosphorylation sites mentioned above-which were used to identify endogenous PANX1 phosphorylation at these two sites-are nonspecific and should not be used to interpret results related to PANX1 phosphorylation. We further provide evidence that neither tyrosine residue is a major phosphorylation site for Src kinase in heterologous expression systems. We call on the field to re-examine the existing paradigm of tyrosine phosphorylation-dependent activation of the PANX1 channel.


Subject(s)
Connexins , Nerve Tissue Proteins , src-Family Kinases , Phosphorylation , Connexins/metabolism , Connexins/genetics , Humans , src-Family Kinases/metabolism , src-Family Kinases/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Tyrosine/metabolism , Animals , HEK293 Cells , Mice
11.
Cell Biochem Funct ; 42(4): e4039, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38751189

ABSTRACT

Platelet hyperreactivity contributes to the pathogenesis of COVID-19, which is associated with a hypercoagulability state and thrombosis disorder. It has been demonstrated that Vitamin D deficiency is associated with the severity of COVID-19 infection. Vitamin D supplement is widely used as a dietary supplement due to its safety and health benefits. In this study, we investigated the direct effects and underlying mechanisms of 1,25(OH)2D3 on platelet hyperreactivity induced by SRAS-CoV-2 spike protein via Western blot and platelet functional studies in vitro. Firstly, we found that 1,25(OH)2D3 attenuated platelet aggregation and Src-mediated signaling. We further observed that 1,25(OH)2D3 attenuated spike protein-potentiated platelet aggregation in vitro. Mechanistically, 1,25(OH)2D3 attenuated spike protein upregulated-integrin αIIbß3 outside-in signaling such as platelet spreading and the phosphorylation of ß3, c-Src and Syk. Moreover, using PP2, the Src family kinase inhibitor to abolish spike protein-stimulated platelet aggregation and integrin αIIbß3 outside-in signaling, the combination of PP2 and 1,25(OH)2D3 did not show additive inhibitory effects on spike protein-potentiated platelet aggregation and the phosphorylation of ß3, c-Src and Syk. Thus, our data suggest that 1,25(OH)2D3 attenuates platelet aggregation potentiated by spike protein via downregulating integrin αIIbß3 outside-in signaling.


Subject(s)
Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex , Signal Transduction , Spike Glycoprotein, Coronavirus , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Spike Glycoprotein, Coronavirus/metabolism , Humans , Signal Transduction/drug effects , SARS-CoV-2/drug effects , COVID-19/metabolism , Blood Platelets/metabolism , Blood Platelets/drug effects , Calcitriol/pharmacology , src-Family Kinases/metabolism , src-Family Kinases/antagonists & inhibitors , Syk Kinase/metabolism , Syk Kinase/antagonists & inhibitors , Phosphorylation/drug effects , COVID-19 Drug Treatment
12.
J Thromb Haemost ; 22(6): 1749-1757, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38811291

ABSTRACT

BACKGROUND: An iron overload status induces ferroptosis, an iron-dependent nonapoptotic cell death, in various pathological conditions. We previously reported that hemin (heme), protoporphyrin-IX with ferric iron, activates platelets via C-type lectin-like receptor-2 (CLEC-2) and glycoprotein VI/FcRγ, but protoporphyrin-IX alone blocks CLEC-2-dependent platelet activation. Therefore, we hypothesized that free iron has the ability to activate platelets. OBJECTIVES: This study aimed to elucidate platelet activation mechanisms of iron (ferric chloride), including the identification of signaling pathways and receptors, and to examine whether platelets regulate ferroptosis. METHODS: Platelet aggregometry, platelet activation marker expression, and protein phosphorylation were examined in ferric chloride-stimulated human and murine platelets. Inhibitors of platelet activation signaling pathways and receptor-deleted platelets were utilized to identify the responsible signaling pathway and receptor. The effect of platelets on ferroptosis of endothelial cells was investigated in vitro. RESULTS: Ferric chloride induced platelet activation dependent on Src family kinase pathways in humans and mice. Ferric chloride-induced platelet aggregation was almost lost in CLEC-2-depleted murine platelets and wild-type platelets preincubated with recombinant CLEC-2 proteins. Furthermore, coculture of wild-type platelets, but not CLEC-2-deficient platelets, attenuated ferroptosis of endothelial cells in vitro. CONCLUSION: Ferric chloride activates platelets via CLEC-2 and Src family kinase pathways, and platelets have a protective role in the ferroptosis of endothelial cells dependent on CLEC-2.


Subject(s)
Blood Platelets , Chlorides , Ferric Compounds , Ferroptosis , Lectins, C-Type , Mice, Inbred C57BL , Platelet Activation , Platelet Aggregation , Signal Transduction , Animals , Blood Platelets/metabolism , Blood Platelets/drug effects , Ferric Compounds/pharmacology , Humans , Platelet Activation/drug effects , Lectins, C-Type/metabolism , Chlorides/metabolism , Platelet Aggregation/drug effects , Ferroptosis/drug effects , src-Family Kinases/metabolism , Phosphorylation , Mice, Knockout , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Mice , Human Umbilical Vein Endothelial Cells/metabolism
13.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 644-651, 2024 Apr 20.
Article in Chinese | MEDLINE | ID: mdl-38708496

ABSTRACT

OBJECTIVE: To observe the effect of Shenqi Chongcao (SQCC) Formula on the ASS1/src/STAT3 signaling pathway in a rat model of lung fibrosis and explore its therapeutic mechanism. METHODS: A total of 120 male SD rats were divided equally into 5 groups, including a blank control group with saline treatment and 4 groups of rat models of idiopathic pulmonary fibrosis induced by intratracheal instillation of bleomycin. One day after modeling, the rat models were treated with daily gavage of 10 mL/kg saline, SQCC decoction (0.423 g/kg), pirfenidone (10 mL/kg), or intraperitoneal injection of arginine deiminase (ADI; 2.25 mg/kg, every 3 days) for 28 days. After the treatments, the lung tissues of the rats were collected for calculating the lung/body weight ratio, observing histopathology using HE and Masson staining, and analyzing the inflammatory cells in BALF using Giemsa staining. Serum chemokine ligand 2 (CCL2) and transforming growth factor-ß1 (TGF-ß1) levels were measured with ELISA. The protein expressions of src, p-srcTry529, STAT3, and p-STAT3Try705 and the mRNA expressions of ASS1, src and STAT3 in the lung tissues were detected using Western blotting and RT-qPCR. RESULTS: The neutrophil, macrophage and lymphocyte counts and serum levels of CCL2 and TGF-ß1 were significantly lower in SQCC, pirfenidone and ADI treatment groups than in the model group at each time point of measurement (P < 0.05). P-srcTry529 and p-STAT3Try705 protein expression levels and ASS1, src, and STAT3 mRNA in the lung tissues were also significantly lower in the 3 treatment groups than in the model group (P < 0.05). CONCLUSION: SQCC Formula can alleviate lung fibrosis in rats possibly by activating the ASS1/src/STAT3 signaling pathway in the lung tissues.


Subject(s)
Drugs, Chinese Herbal , Pulmonary Fibrosis , STAT3 Transcription Factor , Signal Transduction , Animals , Male , Rats , Bleomycin , Chemokine CCL2/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/pharmacology , Inflammation/metabolism , Inflammation/drug therapy , Lung/metabolism , Lung/pathology , Lung/drug effects , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , src-Family Kinases/drug effects , src-Family Kinases/metabolism , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , Transforming Growth Factor beta1/metabolism , Carbon-Carbon Ligases/drug effects , Carbon-Carbon Ligases/metabolism
14.
Dis Model Mech ; 17(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38691000

ABSTRACT

Mechanical stimulation as a mimic of drusen formation in the eye increases the expression of angiogenic factors in retinal pigment epithelial (RPE) cells, but the underlying molecular mechanisms remain unclear. We investigated and characterized the effects of mechanical stimulation on the expression of angiogenic factors in RPE cells both in vitro and in a mouse model. Mechanical stimulation increased the expression of vascular endothelial growth factor (VEGF, encoded by VEGFA) and other angiogenesis-related genes in cultured RPE1 cells. The presence of hypoxia-inducible factor 1α (HIF-1α, encoded by HIF1A) was also increased, and both knockdown of HIF-1α and treatment with the HIF-1α inhibitor CAY10585 attenuated the effect of mechanical stimulation on angiogenesis factor gene expression. Signaling by the tyrosine kinase SRC and p38 mitogen-activated protein kinase was involved in HIF-1α activation and consequent angiogenesis-related gene expression induced by mechanical stimulation. Our results suggest that SRC-p38 and HIF-1α signaling are involved in the upregulation of angiogenic factors in RPE cells by mechanical stimulation. Such in vivo suppression of upregulated expression of angiogenesis-related genes by pharmacological inhibitors of HIF-1α suggests a new potential approach to the treatment of age-related macular degeneration.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Mice, Inbred C57BL , Retinal Pigment Epithelium , Up-Regulation , p38 Mitogen-Activated Protein Kinases , src-Family Kinases , Retinal Pigment Epithelium/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Animals , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Stress, Mechanical , Signal Transduction , Mice , Cell Line , Angiogenesis Inducing Agents/metabolism , Epithelial Cells/metabolism , Humans
15.
Protein Sci ; 33(6): e5023, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38801214

ABSTRACT

Oncogenic mutations can destabilize signaling proteins, resulting in increased or unregulated activity. Thus, there is considerable interest in mapping the relationship between mutations and the stability of signaling proteins, to better understand the consequences of oncogenic mutations and potentially inform the development of new therapeutics. Here, we develop a tool to study protein-kinase stability in live mammalian cells and the effects of the HSP90 chaperone system on the stability of these kinases. We determine the expression levels of protein kinases by monitoring the fluorescence of fluorescent proteins fused to those kinases, normalized to that of co-expressed reference fluorescent proteins. We used this tool to study the dependence of Src- and Raf-family kinases on the HSP90 system. We demonstrate that this sensor reports on destabilization induced by oncogenic mutations in these kinases. We also show that Src-homology 2 and Src-homology 3 domains, which are required for autoinhibition of Src-family kinases, stabilize these kinase domains in the cell. Our expression-calibrated sensor enables the facile characterization of the effects of mutations and small-molecule drugs on protein-kinase stability.


Subject(s)
HSP90 Heat-Shock Proteins , Humans , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/chemistry , src-Family Kinases/metabolism , src-Family Kinases/chemistry , src-Family Kinases/genetics , HEK293 Cells , Protein Stability , Mutation , Enzyme Stability , Fluorescence
16.
J Affect Disord ; 359: 241-252, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38768820

ABSTRACT

BACKGROUND: Postpartum depression (PPD) is a serious psychiatric disorder that has significantly adverse impacts on maternal health. Metabolic abnormalities in the brain are associated with numerous neurological disorders, yet the specific metabolic signaling pathways and brain regions involved in PPD remain unelucidated. METHODS: We performed behavioral test in the virgin and postpartum mice. We used mass spectrometry imaging (MSI) and targeted metabolomics analyses to investigate the metabolic alternation in the brain of GABAAR Delta-subunit-deficient (Gabrd-/-) postpartum mice, a specific preclinical animal model of PPD. Next, we performed mechanism studies including qPCR, Western blot, immunofluorescence staining, electron microscopy and primary astrocyte culture. In the specific knockdown and rescue experiments, we injected the adeno-associated virus into the central amygdala (CeA) of female mice. RESULTS: We identified that prostaglandin D2 (PGD2) downregulation in the CeA was the most outstanding alternation in PPD, and then validated that lipocalin-type prostaglandin D synthase (L-PGDS)/PGD2 downregulation plays a causal role in depressive behaviors derived from PPD in both wild-type and Gabrd-/- mice. Furthermore, we verified that L-PGDS/PGD2 signaling dysfunction-induced astrocytes atrophy is mediated by Src phosphorylation both in vitro and in vivo. LIMITATIONS: L-PGDS/PGD2 signaling dysfunction may be only responsible for the depressive behavior rather than maternal behaviors in the PPD, and it remains to be seen whether this mechanism is applicable to all depression types. CONCLUSION: Our study identified abnormalities in the L-PGDS/PGD2 signaling in the CeA, which inhibited Src phosphorylation and induced astrocyte atrophy, ultimately resulting in the development of PPD in mice.


Subject(s)
Astrocytes , Atrophy , Depression, Postpartum , Disease Models, Animal , Prostaglandin D2 , Signal Transduction , Animals , Astrocytes/pathology , Astrocytes/metabolism , Female , Depression, Postpartum/pathology , Depression, Postpartum/metabolism , Mice , Signal Transduction/physiology , Prostaglandin D2/metabolism , Central Amygdaloid Nucleus/metabolism , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Lipocalins/genetics , Lipocalins/metabolism , src-Family Kinases/metabolism , Mice, Knockout
17.
Cancer Cell ; 42(5): 833-849.e12, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38701792

ABSTRACT

Glucocorticoids have been used for decades to treat lymphomas without an established mechanism of action. Using functional genomic, proteomic, and chemical screens, we discover that glucocorticoids inhibit oncogenic signaling by the B cell receptor (BCR), a recurrent feature of aggressive B cell malignancies, including diffuse large B cell lymphoma and Burkitt lymphoma. Glucocorticoids induce the glucocorticoid receptor (GR) to directly transactivate genes encoding negative regulators of BCR stability (LAPTM5; KLHL14) and the PI3 kinase pathway (INPP5D; DDIT4). GR directly represses transcription of CSK, a kinase that limits the activity of BCR-proximal Src-family kinases. CSK inhibition attenuates the constitutive BCR signaling of lymphomas by hyperactivating Src-family kinases, triggering their ubiquitination and degradation. With the knowledge that glucocorticoids disable oncogenic BCR signaling, they can now be deployed rationally to treat BCR-dependent aggressive lymphomas and used to construct mechanistically sound combination regimens with inhibitors of BTK, PI3 kinase, BCL2, and CSK.


Subject(s)
Glucocorticoids , Receptors, Antigen, B-Cell , Humans , Glucocorticoids/pharmacology , Receptors, Antigen, B-Cell/metabolism , Animals , Signal Transduction/drug effects , Receptors, Glucocorticoid/metabolism , Mice , Cell Line, Tumor , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Molecular Targeted Therapy/methods , Phosphatidylinositol 3-Kinases/metabolism , src-Family Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects
18.
Med Oncol ; 41(6): 156, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750377

ABSTRACT

This study explores the therapeutic potential of phytochemicals derived from Morus alba for colorectal cancer (CRC) treatment. Colorectal cancer is a global health concern with increasing mortality rates, necessitating innovative strategies for prevention and therapy. Employing in silico analysis, molecular docking techniques (MDT), and molecular dynamics simulations (MDS), the study investigates the interactions between Morus alba-derived phytochemicals and key proteins (AKT1, Src, STAT3, EGFR) implicated in CRC progression. ADME/T analysis screens 78 phytochemicals for drug-like and pharmacokinetic properties. The study integrates Lipinski's Rule of Five and comprehensive bioactivity assessments, providing a nuanced understanding of Morus alba phytoconstituent's potential as CRC therapeutic agents. Notably, 14 phytochemicals out of 78 emerge as potential candidates, demonstrating oral bioavailability and favorable bioactivity scores. Autodock 1.5.7 is employed for energy minimization followed by molecular docking with the highest binding energy observed to be - 11.7 kcal/mol exhibited by Kuwanon A against AKT1. Molecular dynamics simulations and trajectory path analysis were conducted between Kuwanon A and AKT1 at the Pleckstrin homology (PH) domain region (TRP80), revealing minimal deviations. In comparison to the standard drug Capivasertib, the phytochemical Kuwanon A emerges as a standout candidate based on computational analysis. This suggests its potential as an alternative to mitigate the limitations associated with the standard drug. The research aims to provide insights for future experimental validations and to stimulate the development of Kuwanon A as a novel, effective therapeutic agent for managing colorectal cancer.


Subject(s)
Colorectal Neoplasms , Molecular Docking Simulation , Molecular Dynamics Simulation , Morus , Phytochemicals , Morus/chemistry , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Phytochemicals/chemistry , Phytochemicals/pharmacology , Phytochemicals/pharmacokinetics , Humans , Proto-Oncogene Proteins c-akt/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , STAT3 Transcription Factor/metabolism , Plant Extracts/chemistry , Plant Extracts/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/chemistry , src-Family Kinases/metabolism
19.
J Virol ; 98(6): e0170523, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38742902

ABSTRACT

Long non-coding RNAs (lncRNAs) represent a new group of host factors involved in viral infection. Current study identified an intergenic lncRNA, LINC08148, as a proviral factor of Zika virus (ZIKV) and Dengue virus 2 (DENV2). Knockout (KO) or silencing of LINC08148 decreases the replication of ZIKV and DENV2. LINC08148 mainly acts at the endocytosis step of ZIKV but at a later stage of DENV2. RNA-seq analysis reveals that LINC08148 knockout downregulates the transcription levels of five endocytosis-related genes including AP2B1, CHMP4C, DNM1, FCHO1, and Src. Among them, loss of Src significantly decreases the uptake of ZIKV. Trans-complementation of Src in the LINC08148KO cells largely restores the caveola-mediated endocytosis of ZIKV, indicating that the proviral effect of LINC08148 is exerted through Src. Finally, LINC08148 upregulates the Src transcription through associating with its transcription factor SP1. This work establishes an essential role of LINC08148 in the ZIKV entry, underscoring a significance of lncRNAs in the viral infection. IMPORTANCE: Long non-coding RNAs (lncRNAs), like proteins, participate in viral infection. However, functions of most lncRNAs remain unknown. In this study, we performed a functional screen based on microarray data and identified a new proviral lncRNA, LINC08148. Then, we uncovered that LINC08148 is involved in the caveola-mediated endocytosis of ZIKV, rather than the classical clathrin-mediated endocytosis. Mechanistically, LINC08148 upregulates the transcription of Src, an initiator of caveola-mediated endocytosis, through binding to its transcription factor SP1. This study identifies a new lncRNA involved in the ZIKV infection, suggesting lncRNAs and cellular proteins are closely linked and cooperate to regulate viral infection.


Subject(s)
Endocytosis , RNA, Long Noncoding , Virus Internalization , Zika Virus Infection , Zika Virus , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics , Zika Virus/genetics , Zika Virus/physiology , Humans , Zika Virus Infection/virology , Zika Virus Infection/metabolism , Zika Virus Infection/genetics , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Caveolae/metabolism , Animals , Virus Replication , Up-Regulation , Dengue Virus/physiology , Dengue Virus/genetics , Chlorocebus aethiops , HEK293 Cells , Vero Cells , src-Family Kinases/metabolism , src-Family Kinases/genetics
20.
Sci Transl Med ; 16(747): eadj7685, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38748774

ABSTRACT

Intrahepatic cholangiocarcinoma (ICC) is an aggressive bile duct malignancy that frequently exhibits isocitrate dehydrogenase (IDH1/IDH2) mutations. Mutant IDH (IDHm) ICC is dependent on SRC kinase for growth and survival and is hypersensitive to inhibition by dasatinib, but the molecular mechanism underlying this sensitivity is unclear. We found that dasatinib reduced p70 S6 kinase (S6K) and ribosomal protein S6 (S6), leading to substantial reductions in cell size and de novo protein synthesis. Using an unbiased phosphoproteomic screen, we identified membrane-associated guanylate kinase, WW, and PDZ domain containing 1 (MAGI1) as an SRC substrate in IDHm ICC. Biochemical and functional assays further showed that SRC inhibits a latent tumor-suppressing function of the MAGI1-protein phosphatase 2A (PP2A) complex to activate S6K/S6 signaling in IDHm ICC. Inhibiting SRC led to activation and increased access of PP2A to dephosphorylate S6K, resulting in cell death. Evidence from patient tissue and cell line models revealed that both intrinsic and extrinsic resistance to dasatinib is due to increased phospho-S6 (pS6). To block pS6, we paired dasatinib with the S6K/AKT inhibitor M2698, which led to a marked reduction in pS6 in IDHm ICC cell lines and patient-derived organoids in vitro and substantial growth inhibition in ICC patient-derived xenografts in vivo. Together, these results elucidated the mechanism of action of dasatinib in IDHm ICC, revealed a signaling complex regulating S6K phosphorylation independent of mTOR, suggested markers for dasatinib sensitivity, and described a combination therapy for IDHm ICC that may be actionable in the clinic.


Subject(s)
Adaptor Proteins, Signal Transducing , Cholangiocarcinoma , Dasatinib , Isocitrate Dehydrogenase , Mutation , src-Family Kinases , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/pathology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/genetics , Humans , Dasatinib/pharmacology , Mutation/genetics , src-Family Kinases/metabolism , src-Family Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Isocitrate Dehydrogenase/metabolism , Isocitrate Dehydrogenase/genetics , Animals , Cell Adhesion Molecules/metabolism , Cell Proliferation/drug effects , Phosphorylation/drug effects , Signal Transduction/drug effects , Mice , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/drug therapy , Ribosomal Protein S6 Kinases, 70-kDa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...