Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 519
Filter
1.
Cells ; 11(21)2022 10 27.
Article in English | MEDLINE | ID: mdl-36359784

ABSTRACT

BACKGROUND: Rhabdomyolysis is the collapse of damaged skeletal muscle and the leakage of muscle-cell contents, such as electrolytes, myoglobin, and other sarcoplasmic proteins, into the circulation. The glomeruli filtered these products, leading to acute kidney injury (AKI) through several mechanisms, such as intratubular obstruction secondary to protein precipitation. The prognosis is highly mutable and depends on the underlying complications and etiologies. New therapeutic plans to reduce AKI are now needed. Up to now, several cellular pathways, with the nuclear factor kappa beta (NF-kB), as well as the proinflammatory effects on epithelial and tubular epithelial cells, have been recognized as the major pathway for the initiation of the matrix-producing cells in AKI. Recently, it has been mentioned that periostin (POSTN), an extracellular matrix protein, is involved in the development of inflammation through the modulation of the NF-kB pathway. However, how POSTN develops the inflammation protection in AKI by rhabdomyolysis is uncertain. This study aimed to investigate the role of POSTN in a rhabdomyolysis mice model of AKI induced by an intramuscular injection of 50% glycerol. METHODS: In vivo, we performed an intramuscular injection of 50% glycerol (5 mg/kg body weight) to make rhabdomyolysis-induced AKI. We examined the expression level of POSTN through the progression of AKI after glycerol intramuscular injection for C57BL/6J wildtype (WT) mice. We sacrificed mice at 72 h after glycerol injection. We made periostin-null mice to examine the role of POSTN in acute renal failure. The role of periostin was further examined through in vitro methods. The development of renal inflammation is linked with the NF-kB pathway. To examine the POSTN function, we administrated hemin (100 µM) on NIH-3T3 fibroblast cells, and the following signaling pathways were examined. RESULTS: The expression of periostin was highly increased, peaking at about 72 h after glycerol injection. The expression of inflammation-associated mRNAs such as monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-a) and IL-6, and tubular injury score in H-E staining were more reduced in POSTN-null mice than WT mice at 72 h after glycerol injection. CONCLUSION: POSTN was highly expressed in the kidney through rhabdomyolysis and was a positive regulator of AKI. Targeting POSTN might propose a new therapeutic strategy against the development of acute renal failure.


Subject(s)
Acute Kidney Injury , Cell Adhesion Molecules , Animals , Mice , Acute Kidney Injury/chemically induced , Acute Kidney Injury/complications , Acute Kidney Injury/pathology , Disease Models, Animal , Glycerol/pharmacology , Inflammation/drug therapy , Inflammation/pathology , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Rhabdomyolysis/complications , Rhabdomyolysis/chemically induced , Rhabdomyolysis/pathology , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/metabolism
2.
Pharmacol Rev ; 74(4): 1028-1048, 2022 10.
Article in English | MEDLINE | ID: mdl-36113879

ABSTRACT

Several integral membrane proteins undergo regulated intramembrane proteolysis (RIP), a tightly controlled process through which cells transmit information across and between intracellular compartments. RIP generates biologically active peptides by a series of proteolytic cleavage events carried out by two primary groups of enzymes: sheddases and intramembrane-cleaving proteases (iCLiPs). Following RIP, fragments of both pore-forming and non-pore-forming ion channel subunits, as well as immunoglobulin super family (IgSF) members, have been shown to translocate to the nucleus to function in transcriptional regulation. As an example, the voltage-gated sodium channel ß1 subunit, which is also an IgSF-cell adhesion molecule (CAM), is a substrate for RIP. ß1 RIP results in generation of a soluble intracellular domain, which can regulate gene expression in the nucleus. In this review, we discuss the proposed RIP mechanisms of voltage-gated sodium, potassium, and calcium channel subunits as well as the roles of their generated proteolytic products in the nucleus. We also discuss other RIP substrates that are cleaved by similar sheddases and iCLiPs, such as IgSF macromolecules, including CAMs, whose proteolytically generated fragments function in the nucleus. Importantly, dysfunctional RIP mechanisms are linked to human disease. Thus, we will also review how understanding RIP events and subsequent signaling processes involving ion channel subunits and IgSF proteins may lead to the discovery of novel therapeutic targets. SIGNIFICANCE STATEMENT: Several ion channel subunits and immunoglobulin superfamily molecules have been identified as substrates of regulated intramembrane proteolysis (RIP). This signal transduction mechanism, which generates polypeptide fragments that translocate to the nucleus, is an important regulator of gene transcription. RIP may impact diseases of excitability, including epilepsy, cardiac arrhythmia, and sudden death syndromes. A thorough understanding of the role of RIP in gene regulation is critical as it may reveal novel therapeutic strategies for the treatment of previously intractable diseases.


Subject(s)
Cell Adhesion Molecules , Ion Channels , Proteolysis , Calcium Channels/metabolism , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/metabolism , Humans , Ion Channels/drug effects , Ion Channels/metabolism , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Peptide Hydrolases/metabolism , Peptides/metabolism , Potassium/metabolism , Potassium Channels, Voltage-Gated , Proteolysis/drug effects , Sodium/metabolism
3.
Anticancer Drugs ; 33(1): e235-e246, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34419964

ABSTRACT

One cost-effective way for identifying novel cancer therapeutics is in the repositioning of available drugs for which current therapies are inadequate. Levofloxacin prevents DNA duplication in bacteria by inhibiting the activity of DNA helicase. As eukaryotic cells have similar intracellular biologic characteristics as prokaryotic cells, we speculate that antibiotics inhibiting DNA duplication in bacteria may also affect the survival of cancer cells. Here we report that levofloxacin significantly inhibited the proliferation and clone formation of cancer cells and xenograft tumor growth through cell cycle arrest at G2/M and by enhancing apoptosis. Levofloxacin significantly altered gene expression in a direction favoring anticancer activity. THBS1 and LAPTM5 were dose-dependently upregulated whereas SRD5A3, MFAP5 and P4HA1 were downregulated. Pathway analysis revealed that levofloxacin significantly regulated canonical oncogenic pathways. Specific network enrichment included a MAPK/apoptosis/cytokine-cytokine receptor interaction pathway network that associates with cell growth, differentiation, cell death, angiogenesis and development and repair processes and a bladder cancer/P53 signaling pathway network mediating the inhibition of angiogenesis and metastasis. THBS1 overlapped in 16 of the 22 enriched apoptotic pathways and the 2 pathways in the bladder cancer/P53 signaling pathway network. P4HA1 enriched in 7 of the top 10 molecular functions regulated by differential downregulated genes. Our results indicate that levofloxacin has broad-spectrum anticancer activity with the potential to benefit cancer patients already treated or requiring prophylaxis for an infectious syndrome. The efficacy we find with levofloxacin may provide insight into the discovery and the design of novel less toxic anticancer drugs.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Levofloxacin/pharmacology , Animals , Apoptosis/drug effects , Cell Adhesion Molecules/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytokines/drug effects , DNA Helicases/drug effects , Dose-Response Relationship, Drug , Humans , Male , Membrane Proteins/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinases/drug effects , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
4.
Mol Med Rep ; 24(4)2021 Oct.
Article in English | MEDLINE | ID: mdl-34396446

ABSTRACT

Recruitment of lymphocytes to the vascular wall contributes to the pathogenesis of atherosclerosis (AS). The expression of cellular adhesion molecules, such as vascular cell adhesion molecule­1 and intercellular adhesion molecule­1, serves a critical role in mediating lymphocyte adhesion to the vascular wall. Cholesterol loading induces the expression of adhesion molecules in vascular smooth muscle cells (VSMCs), but the underlying mechanism is not completely understood. The present study aimed to investigate the mechanism underlying the effects of cholesterol on adhesion molecule expression, and whether metformin protected VSMCs against cholesterol­induced functional alterations. Human VSMCs were loaded with cholesterol and different concentrations of metformin. The expression levels of adhesion molecules were assessed via reverse transcription­quantitative PCR and western blotting. Reactive oxygen species (ROS) accumulation and levels were quantified via fluorescence assays and spectrophotometry, respectively. AMP­activated protein kinase (AMPK), p38 MAPK and NF­κB signaling pathway­related protein expression levels were evaluated via western blotting. Compared with the control group, cholesterol loading significantly upregulated adhesion molecule expression levels on VSMCs by increasing intracellular ROS levels and activating the p38 MAPK and NF­κB signaling pathways. Metformin decreased cholesterol­induced VSMC damage by activating the AMPK signaling pathway, and suppressing p38 MAPK and NF­κB signaling. The present study indicated the therapeutic potential of metformin for VSMC protection, reduction of monocyte adhesion, and ultimately, the prevention and treatment of AS.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Adhesion Molecules/drug effects , Cholesterol/adverse effects , Metformin/pharmacology , Muscle, Smooth, Vascular/metabolism , Signal Transduction/drug effects , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Monocytes/metabolism , Myocytes, Smooth Muscle/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor RelA , Vascular Cell Adhesion Molecule-1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Molecules ; 26(11)2021 May 25.
Article in English | MEDLINE | ID: mdl-34070457

ABSTRACT

Cisplatin and derivatives are highly effective in the treatment of a wide range of cancer types; however, these metallodrugs display low selectivity, leading to severe side effects. Additionally, their administration often results in the development of chemoresistance, which ultimately results in therapeutic failure. This scenario triggered the study of other transition metals with innovative pharmacological profiles as alternatives to platinum, ruthenium- (e.g., KP1339 and NAMI-A) and gold-based (e.g., Auranofin) complexes being among the most advanced in terms of clinical evaluation. Concerning the importance of improving the in vivo selectivity of metal complexes and the current relevance of ruthenium and gold metals, this review article aims to survey the main research efforts made in the past few years toward the design and biological evaluation of target-specific ruthenium and gold complexes. Herein, we give an overview of the inorganic and organometallic molecules conjugated to different biomolecules for targeting membrane proteins, namely cell adhesion molecules, G-protein coupled receptors, and growth factor receptors. Complexes that recognize the progesterone receptors or other targets involved in metabolic pathways such as glucose transporters are discussed as well. Finally, we describe some complexes aimed at recognizing cell organelles or compartments, mitochondria being the most explored. The few complexes addressing targeted gene therapy are also presented and discussed.


Subject(s)
Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Gold Compounds/pharmacology , Ruthenium Compounds/pharmacology , Antineoplastic Agents/administration & dosage , Cell Adhesion Molecules/drug effects , Cell Line, Tumor , Coordination Complexes/administration & dosage , Gold Compounds/administration & dosage , Humans , Receptors, G-Protein-Coupled/drug effects , Receptors, Growth Factor/drug effects , Ruthenium Compounds/administration & dosage
6.
Clin Exp Allergy ; 51(7): 915-931, 2021 07.
Article in English | MEDLINE | ID: mdl-34037993

ABSTRACT

BACKGROUND: Type 2 inflammation is common in numerous atopic/allergic diseases and can be identified by elevated biomarker levels. Dupilumab, a fully human monoclonal antibody, blocks the shared receptor component for interleukin-4 and interleukin-13, key and central drivers of type 2 inflammation. OBJECTIVE: Assessment of dupilumab effect on type 2 inflammatory biomarkers in atopic dermatitis (AD), asthma, chronic rhinosinusitis with nasal polyps (CRSwNP) and eosinophilic esophagitis (EoE). METHODS: Data were extracted from three randomized placebo-controlled trials of dupilumab in AD (NCT02277743, N = 671; NCT02277769, N = 708; NCT02260986, N = 740); and one each in asthma (NCT02414854, N = 1902); CRSwNP (NCT02898454, N = 448); and EoE (NCT02379052, N = 47). Biomarkers assessed were serum thymus and activation-regulated chemokine (TARC), plasma eotaxin-3, serum total immunoglobulin E (IgE), serum periostin and blood eosinophil count. RESULTS: Dupilumab versus placebo significantly suppressed most type 2 inflammatory biomarker levels across all studies/indications where data were assessed. Reductions in serum TARC, plasma eotaxin-3 and serum periostin occurred rapidly, whereas reductions in serum total IgE were more gradual. Across diseases, at the end of treatment, median percentage change from baseline in TARC levels ranged from -24.8% to -88.6% (placebo +2.6% to -53.6%); -38.2% to -51.5% (placebo +8.3% to -0.16%) in eotaxin-3; -24.8% to -76.7% (placebo +8.3% to -4.4%) in total IgE; and -13.6% to -41.1% (placebo +10.1% to -6.94%) in periostin levels. Blood eosinophil responses to dupilumab varied by disease, with minimal changes in AD in the SOLO studies (median percentage change from baseline to end of treatment: 0% [95% CI: -15.8, 0]); transient increases followed by decreases to below-baseline levels in asthma (-14.6% [-20.0, -7.7]) and CRSwNP (-29.4% [-40.0, -16.3]); and significant decreases in EoE (-50.0% [-50.0, -33.3]). CONCLUSION AND CLINICAL RELEVANCE: Dupilumab reduced levels of type 2 biomarkers across clinical studies in patients with AD, asthma, CRSwNP and EoE.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Biomarkers/blood , Hypersensitivity, Immediate/drug therapy , Hypersensitivity, Immediate/immunology , Cell Adhesion Molecules/blood , Cell Adhesion Molecules/drug effects , Chemokine CCL17/blood , Chemokine CCL17/drug effects , Chemokine CCL26/blood , Chemokine CCL26/drug effects , Eosinophils/drug effects , Humans , Immunoglobulin E/blood , Immunoglobulin E/drug effects , Inflammation/drug therapy , Inflammation/immunology , Randomized Controlled Trials as Topic
7.
Behav Brain Res ; 410: 113353, 2021 07 23.
Article in English | MEDLINE | ID: mdl-33979656

ABSTRACT

Selective serotonin reuptake inhibitors (SSRIs) are the most widely used treatment by women experiencing depression during pregnancy. However, the effects of maternal SSRI use on early offspring development remain poorly understood. Recent studies suggest that SSRIs can modify the gut microbiota and interact directly with particular gut bacteria, raising the question of whether the gut microbiome impacts host responses to SSRIs. In this study, we investigate effects of prenatal SSRI exposure on fetal neurodevelopment and further evaluate potential modulatory influences of the maternal gut microbiome. We demonstrate that maternal treatment with the SSRI fluoxetine induces widespread alterations in the fetal brain transcriptome during midgestation, including increases in the expression of genes relevant to synaptic organization and neuronal signaling and decreases in the expression of genes related to DNA replication and mitosis. Notably, maternal fluoxetine treatment from E7.5 to E14.5 has no overt effects on the composition of the maternal gut microbiota. However, maternal pretreatment with antibiotics to deplete the gut microbiome substantially modifies transcriptional responses of the fetal brain to maternal fluoxetine treatment. In particular, maternal fluoxetine treatment elevates localized expression of the opioid binding protein/cell adhesion molecule like gene Opcml in the fetal thalamus and lateral ganglionic eminence, which is prevented by maternal antibiotic treatment. Together, these findings reveal that maternal fluoxetine treatment alters gene expression in the fetal brain through pathways that are impacted, at least in part, by the presence of the maternal gut microbiota.


Subject(s)
Brain/drug effects , Cell Adhesion Molecules/drug effects , Embryo, Mammalian/drug effects , Fluoxetine/pharmacology , Gastrointestinal Microbiome/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Selective Serotonin Reuptake Inhibitors/pharmacology , Transcriptome/drug effects , Animals , Disease Models, Animal , Female , Fluoxetine/administration & dosage , GPI-Linked Proteins/drug effects , Male , Mice , Mice, Inbred C57BL , Pregnancy , Selective Serotonin Reuptake Inhibitors/administration & dosage
8.
Eur J Pharmacol ; 900: 174038, 2021 Jun 05.
Article in English | MEDLINE | ID: mdl-33737008

ABSTRACT

Subarachnoid hemorrhage (SAH) due to rupture of an intracranial aneurysm leads to vasospasm resulting in delayed cerebral ischemia. Therapeutic options are currently limited to hemodynamic optimization and nimodipine, which have marginal clinical efficacy. Nitric oxide (NO) modulates cerebral blood flow through activation of the cGMP-Protein Kinase G (PKG) pathway. Our hypothesis is that SAH results in downregulation of signaling components in the NO-PKG pathway which could explain why treatments for vasospasm targeting this pathway lack efficacy and that treatment with a cell permeant phosphopeptide mimetic of downstream effector prevents delayed vasospasm after SAH. Using a rat endovascular perforation model, reduced levels of NO-PKG pathway molecules were confirmed. Additionally, it was determined that expression and phosphorylation of a PKG substrate: Vasodilator-stimulated phosphoprotein (VASP) was downregulated. A family of cell permeant phosphomimetic of VASP (VP) was wasdesigned and shown to have vasorelaxing property that is synergistic with nimodipine in intact vascular tissuesex vivo. Hence, treatment targeting the downstream effector of the NO signaling pathway, VASP, may bypass receptors and signaling elements leading to vasorelaxation and that treatment with VP can be explored as a therapeutic strategy for SAH induced vasospasm and ameliorate neurological deficits.


Subject(s)
Phosphopeptides/therapeutic use , Subarachnoid Hemorrhage/drug therapy , Vasodilator Agents/therapeutic use , Vasospasm, Intracranial/drug therapy , Animals , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/metabolism , Cyclic GMP-Dependent Protein Kinases/drug effects , Down-Regulation , Drug Design , Drug Synergism , Microfilament Proteins/drug effects , Microfilament Proteins/metabolism , Molecular Mimicry , Nimodipine/pharmacology , Nitric Oxide/metabolism , Phosphopeptides/pharmacokinetics , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Subarachnoid Hemorrhage/metabolism , Swine , Vasodilator Agents/pharmacokinetics
9.
Methods Mol Biol ; 2201: 163-169, 2021.
Article in English | MEDLINE | ID: mdl-32975797

ABSTRACT

Opioids play a pivotal role in pain transmission but are also able to modulate immune cell functions. In the last decades a connection between opioids and integrins-adhesion molecules involved, among many other processes, in leukocyte recruitment at inflamed site-has been established. To study immune cell integrin-mediated adhesion, cell adhesion assay is a simple, reproducible, and valuable tool capable of unraveling concentration-dependent effects of a test candidate on integrin-mediated cell adhesion.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Adhesion/drug effects , Immunohistochemistry/methods , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacology , Animals , Cell Adhesion/physiology , Cell Adhesion Molecules/drug effects , Humans , Immunologic Factors/pharmacology , Inflammation/metabolism , Integrins/drug effects , Integrins/metabolism , Jurkat Cells , Leukocytes/metabolism , U937 Cells
10.
Cancer Gene Ther ; 28(1-2): 18-26, 2021 02.
Article in English | MEDLINE | ID: mdl-32595215

ABSTRACT

OPCML is a highly conserved glycosyl phosphatidylinositol (GPI)-anchored protein belonging to the IgLON family of cell adhesion molecules. OPCML functions as a tumor suppressor and is silenced in over 80% of ovarian cancers by loss of heterozygosity and by epigenetic mechanisms. OPCML inactivation is also observed in many other cancers suggesting a conservation of tumor suppressor function. Although epigenetic silencing and subsequent loss of OPCML expression correlate with poor progression-free and overall patient survival, its mechanism of action is only starting to be fully elucidated. Recent discoveries have demonstrated that OPCML exerts its tumor suppressor effect by inhibiting several cancer hallmark phenotypes in vitro and abrogating tumorigenesis in vivo, by downregulating/inactivating a specific spectrum of Receptor Tyrosine Kinases (RTKs), including EphA2, FGFR1, FGFR3, HER2, HER4, and AXL. This modulation of RTKs can also sensitize ovarian and breast cancers to lapatinib, erlotinib, and anti-AXL therapies. Furthermore, OPCML has also been shown to function in synergy with the tumor suppressor phosphatase PTPRG to inactivate pro-metastatic RTKs such as AXL. Recently, the identification of inactivating point mutations and the elucidation of the crystal structure of OPCML have provided valuable insights into its structure-function relationships, giving rise to its potential as an anti-cancer therapeutic.


Subject(s)
Cell Adhesion Molecules/drug effects , Glycosylphosphatidylinositols/therapeutic use , Neoplasms/drug therapy , GPI-Linked Proteins/drug effects , Glycosylphosphatidylinositols/pharmacology , Humans
11.
Front Immunol ; 11: 583385, 2020.
Article in English | MEDLINE | ID: mdl-33312173

ABSTRACT

Type 1 diabetesmellitus (T1D) is caused by partial destruction of the insulin-producing beta cells in the pancreas and is a major issue for public health care worldwide. Reduced or impaired immunological responses, which render patients more susceptible to infections, have been observed in T1D, and this dysfunction is often related to a lack of insulin in the blood. Paracoccidioidomycosis is an important systemic mycosis endemic in Latin America. To evaluate the effects of T1D on this fungal infection and the modulatory effects of insulin, we induced diabetes in C57Bl/6 male mice (alloxan, 60 mg/kg), infected the mice (Pb18, 1 x 106 cells), and treated the mice with neutral protamine Hagedorn (NPH) insulin (2 IU/600 mg/dL blood glucose). Twenty-four hours after infection, infected diabetic mice showed reduced secretion of interferon (IFN)-γ and interleukine (IL)-12 p70 compared to infected nondiabetic controls. On the 45th day of infection, infected diabetic mice presented higher IFN-γ levels, a higher tumor necrosis factor (TNF)-α:IL-10 ratio, and lower adhesion molecule expression levels than nondiabetic mice. In the in vitro experiments, alveolar macrophages from diabetic animals showed reduced phagocytic activity compared to those from control animals at 4, 12, and 24 h. In infected diabetic mice, treatment with insulin restored IL-12 p70 levels at 24 h of infection, reduced IFN-γ levels and the TNF-α:IL-10 ratio at 45 days, and restored vascular cell adhesion molecule (VCAM)-1 expression in pulmonary blood vessels, and this treatment reduced the diminished phosphorylation of extracellular signal-regulated kinases (ERK) and increased nuclear factor-kappa-B(iκb)-α and jun amino-terminal kinases (JNK) p46 levels in infected nondiabetic mice. In addition, insulin promoted increased phagocytic activity in the alveolar macrophages of diabetic mice. These data suggest that T1D mice are more susceptible to Pb18 infection and that insulin modulates this inflammation in diabetic mice by augmenting the expression of adhesion molecules and leukocytes in the lungs and by reducing chronic inflammation.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Insulin/pharmacology , Lung/drug effects , Paracoccidioidomycosis/immunology , Animals , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/immunology , Cytokines/drug effects , Cytokines/immunology , Diabetes Mellitus, Type 1/complications , Leukocytes/drug effects , Leukocytes/immunology , Lung/immunology , Male , Mice , Mice, Inbred C57BL
12.
Neurobiol Dis ; 146: 105131, 2020 12.
Article in English | MEDLINE | ID: mdl-33053430

ABSTRACT

As researchers across the globe have focused their attention on understanding SARS-CoV-2, the picture that is emerging is that of a virus that has serious effects on the vasculature in multiple organ systems including the cerebral vasculature. Observed effects on the central nervous system include neurological symptoms (headache, nausea, dizziness), fatal microclot formation and in rare cases encephalitis. However, our understanding of how the virus causes these mild to severe neurological symptoms and how the cerebral vasculature is impacted remains unclear. Thus, the results presented in this report explored whether deleterious outcomes from the SARS-CoV-2 viral spike protein on primary human brain microvascular endothelial cells (hBMVECs) could be observed. The spike protein, which plays a key role in receptor recognition, is formed by the S1 subunit containing a receptor binding domain (RBD) and the S2 subunit. First, using postmortem brain tissue, we show that the angiotensin converting enzyme 2 or ACE2 (a known binding target for the SARS-CoV-2 spike protein), is ubiquitously expressed throughout various vessel calibers in the frontal cortex. Moreover, ACE2 expression was upregulated in cases of hypertension and dementia. ACE2 was also detectable in primary hBMVECs maintained under cell culture conditions. Analysis of cell viability revealed that neither the S1, S2 or a truncated form of the S1 containing only the RBD had minimal effects on hBMVEC viability within a 48 h exposure window. Introduction of spike proteins to invitro models of the blood-brain barrier (BBB) showed significant changes to barrier properties. Key to our findings is the demonstration that S1 promotes loss of barrier integrity in an advanced 3D microfluidic model of the human BBB, a platform that more closely resembles the physiological conditions at this CNS interface. Evidence provided suggests that the SARS-CoV-2 spike proteins trigger a pro-inflammatory response on brain endothelial cells that may contribute to an altered state of BBB function. Together, these results are the first to show the direct impact that the SARS-CoV-2 spike protein could have on brain endothelial cells; thereby offering a plausible explanation for the neurological consequences seen in COVID-19 patients.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Blood-Brain Barrier/metabolism , Capillary Permeability/physiology , Endothelial Cells/metabolism , Inflammation/metabolism , Matrix Metalloproteinases/metabolism , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/physiology , Blood-Brain Barrier/drug effects , COVID-19 , Capillary Permeability/drug effects , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/metabolism , Cell Survival/drug effects , Dementia/metabolism , Electric Impedance , Endothelial Cells/drug effects , Frontal Lobe/metabolism , Humans , Hypertension/metabolism , In Vitro Techniques , Intercellular Junctions/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lab-On-A-Chip Devices , Matrix Metalloproteinases/drug effects , Primary Cell Culture , Protein Domains , Protein Subunits/metabolism , Protein Subunits/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Spike Glycoprotein, Coronavirus/pharmacology
13.
Pharm Biol ; 58(1): 646-654, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32658562

ABSTRACT

CONTEXT: Palmatine, a biologically active isoquinoline alkaloid, possesses multiple pharmaceutical activities against mucosal infection and inflammation. OBJECTIVE: There are no reports about the influence of palmatine on uterine mucosal epithelial cells. MATERIALS AND METHODS: We used proteomics to analyse differentially expressed proteins (DEPs) in goat endometrial epithelial cells (EECs) stimulated by lipopolysaccharide (LPS, 5 µg/mL, the dosage can induce inflammatory response, according to our previous study) for 12 h and then treated with palmatine (80 µg/mL) for 8 h; the dosage was selected based on MTT assay. The EECs without any treatment were used as controls. Every group was treated in triplicate. RESULTS: A total of 428 DEPs in LPS-stimulated group and 486 DEPs in the palmatine-treated group were identified. Functional annotation analysis showed that palmatine mainly regulated the protein expression of structural molecules involved in the response to stimuli. Pathway analysis showed that cell adhesion molecule (CaM) pathways were most significant enriched due to palmatine treatment. Junction adhesion molecule 1 (JAM1), nectin 1 (NECT1) and cadherin 5 (CDH5), which play important roles in the transepithelial migration (TEpM) of leukocytes, were significantly downregulated by palmatine. Meanwhile, other proteins essential to the maintenance of cell adhesion and those that facilitate leukocyte migration were upregulated after palmatine treatment. Discussion and conclusions: The results suggested that palmatine regulates the expression of CaMs to affect TEpM during uterine mucosal inflammation and provides novel insight to understanding and developing palmatine pharmacology. Palmatine is a promising drug for treatment of mucosal inflammation.


Subject(s)
Berberine Alkaloids/pharmacology , Endometrium/drug effects , Epithelial Cells/drug effects , Proteomics , Animals , Cell Adhesion/drug effects , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/metabolism , Cell Movement/drug effects , Cells, Cultured , Down-Regulation/drug effects , Endometrium/cytology , Epithelial Cells/metabolism , Female , Goats , Inflammation/drug therapy , Inflammation/pathology , Leukocytes/drug effects , Leukocytes/metabolism , Lipopolysaccharides , Transendothelial and Transepithelial Migration/drug effects
14.
J Cell Physiol ; 235(11): 8334-8344, 2020 11.
Article in English | MEDLINE | ID: mdl-32239671

ABSTRACT

E-cadherin, a central component of the adherens junction (AJ), is a single-pass transmembrane protein that mediates cell-cell adhesion. The loss of E-cadherin surface expression, and therefore cell-cell adhesion, leads to increased cell migration and invasion. Treatment of colorectal cancer (CRC)-derived cells (SW-480 and HT-29) with 2.0 mM metformin promoted a redistribution of cytosolic E-cadherin to de novo formed puncta along the length of the contacting membranes of these cells. Metformin also promoted translocation from the cytosol to the plasma membrane of p120-catenin, another core component of the AJs. Furthermore, E-cadherin and p120-catenin colocalized with ß-catenin at cell-cell contacts. Western blot analysis of lysates of CRC-derived cells revealed a substantial metformin-induced increase in the level of p120-catenin as well as E-cadherin phosphorylation on Ser838/840 , a modification associated with ß-catenin/E-cadherin interaction. These modifications in E-cadherin, p120-catenin and ß-catenin localization suggest that metformin induces rebuilding of AJs in CRC-derived cells. Those modifications were accompanied by the inhibition of focal adhesion kinase (FAK), as revealed by a significant decrease in the phosphorylation of FAK at Tyr397 and paxillin at Tyr118 . These changes were associated with a reduction in the numbers, but an increase in the size, of focal adhesions and by the inhibition of cell migration. Overall, these observations indicate that metformin targets multiple pathways associated with CRC development and progression.


Subject(s)
Adherens Junctions/drug effects , Cell Movement/drug effects , Colorectal Neoplasms/pathology , Focal Adhesion Kinase 1/metabolism , Metformin/pharmacology , Adherens Junctions/metabolism , Cell Adhesion/drug effects , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Down-Regulation , Focal Adhesion Kinase 1/drug effects , Humans , Protein Transport/drug effects
15.
J Pain ; 21(7-8): 892-904, 2020.
Article in English | MEDLINE | ID: mdl-31917219

ABSTRACT

NrCAM, a neuronal cell adhesion molecule in the L1 family of the immunoglobulin superfamily, is subjected to extensively alternative splicing and involved in neural development and some disorders. The aim of this study was to explore the role of Nrcam mRNA alternative splicing in neuropathic pain. A next generation RNA sequencing analysis of dorsal root ganglions (DRGs) showed the differential expression of two splicing variants of Nrcam, Nrcam+10 and Nrcam-10, in the injured DRG after the fourth lumbar spinal nerve ligation (SNL) in mice. SNL increased the exon 10 insertion, resulting in an increase in the amount of Nrcam+10 and a corresponding decrease in the level of Nrcam-10 in the injured DRG. An antisense oligonucleotide (ASO) that specifically targeted exon 10 of Nrcam gene (Nrcam ASO) repressed RNA expression of Nrcam+10 and increased RNA expression of Nrcam-10 in in vitro DRG cell culture. Either DRG microinjection or intrathecal injection of Nrcam ASO attenuated SNL-induced the development of mechanical allodynia, thermal hyperalgesia, or cold allodynia. Nrcam ASO also relieved SNL- or chronic compression of DRG (CCD)-induced the maintenance of pain hypersensitivities in male and female mice. PERSPECTIVE: We conclude that the relative levels of alternatively spliced Nrcam variants are critical for neuropathic pain genesis. Targeting Nrcam alternative splicing via the antisense oligonucleotides may be a new potential avenue in neuropathic pain management.


Subject(s)
Cell Adhesion Molecules , Ganglia, Spinal/metabolism , Hyperalgesia , Neuralgia , Oligonucleotides, Antisense/pharmacology , Alternative Splicing , Animals , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Injections, Spinal , Ligation , Male , Mice , Mice, Inbred C57BL , Neuralgia/drug therapy , Neuralgia/metabolism , Oligonucleotides, Antisense/administration & dosage , Sequence Analysis, RNA , Spinal Nerves/surgery
16.
Am J Respir Cell Mol Biol ; 62(2): 204-216, 2020 02.
Article in English | MEDLINE | ID: mdl-31505128

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized as progressive and irreversible fibrosis in the interstitium of lung tissues. There is still an unmet need to develop a novel therapeutic drug for IPF. We have previously demonstrated that periostin, a matricellular protein, plays an important role in the pathogenesis of pulmonary fibrosis. However, the underlying mechanism of how periostin causes pulmonary fibrosis remains unclear. In this study, we sought to learn whether the cross-talk between TGF-ß (transforming growth factor-ß), a central mediator in pulmonary fibrosis, and periostin in lung fibroblasts leads to generation of pulmonary fibrosis and whether inhibitors for integrin αVß3, a periostin receptor, can block pulmonary fibrosis in model mice and the TGF-ß signals in fibroblasts from patients with IPF. We found that cross-talk exists between TGF-ß and periostin signals via αVß3/ß5 converging into Smad3. This cross-talk is necessary for the expression of TGF-ß downstream effector molecules important for pulmonary fibrosis. Moreover, we identified several potent integrin low-molecular-weight inhibitors capable of blocking cross-talk with TGF-ß signaling. One of the compounds, CP4715, attenuated bleomycin-induced pulmonary fibrosis in vivo in mice and the TGF-ß signals in vitro in fibroblasts from patients with IPF. These results suggest that the cross-talk between TGF-ß and periostin can be targeted for pulmonary fibrosis and that CP4715 can be a potential therapeutic agent to block this cross-talk.


Subject(s)
Idiopathic Pulmonary Fibrosis/metabolism , Lung Diseases/metabolism , Transforming Growth Factor beta/metabolism , Animals , Bleomycin/pharmacology , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Mice , Piperidines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Smad3 Protein/genetics
17.
Exp Neurol ; 324: 113128, 2020 02.
Article in English | MEDLINE | ID: mdl-31759899

ABSTRACT

MicroRNA-592 (miR-592) has been reported to play a significant role in mediating neuronal activity, but its possible link with Alzheimer's disease (AD) remains unclear. We aimed to explore the mechanism of miR-592 in oxidative stress (OS) injury of astrocytes (ASTs) from AD rat models induced by D-galactose or Aß25-35 injection. Bioinformatics website and dual-luciferase reporter gene assay clarified the binding affinity between miR-592 and KIAA0319. KIAA0319 was identified as a target gene of miR-592. The mechanism of miR-592, KIAA0319 and the Keap1/Nrf2/ARE signaling pathway in AD was examined after transducing miR-592 mimic, miR-592 inhibitor and siRNA-KIAA0319 into ASTs to query cell viability, OS injury and reactive oxygen species (ROS). The rat models of AD Exhibited highly expressed miR-592 and poorly expressed KIAA0319. Furthermore, inhibition of miR-592 diminished C-Keap1 expression and enhanced N-Nrf2 and NQO1 expression, thus promoting cell viability and reducing OS injury of ASTs. Taken together, these findings suggested that the downregulation of miR-592 inhibited OS injury of ASTs in rat models of AD by up-regulating KIAA0319 through the activation of the Keap1/Nrf2/ARE signaling pathway.


Subject(s)
Alzheimer Disease/drug therapy , Astrocytes/drug effects , Astrocytes/metabolism , Carboxylic Ester Hydrolases/drug effects , Cell Adhesion Molecules/drug effects , Kelch-Like ECH-Associated Protein 1/drug effects , MicroRNAs/metabolism , NF-E2-Related Factor 2/drug effects , Oxidative Stress/drug effects , Signal Transduction/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid beta-Peptides , Animals , Computational Biology , Galactose , Male , Maze Learning , Peptide Fragments , Rats , Rats, Sprague-Dawley
18.
Rev. esp. enferm. dig ; 111(11): 823-827, nov. 2019. ilus, tab, graf
Article in English | IBECS | ID: ibc-190504

ABSTRACT

Background and aims: to investigate the potential effect and mechanism of Salvia miltiorrhiza in Gynura segetum-induced hepatic sinusoidal obstruction syndrome (HSOS). Methods: the mice were gavaged with PBS, Gynura segetum or Gynura segetum, along with 100 or 200 mg/kg Salvia miltiorrhiza. Histological scoring and liver function were performed. The expression of tumor necrosis factor-alpha (TNF-alfa), vascular cellular adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1) and nuclear transcription factor P65 (NF-κBp65) were determined by reverse transcriptase polymerase chain reaction (RT-PCR) and western blot. Results: liver function were effectively improved in the Salvia miltiorrhiza groups. The levels of TNF-alfa, VCAM-1, ICAM-1 and NF-κBp65 were significantly lower in the Salvia miltiorrhiza groups than in the Gynura segetum group. Conclusions: Salvia miltiorrhiza has a therapeutic effect on Gynura segetum-induced HSOS


No disponible


Subject(s)
Animals , Rats , Salvia miltiorrhiza , Plant Extracts/pharmacokinetics , Hepatic Veno-Occlusive Disease/drug therapy , Cell Adhesion Molecules/drug effects , Tumor Necrosis Factor-alpha/drug effects , Vascular Cell Adhesion Molecule-1/drug effects , Transcription Factor RelA/drug effects , Disease Models, Animal , Hepatic Veno-Occlusive Disease/chemically induced , Liver Function Tests/methods , Protective Agents/analysis
19.
Respir Med ; 152: 51-59, 2019 06.
Article in English | MEDLINE | ID: mdl-31128610

ABSTRACT

BACKGROUND: Aspirin desensitization (AD) is an effective and safe therapeutic option for patients with nonsteroidal anti-inflammatory drugs (NSAIDs)-exacerbated respiratory disease (N-ERD). The mechanisms driving its beneficial effects remain poorly understood. OBJECTIVE: To investigate the effect of long-term AD on clinical, biochemical and radiological changes in N-ERD patients. METHODS: The study group consisted of twenty-three individuals with N-ERD who underwent AD, followed by ingestion of 325 mg aspirin twice daily. Twenty patients completed the 52 weeks of AD. The following evaluations were conducted at baseline and in the 52nd week of AD: (i) clinical: asthma exacerbations, Asthma Control Test (ACT), Visual Analogue Scale (VAS) for the assessment of nasal symptoms; (ii) blood and induced sputum supernatant (ISS) periostin, (iii) phenotypes based on induced sputum (IS) cells, (iiii) ISS and nasal lavage (NL) concentration of prostaglandin D2 (PGD2), prostaglandin E2 (PGE2), tetranor-PGD-M, tetranor-PGE-M, 8-iso-PGE2, leukotriene B4 (LTB4), LTC4, LTD4 and LTE4, and urine LTE4. RESULTS: A significant improvement was observed in ACT (P = 0.02) and VAS score (P = 0.008) in the 52nd week of AD. ISS periostin and IS eosinophil count decreased significantly in the 52nd week of AD (P = 0.04 and P = 0.01, respectively). ISS and NL eicosanoid concentrations did not change following long-term AD. CONCLUSION: and Clinical Relevance: AD is associated with a decrease in sputum periostin biosynthesis, which may prevent the recruitment of eosinophils into respiratory tissue and be one of explanation of the clinical benefits of AD. Long-term aspirin administration does not lead to an imbalance between pro- and anti-inflammatory ISS eicosanoids.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Aspirin/adverse effects , Desensitization, Immunologic/methods , Respiration Disorders/immunology , Sputum/metabolism , Adult , Aged , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Aspirin/administration & dosage , Aspirin/therapeutic use , Asthma/chemically induced , Asthma/metabolism , Asthma/physiopathology , Biomarkers/blood , Biomarkers/urine , Carrier Proteins/metabolism , Cell Adhesion Molecules/blood , Cell Adhesion Molecules/drug effects , Eicosanoids/metabolism , Eosinophils/drug effects , Female , Humans , Lipoproteins/metabolism , Male , Middle Aged , Nasal Lavage Fluid/immunology , Prospective Studies , Respiration Disorders/chemically induced , Symptom Flare Up , Trans-Activators/metabolism , Visual Analog Scale
20.
Mar Drugs ; 17(5)2019 May 08.
Article in English | MEDLINE | ID: mdl-31071969

ABSTRACT

Blood circulation disorders, such as hyperlipidemia and arteriosclerosis, are not easily cured by dietary supplements, but they can be mitigated. Although Ecklonia cava extract (ECE), as dietary supplements, are associated with improving the conditions, there are not many studies verifying the same. In this study, the beneficial effect of ECE and leaf of Ginkgo biloba extract (GBE), which is a well-known dietary supplement, were first confirmed in a diet induced-obese model. Afterwards, 4 phlorotannins were isolated from ECE, and their inhibitory effects on vascular cell dysfunction were validated. Pyrogallol-phloroglucinol-6,6-bieckol (PPB) was selected to be orally administered in two mice models: the diet induced obese model and diet induced hypertension model. After four weeks of administration, the blood pressure of all mice was measured, after which they were subsequently sacrificed. PPB was found to significantly improve blood circulation, including a reduction of adhesion molecule expression, endothelial cell (EC) death, excessive vascular smooth muscle cell (VSMC) proliferation and migration, blood pressure, and lipoprotein and cholesterol levels. Based on the excellent efficacy in diet-induced mouse models of obese and hypertension, our results demonstrate that PPB is a valuable active compound from among the phlorotannins that were isolated and it has the potential to be used in functional foods for improving the blood circulation.


Subject(s)
Blood Circulation/drug effects , Dioxins/pharmacology , Hypertension/drug therapy , Obesity/drug therapy , Phloroglucinol/pharmacology , Pyrogallol/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Blood Pressure , Cell Adhesion Molecules/drug effects , Cell Adhesion Molecules/metabolism , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Diet , Ginkgo biloba , Hypertension/chemically induced , Mice , Mice, Inbred C57BL , Models, Animal , Obesity/chemically induced , Phaeophyceae/chemistry , Plant Extracts/pharmacology , RNA, Messenger , Signal Transduction , Tannins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...