Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Hepatology ; 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39042837

ABSTRACT

BACKGROUND AND AIMS: Liver fibrosis represents a global health burden, given the paucity of approved antifibrotic therapies. Liver sinusoidal endothelial cells (LSECs) play a major gatekeeping role in hepatic homeostasis and liver disease pathophysiology. In early tumorigenesis, runt-related transcription factor 3 (RUNX3) functions as a sentinel; however, its function in liver fibrosis in LSECs remains unclear. This study aimed to investigate the role of RUNX3 as an important regulator of the gatekeeping functions of LSECs and explore novel angiocrine regulators of liver fibrosis. APPROACH AND RESULTS: Mice with endothelial Runx3 deficiency develop gradual and spontaneous liver fibrosis secondary to LSEC dysfunction, thereby more prone to liver injury. Mechanistic studies in human immortalized LSECs and mouse primary LSECs revealed that IL-6/JAK/STAT-3 pathway activation was associated with LSEC dysfunction in the absence of RUNX3. Single-cell RNA sequencing and quantitative RT-PCR revealed that leucine-rich alpha-2-glycoprotein 1 (LRG1) was highly expressed in RUNX3-deficient and dysfunctional LSECs. In in vitro and coculture experiments, RUNX3-depleted LSECs secreted LRG1, which activated hepatic stellate cells via TGFBR1-SMAD2/3 signaling in a paracrine manner. Furthermore, circulating LRG1 levels were elevated in mouse models of liver fibrosis and in patients with fatty liver and cirrhosis. CONCLUSIONS: RUNX3 deficiency in the endothelium induces LSEC dysfunction, LRG1 secretion, and liver fibrosis progression. Therefore, endothelial RUNX3 is a crucial gatekeeping factor in LSECs, and profibrotic angiocrine LRG1 may be a novel target for combating liver fibrosis.

2.
Proc Natl Acad Sci U S A ; 114(45): 11926-11931, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29078297

ABSTRACT

Insulin resistance, a key etiological factor in metabolic syndrome, is closely linked to ectopic lipid accumulation and increased intracellular Ca2+ concentrations in muscle and liver. However, the mechanism by which dysregulated intracellular Ca2+ homeostasis causes insulin resistance remains elusive. Here, we show that increased intracellular Ca2+ acts as a negative regulator of insulin signaling. Chronic intracellular Ca2+ overload in hepatocytes during obesity and hyperlipidemia attenuates the phosphorylation of protein kinase B (Akt) and its key downstream signaling molecules by inhibiting membrane localization of pleckstrin homology (PH) domains. Pharmacological approaches showed that elevated intracellular Ca2+ inhibits insulin-stimulated Akt phosphorylation and abrogates membrane localization of various PH domain proteins such as phospholipase Cδ and insulin receptor substrate 1, suggesting a common mechanism inhibiting the membrane targeting of PH domains. PH domain-lipid overlay assays confirmed that Ca2+ abolishes the binding of various PH domains to phosphoinositides (PIPs) with two adjacent phosphate groups, such as PI(3,4)P2, PI(4,5)P2, and PI(3,4,5)P3 Finally, thermodynamic analysis of the binding interaction showed that Ca2+-mediated inhibition of targeting PH domains to the membrane resulted from the tight binding of Ca2+ rather than PH domains to PIPs forming Ca2+-PIPs. Thus, Ca2+-PIPs prevent the recognition of PIPs by PH domains, potentially due to electrostatic repulsion between positively charged side chains in PH domains and the Ca2+-PIPs. Our findings provide a mechanistic link between intracellular Ca2+ dysregulation and Akt inactivation in insulin resistance.


Subject(s)
Calcium/metabolism , Cell Membrane/metabolism , Insulin Resistance/physiology , Phosphatidylinositols/metabolism , Pleckstrin Homology Domains/physiology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Diet, High-Fat , Glucose Intolerance/pathology , Hyperinsulinism/pathology , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Obesity/pathology , Phospholipase C delta/metabolism , Phosphorylation , Protein Binding
3.
Cell Oncol (Dordr) ; 46(1): 117-132, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36329364

ABSTRACT

PURPOSE: Tumor microenvironment consists of various kind of cells, forming complex interactions and signal transductions for tumor growth. Due to this complexity, targeting multiple kinases could yield improved clinical outcomes. In this study, we aimed to investigate the potential of myriocin, from Mycelia sterilia, as a novel dual-kinase inhibitor and suggest myriocin as a candidate for combined chemotherapy. METHODS: We initially evaluated the anti-tumor and anti-metastatic effect of myriocin in mouse allograft tumor models. We examined the effects of myriocin on angiogenesis and tumor vasculature using in vitro, in vivo, and ex vivo models, and also tested the anti-migration effect of myriocin in in vitro models. Next, we explored the effects of myriocin alone and in combination with cisplatin on tumor growth and vascular normalization in mouse models. RESULTS: We found that myriocin inhibited tumor growth and lung metastasis in mouse allograft tumor models. Myriocin induced normalization of the tumor vasculature in the mouse models. We also found that myriocin suppressed angiogenesis through the VEGFR2/PI3K/AKT pathway in endothelial cells (ECs), as well as cancer cell migration by blocking the IκBα/NF-κB(p65)/MMP-9 pathway. Finally, we found that myriocin enhanced the drug delivery efficacy of cisplatin by increasing the integrity of tumor vasculature in the mouse models, which synergistically increased the anti-tumor activity of cisplatin. CONCLUSION: We suggest that myriocin is a novel potent anti-cancer agent that dually targets both VEGFR2 in ECs and IκBα in cancer cells, and exerts more pronounced anti-tumor effects than with either kinase being inhibited alone.


Subject(s)
Cisplatin , Lung Neoplasms , Mice , Animals , Cisplatin/pharmacology , NF-KappaB Inhibitor alpha , Endothelial Cells , Phosphatidylinositol 3-Kinases , Cell Proliferation , Neovascularization, Pathologic , Cell Line, Tumor , Tumor Microenvironment
4.
J Nutr Biochem ; 112: 109212, 2023 02.
Article in English | MEDLINE | ID: mdl-36370926

ABSTRACT

Dietary restriction through low-calorie intake or intermittent fasting benefits many organs, including the brain. This study investigated the neuroprotective effects of fasting in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. We found that fasting every other day rather than weekly increased the levels of brain-derived neurotrophic factor and glial-derived neurotrophic factor in the nigrostriatal pathway. Therefore, we maintained the animals on alternate-day fasting for 2 weeks and injected MPTP (30 mg/kg/day, intraperitoneally [i.p.]) for five days. We observed that alternate-day fasting attenuated MPTP-induced dopaminergic neuronal loss and astroglial activation in the substantia nigra and the striatum. Moreover, neurochemical analysis using high-performance liquid chromatography showed that alternate-day fasting reduced MPTP-induced depletion of striatal dopamine. Consistent with these results, behavioral tests showed that fasting suppressed the motor impairment caused by MPTP. Furthermore, fasting increased the phosphorylation of phosphatidylinositol-3-kinase and protein kinase B, which are downstream signaling molecules of neurotrophic factors. Fasting also increased the phosphorylation of extracellular signal-regulated protein kinase and cAMP response element-binding protein, further supporting the involvement of neurotrophic factors in the observed neuroprotective effects. Hence, our results demonstrated the dopaminergic neuroprotection of intermittent fasting in an MPTP mouse model of Parkinson's disease, supporting the idea that fasting could be an instrumental tool for preventing neurodegeneration in the brain.


Subject(s)
Neuroprotective Agents , Parkinson Disease , Animals , Mice , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Intermittent Fasting , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Neuroprotective Agents/metabolism , Parkinson Disease/metabolism , Substantia Nigra
5.
Arch Pharm Res ; 46(7): 629-645, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37468765

ABSTRACT

Macrophages within the tumor microenvironment (TME), referred to as tumor-associated macrophages (TAMs), are involved in various aspects of tumor progression including initiation, angiogenesis, metastasis, immunosuppression, and resistance to therapy. Myriocin, a natural compound isolated from Mycelia sterilia, is an immunosuppressant that inhibits tumor growth and angiogenesis. However, the mechanisms underlying the effects of myriocin on TAMs and TAM-mediated tumor growth have not yet been elucidated. In this study, we determined the effects of myriocin on TAMs and the underlying mechanism in vitro and in vivo. Myriocin significantly suppressed monocyte-macrophage differentiation and M2 polarization of macrophages but not M1 polarization. In addition, myriocin inhibited the expression of anti-inflammatory cytokines and secretion of proangiogenic factors, such as vascular endothelial growth factor, in M2 macrophages as well as M2-induced endothelial cell permeability. Myriocin also inhibited the PI3K/Akt/mTOR signaling pathway in M2 macrophages. Myriocin reduced the population of M2-like TAMs within the tumor tissue of a mouse allograft tumor model but not that of M1-like TAMs. Moreover, combined treatment with myriocin and cisplatin synergistically suppressed tumor growth and enhanced survival rate in mice by reducing the population of M2-like TAMs. Overall, these results suggest that myriocin inhibits tumor growth by remodeling the TME through suppression of differentiation and polarization of M2-like TAMs via the PI3K/Akt/mTOR signaling pathway.


Subject(s)
Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Animals , Mice , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Line, Tumor , TOR Serine-Threonine Kinases/metabolism , Macrophages , Tumor Microenvironment
6.
Arch Pharm Res ; 44(1): 1-15, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33230600

ABSTRACT

The role of angiogenesis in the growth of organs and tumors is widely recognized. Vascular-organ interaction is a key mechanism and a concept that enables an understanding of all biological phenomena and normal physiology that is essential for human survival under pathological conditions. Recently, vascular endothelial cells have been classified as a type of innate immune cells that are dependent on the pathological situations. Moreover, inflammatory cytokines and signaling regulators activated upon exposure to infection or various stresses play crucial roles in the pathological function of parenchymal cells, peripheral immune cells, stromal cells, and cancer cells in tissues. Therefore, vascular-organ interactions as a vascular microenvironment or tissue microenvironment under physiological and pathological conditions are gaining popularity as an interesting research topic. Here, we review vascular contribution as a major factor in microenvironment homeostasis in the pathogenesis of normal as well as cancerous tissues. Furthermore, we suggest that the normalization strategy of pathological angiogenesis could be a promising therapeutic target for various diseases, including cancer.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Endothelium, Vascular/pathology , Neoplasms/drug therapy , Neovascularization, Pathologic/immunology , Angiogenesis Inhibitors/pharmacology , Animals , Cytokines/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/metabolism , Neoplasms/blood supply , Neoplasms/immunology , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
7.
Mol Neurobiol ; 55(1): 554-566, 2018 01.
Article in English | MEDLINE | ID: mdl-27975170

ABSTRACT

Neurotrophic factors are essential for neuronal survival, plasticity, and development and have been implicated in the action mechanism of antidepressants. In this study, we assessed the neurotrophic factor-inducing and neuroprotective properties of antidepressants. In the first part of the study, we found that fluoxetine, imipramine, and milnacipran (i.p., 20 mg/kg/day for 1 week or 3 weeks) upregulated brain-derived neurotrophic factor in the striatum and substantia nigra both at 1 week and 3 weeks. In contrast, an increase in the glial-derived neurotrophic factor was more obvious at 3 weeks after the antidepressants treatment. Specifically, it was found that fluoxetine and imipramine are more potent in raising the levels of neurotrophic factors than milnacipran. Furthermore, antidepressants elevated the phosphorylation of extracellular signal-regulated-protein kinase (ERK1/2) and the serine/threonine kinase Akt. In the second part of the study, we compared the neuroprotective effects of fluoxetine, imipramine, and milnacipran in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of Parkinson's disease. Pretreament with fluoxetine, imipramine or milnacipran for 3 weeks reduced MPTP-induced dopaminergic neurodegeneration and microglial activation in the nigrostriatal pathway. Neurochemical analysis by HPLC exhibited that antidepressants attenuated the depletion of striatal dopamine. In consistent, beam test showed that behavioral impairment was ameliorated by antidepressants. Neuroprotective effects were more prominent in the fluoxetine or imipramine treatment group than in milnacipran treatment group. Finally, we found that neuroprotection of the antidepressants against 1-methyl-4-phenylpyridinium neurotoxicity in SH-SY5Y cells was attenuated by ERK or Akt inhibitor. These results indicate that neuroprotection by antidepressants might be associated with the induction of neurotrophic factors, and antidepressant could be a potential therapeutic intervention for treatment of Parkinson's disease.


Subject(s)
Antidepressive Agents/therapeutic use , Nerve Growth Factors/metabolism , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Up-Regulation , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Antidepressive Agents/pharmacology , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Humans , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/enzymology , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Substantia Nigra/drug effects , Substantia Nigra/pathology , Substantia Nigra/physiopathology , Up-Regulation/drug effects
8.
Neurosci Res ; 114: 62-69, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27667002

ABSTRACT

Accumulating evidence suggests that chronic inflammation plays a role in the progressive dopaminergic neurodegeneration that occurs in Parkinson's disease. It has been hypothesized that inflammation mediates neuronal damage via exacerbation of a vicious cycle of oxidative stress and mitochondrial dysfunction. The bacterial endotoxin, lipopolysaccharide (LPS), induces microglial activation and inflammation driven dopaminergic neurodegeneration. In order to test the hypothesis that LPS-induced inflammatory response might damage mitochondrial structure and function leading to nigral dopaminergic neuron loss, we injected LPS or saline into the striatum of rats. Here, we found that intrastriatal LPS induced deficit in mitochondrial respiration, damage to mitochondrial cristae, mitochondrial oxidation and nitration. Finally, we found significant loss of dopaminergic neurons in the substantia nigra one week after LPS injection. This study indicates that LPS-induced dopaminergic neurodegeneration might be exerted by mitochondrial injury.


Subject(s)
Lipopolysaccharides/toxicity , Mitochondrial Diseases/etiology , Striatonigral Degeneration , Substantia Nigra , Animals , Cell Count , Disease Models, Animal , Male , Microscopy, Electron , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Striatonigral Degeneration/chemically induced , Striatonigral Degeneration/complications , Striatonigral Degeneration/pathology , Substantia Nigra/metabolism , Substantia Nigra/pathology , Substantia Nigra/ultrastructure , Tyrosine 3-Monooxygenase/metabolism
9.
Neuropharmacology ; 125: 396-407, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28807678

ABSTRACT

In spite of the massive research for the identification of neurorestorative or neuroprotective intervention for curing Parkinson's disease (PD), there is still lack of clinically proven neuroprotective agents. Metformin, a common anti-hyperglycemic drug has been known to possess neuroprotective properties. However, specific mechanisms by which metformin protects neurons from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxicity remain to be elucidated. In this study, we assessed the neuroprotective effects of metformin in the subchronic MPTP model of PD, and explored its feasible mechanisms for neuroprotection. Animals received saline or MPTP injection (30 mg/kg/day) for the first 7 days, and then saline or metformin (200 mg/kg/day) for the next 7 days. Immunohistochemical stainings showed that metformin rescued the tyrosine hydroxylase-positive neurons and attenuated astroglial activation in the nigrostriatal pathway. In parallel, metformin restored dopamine depletion and behavioral impairments exerted by MPTP. Western blot analysis revealed that metformin ameliorated MPTP-induced α-synuclein phosphorylation which was accompanied by increased methylation of protein phosphatase 2A (PP2A), a phosphatase related to α-synuclein dephosphorylation. Moreover, the metformin regimen significantly increased the level of brain derived neurotrophic factor in the substantia nigra, and activated signaling pathways related to cell survival. Proof of concept study revealed that inhibition of PP2A or tropomyosin receptor kinase B reversed neuroprotective property of metformin in SH-SY5Y cells. Our results indicate that metformin provides neuroprotection against MPTP neurotoxicity, which might be mediated by inhibition of α-synuclein phosphorylation and induction of neurotrophic factors.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , MPTP Poisoning/drug therapy , MPTP Poisoning/metabolism , Metformin/pharmacology , Neuroprotective Agents/pharmacology , alpha-Synuclein/metabolism , Animals , Antiparkinson Agents/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Humans , MPTP Poisoning/pathology , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Phosphorylation/drug effects , Proof of Concept Study , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/metabolism , Receptor, trkB/antagonists & inhibitors , Receptor, trkB/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
10.
Neurochem Int ; 102: 79-88, 2017 01.
Article in English | MEDLINE | ID: mdl-27956238

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by prominent loss of the nigral dopaminergic neurons and motor symptoms, such as resting tremor and bradykinesia. Evidence suggests that neuroinflammation may play a critical role in PD pathogenesis. Interleukin (IL)-32 is a newly-identified proinflammatory cytokine, which regulates innate and adaptive immune responses by activating p38 MAPK and NF-κB signaling pathways. The cytokine has been implicated in cancers and autoimmune, inflammatory, and infectious diseases. In this study, we attempted to identify the effects of IL-32ß on dopaminergic neurotoxicity induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), using IL-32ß transgenic mice. Male wild type and IL-32ß transgenic mice received intraperitoneal injections of vehicle or MPTP (15 mg/kg × 4). Immunohistochemistry showed that overexpression of IL-32ß significantly increased MPTP-mediated loss of dopaminergic neurons in the substantia nigra and deletion of tyrosine hydroxylase-positive fibers in the striatum. Dopamine depletion in the striatum and deficit in locomotor activity were enhanced in IL-32ß transgenic mice. These results were accompanied by higher neuroinflammatory responses in the brains of transgenic mice. Finally, we found that IL-32ß exaggerated MPTP-mediated activation of p38 MAPK and JNK pathways, which have been shown to be involved in MPTP neurotoxicity. These results suggest that IL-32ß exacerbates MPTP neurotoxicity through enhanced neuroinflammatory responses.


Subject(s)
Corpus Striatum/drug effects , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Interleukins/genetics , MPTP Poisoning , Animals , Corpus Striatum/metabolism , Disease Models, Animal , MPTP Poisoning/metabolism , Mice, Transgenic , Substantia Nigra/drug effects , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL