Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.983
Filter
1.
CNS Neurosci Ther ; 30(8): e14913, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39123294

ABSTRACT

BACKGROUND: Hyperglycemia-induced neuroinflammation significantly contributes to diabetic neuropathic pain (DNP), but the underlying mechanisms remain unclear. OBJECTIVE: To investigate the role of Sirt3, a mitochondrial deacetylase, in hyperglycemia-induced neuroinflammation and DNP and to explore potential therapeutic interventions. METHOD AND RESULTS: Here, we found that Sirt3 was downregulated in spinal dorsal horn (SDH) of diabetic mice by RNA-sequencing, which was further confirmed at the mRNA and protein level. Sirt3 deficiency exacerbated hyperglycemia-induced neuroinflammation and DNP by enhancing microglial aerobic glycolysis in vivo and in vitro. Overexpression of Sirt3 in microglia alleviated inflammation by reducing aerobic glycolysis. Mechanistically, high-glucose stimulation activated Akt, which phosphorylates and inactivates FoxO1. The inactivation of FoxO1 diminished the transcription of Sirt3. Besides that, we also found that hyperglycemia induced Sirt3 degradation via the mitophagy-lysosomal pathway. Blocking Akt activation by GSK69093 or metformin rescued the degradation of Sirt3 protein and transcription inhibition of Sirt3 mRNA, which substantially diminished hyperglycemia-induced inflammation. Metformin in vivo treatment alleviated neuroinflammation and diabetic neuropathic pain by rescuing hyperglycemia-induced Sirt3 downregulation. CONCLUSION: Hyperglycemia induces metabolic reprogramming and inflammatory activation in microglia through the regulation of Sirt3 transcription and degradation. This novel mechanism identifies Sirt3 as a potential drug target for treating DNP.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Neuropathies , Down-Regulation , Glycolysis , Hyperglycemia , Mice, Inbred C57BL , Microglia , Sirtuin 3 , Animals , Sirtuin 3/metabolism , Sirtuin 3/genetics , Mice , Glycolysis/drug effects , Glycolysis/physiology , Down-Regulation/drug effects , Down-Regulation/physiology , Hyperglycemia/metabolism , Microglia/metabolism , Microglia/drug effects , Male , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/metabolism , Inflammation/metabolism , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/etiology , Metformin/pharmacology
2.
Curr Opin Neurobiol ; 88: 102901, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39142020

ABSTRACT

Stress-related and substance use disorders are both characterized by disruptions in reward-related behaviors, and these disorders are often comorbid with one another. Recent investigations have identified a novel mechanism of inhibitory plasticity induced by both stress and substance use within the ventral tegmental area (VTA), a key region in reward processing. This mechanism involves the neuron-specific potassium chloride cotransporter isoform 2 (KCC2), which is essential in modulating inhibitory signaling through the regulation of intracellular chloride (Cl-) in VTA GABA neurons. Experiences, such as exposure to stress or substance use, diminish KCC2 expression in VTA GABA neurons, leading to abnormal reward-related behaviors. Here, we review literature suggesting that KCC2 downregulation contributes to irregular dopamine (DA) transmission, impacting multiple reward circuits and promoting maladaptive behaviors. Activating KCC2 restores canonical GABA functioning and reduces behavioral deficits in preclinical models, leading us to advocate for KCC2 as a target for therapies aimed at alleviating and mitigating various stress-related and substance use disorders.


Subject(s)
Down-Regulation , K Cl- Cotransporters , Stress, Psychological , Substance-Related Disorders , Symporters , Animals , Humans , Down-Regulation/physiology , Mesencephalon/metabolism , Reward , Stress, Psychological/metabolism , Substance-Related Disorders/metabolism , Symporters/metabolism , Ventral Tegmental Area/metabolism
3.
J Pain ; 25(10): 104598, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38866121

ABSTRACT

The voltage-gated sodium channel ß2 subunit protein (SCN2B) plays a crucial role in neuropathic pain. However, the role and mechanisms of SCN2B in orofacial neuropathic pain are still unclear. This study aimed to investigate the upstream regulatory mechanisms of SCN2B in the trigeminal ganglion (TG) underlying orofacial neuropathic pain. Chronic constriction injury of the infraorbital nerve (CCI-ION) of mice was performed to establish the model of orofacial neuropathic pain. Von Frey filament test was performed to detect the head withdrawal threshold (HWT) of mice. Quantitative reverse transcription-polymerase chain, western blotting (WB), fluorescence in situ hybridization, and immunofluorescence (IF) staining were used to detect the expression and distribution of SCN2B and miR-6954-3p in the TG of mice. A luciferase activity assay was carried out to prove the binding between SCN2B messenger ribonucleic acid (mRNA) and miR-6954-3p. After the CCI-ION surgery, the levels of Scn2b mRNA and protein significantly increased and miR-6954-3p decreased in the TG of mice with decreasing HWT. IF staining revealed that SCN2B was expressed specifically in the TG neurons. Silencing SCN2B in the TG of CCI-ION mice significantly increased the HWT. Importantly, the 3'-untranslated region of Scn2b mRNA was proved to bind with miR-6954-3p. Fluorescence in situ hybridization and IF staining demonstrated that miR-6954-3p was expressed in TG neurons and co-expressed with SCN2B. Furthermore, intraganglionic injection of miR-6954-3p agomir into the TG of CCI-ION mice resulted in the downregulation of SCN2B and increased the HWT. These findings suggest that the downregulation of miR-6954-3p in the TG promotes orofacial neuropathic pain by promoting SCN2B expression following trigeminal nerve injury. PERSPECTIVE: This study points to the important role of SCN2B in orofacial neuropathic pain. Furthermore, miR-6954-3p is proven to regulate the expression of SCN2B by binding to the 3'-untranslated region of Scn2b mRNA. These findings indicate that SCN2B and miR-6954-3p are potential therapeutic targets for the treatment of orofacial neuropathic pain.


Subject(s)
Down-Regulation , Facial Pain , MicroRNAs , Neuralgia , Voltage-Gated Sodium Channel beta-2 Subunit , Animals , Male , Mice , Disease Models, Animal , Down-Regulation/physiology , Facial Pain/metabolism , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics , Neuralgia/metabolism , Trigeminal Ganglion/metabolism , Voltage-Gated Sodium Channel beta-2 Subunit/metabolism , Female
4.
Int J Cardiol ; 408: 132158, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38744338

ABSTRACT

BACKGROUND: Cardiomyocyte apoptosis plays a vital role in myocardial ischemia-reperfusion (MI/R) injury; however, the role of beclin1 (BECN1) remains unclear. This study aimed at revealing the function of BECN1 during cardiomyocyte apoptosis after MI/R injury. METHODS: In vivo, TTC and Evan's blue double staining was applied to verify the gross morphological alteration in both wild type (WT) mice and BECN1 transgene mice (BECN1-TG), and TUNEL staining and western blot were adopted to evaluate the cardiomyocyte apoptosis. In vitro, a hypoxia/reoxygenation (H/R) model was established in H9c2 cells to simulate MI/R injury. Proteomics analysis was preformed to verify if apoptosis occurs in the H/R cellular model. And apoptosis factors, RIPK1, Caspase-1, Caspase-3, and cleaved Caspase-3, were investigated using western bolting. In addition, the mRNA level were verified using RT-PCR. To further investigate the protein interactions small interfering RNA and lentiviral transfection were used. To continue investigate the protein interactions, immunofluorescence and coimmunoprecipitation were applied. RESULTS: Morphologically, BECN1 significantly attenuated the apoptosis from TTC-Evan's staining, TUNEL, and cardiac tissue western blot. After H/R, a RIPK1-induced complex (complex II) containing RIPK1, Caspase-8, and FADD was formed. Thereafter, cleaved Caspase-3 was activated, and myocyte apoptosis occurred. However, BECN1 decreased the expression of RIPK1, Caspase-8, and FADD. Nevertheless, BECN1 overexpression increased RIPK1 ubiquitination before apoptosis by inhibiting OTUD1. CONCLUSIONS: BECN1 regulates FADD/RIPK1/Caspase-8 complex formation via RIPK1 ubiquitination by downregulating OTUD1 in C-Caspase-3-induced myocyte apoptosis after MI/R injury. Therefore, BECN1 can function as a cardioprotective candidate.


Subject(s)
Apoptosis , Beclin-1 , Caspase 8 , Down-Regulation , Fas-Associated Death Domain Protein , Myocardial Reperfusion Injury , Myocytes, Cardiac , Receptor-Interacting Protein Serine-Threonine Kinases , Ubiquitination , Animals , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Fas-Associated Death Domain Protein/metabolism , Apoptosis/physiology , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Caspase 8/metabolism , Beclin-1/metabolism , Ubiquitination/physiology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Down-Regulation/physiology , Male , Mice, Transgenic , Mice, Inbred C57BL , Cells, Cultured
5.
Neurochem Res ; 49(8): 2105-2119, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38819696

ABSTRACT

Ischemic stroke involves various pathological processes, among which ferroptosis is crucial. Previous studies by our group have indicated that electroacupuncture (EA) mitigates ferroptosis after ischemic stroke; however, the precise mechanism underlying this effect remains unclear. In the present study, we developed a rat model of middle cerebral artery occlusion/reperfusion. We chose the main acupoint of the treatment methods of the "Awakening and Opening of the Brain". Rats' neurological function and motor coordination were evaluated by neurological function score and the rotarod test, respectively, and the volume of cerebral infarction was analyzed by 2,3,5-triphenyltetrazolium chloride Staining. The cerebrovascular conditions were visualized by time-of-flight magentic resonance angiography. In addition, we detected changes in lipid peroxidation and endogenous antioxidant activity by measuring the malondialdehyde, glutathione, superoxide dismutase activities, glutathione/oxidized glutathione and reduced nicotinamide adenine dinucleotide phosphate/oxidized nicotinamide adenine dinucleotide phosphate ratios. Inductively coupled plasma-mass spectrometry, western blot, reverse transcription-polymerase chain reaction, fluoro-jade B staining, immunofluorescence analysis, and transmission electron microscopy were utilized to examine the influence of EA. The results indicate that EA treatment was effective in reversing neurological impairment, neuronal damage, and protecting mitochondrial morphology and decreasing the cerebral infarct volume in the middle cerebral artery occlusion/reperfusion rat model. EA reduced iron levels, inhibited lipid peroxidation, increased endogenous antioxidant activity, modulated the expression of several ferroptosis-related proteins, and promoted nuclear factor-E2-related factor 2 (Nrf2) nuclear translocation. However, the protective effect of EA was hindered by the Nrf2 inhibitor ML385. These findings suggest that EA can suppress ferroptosis and decrease damage caused by cerebral ischemia/reperfusion by activating Nrf2 and increasing the protein expression of solute carrier family 7 member 11 and glutathione peroxidase 4.


Subject(s)
Electroacupuncture , Ferroptosis , Infarction, Middle Cerebral Artery , NF-E2-Related Factor 2 , Phospholipid Hydroperoxide Glutathione Peroxidase , Rats, Sprague-Dawley , Animals , Ferroptosis/physiology , Electroacupuncture/methods , NF-E2-Related Factor 2/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Male , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/therapy , Reperfusion Injury/metabolism , Rats , Neurons/metabolism , Down-Regulation/physiology
6.
Glia ; 72(8): 1374-1391, 2024 08.
Article in English | MEDLINE | ID: mdl-38587131

ABSTRACT

Oligodendrocytes and astrocytes are metabolically coupled to neuronal compartments. Pyruvate and lactate can shuttle between glial cells and axons via monocarboxylate transporters. However, lactate can only be synthesized or used in metabolic reactions with the help of lactate dehydrogenase (LDH), a tetramer of LDHA and LDHB subunits in varying compositions. Here we show that mice with a cell type-specific disruption of both Ldha and Ldhb genes in oligodendrocytes lack a pathological phenotype that would be indicative of oligodendroglial dysfunctions or lack of axonal metabolic support. Indeed, when combining immunohistochemical, electron microscopical, and in situ hybridization analyses in adult mice, we found that the vast majority of mature oligodendrocytes lack detectable expression of LDH. Even in neurodegenerative disease models and in mice under metabolic stress LDH was not increased. In contrast, at early development and in the remyelinating brain, LDHA was readily detectable in immature oligodendrocytes. Interestingly, by immunoelectron microscopy LDHA was particularly enriched at gap junctions formed between adjacent astrocytes and at junctions between astrocytes and oligodendrocytes. Our data suggest that oligodendrocytes metabolize lactate during development and remyelination. In contrast, for metabolic support of axons mature oligodendrocytes may export their own glycolysis products as pyruvate rather than lactate. Lacking LDH, these oligodendrocytes can also "funnel" lactate through their "myelinic" channels between gap junction-coupled astrocytes and axons without metabolizing it. We suggest a working model, in which the unequal cellular distribution of LDH in white matter tracts facilitates a rapid and efficient transport of glycolysis products among glial and axonal compartments.


Subject(s)
Axons , Glycolysis , L-Lactate Dehydrogenase , Oligodendroglia , Animals , Oligodendroglia/metabolism , Axons/metabolism , L-Lactate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/genetics , Glycolysis/physiology , Mice , Down-Regulation/physiology , Mice, Inbred C57BL , Lactate Dehydrogenase 5/metabolism , Astrocytes/metabolism , Astrocytes/ultrastructure , Mice, Transgenic , Isoenzymes/metabolism , Isoenzymes/genetics , Gap Junctions/metabolism , Gap Junctions/ultrastructure , Mice, Knockout
7.
New Phytol ; 237(5): 1667-1683, 2023 03.
Article in English | MEDLINE | ID: mdl-36444526

ABSTRACT

One of the strategies that plants adopt to cope with an unfavorable environment is to sacrifice their growth for tolerance. Although moderate salt stress can induce root growth inhibition, the molecular mechanisms regulating this process have yet to be elucidated. Here, we found that overexpression of a zinc finger-homeodomain family transcription factor, HOMEOBOX PROTEIN 24 (HB24), led to longer primary roots than in the wild-type in the presence of 125 mM NaCl, whereas this phenotype was reversed for the hb24 loss-of-function mutant, indicating a negative impact of HB24 on salt-induced root growth inhibition. We then found that salt stress triggered the degradation of HB24 via the ubiquitin-proteasome pathway, as mediated by a plant U-box type E3 ubiquitin ligase 30 (PUB30) that directly targets HB24. We verified that HB24 is able to directly bind to the promoters of Sugars Will Eventually be Exported Transporter 11/12 (SWEET11/12) to regulate their expression in roots. Through genetic and biochemical assays, we further demonstrated that the HB24-SWEET11 module plays a negative role in salt-induced root growth inhibition. Therefore, we propose that under salt stress, PUB30 mediates HB24's degradation, thereby downregulating the expression of SWEET11, resulting in reduced sucrose supply and root growth inhibition.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Plant Roots , Salt Stress , Sucrose , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Down-Regulation/genetics , Down-Regulation/physiology , Gene Expression Regulation, Plant , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Membrane Transport Proteins/metabolism , Plant Roots/genetics , Plant Roots/growth & development , Plant Roots/metabolism , Plants, Genetically Modified/metabolism , Salt Stress/genetics , Salt Stress/physiology , Stress, Physiological/genetics , Sucrose/metabolism , Ubiquitin-Protein Ligases/metabolism
8.
Int J Mol Sci ; 23(4)2022 Feb 15.
Article in English | MEDLINE | ID: mdl-35216267

ABSTRACT

Bladder cancer (BC) has a high recurrence rate worldwide. The aim of this study was to evaluate the role of fatty acid binding protein 6 (FABP6) in proliferation and migration in human bladder cancer cells. Cell growth was confirmed by MTT and colony formation assay. Western blotting was used to explore protein expressions. Wound healing and Transwell assays were performed to evaluate the migration ability. A xenograft animal model with subcutaneous implantation of BC cells was generated to confirm the tumor progression. Knockdown of FABP6 reduced cell growth in low-grade TSGH-8301 and high-grade T24 cells. Cell cycle blockade was observed with the decrease of CDK2, CDK4, and Ki67 levels in FABP6-knockdown BC cells. Interestingly, knockdown of FBAP6 led to downregulation of autophagic markers and activation of AKT-mTOR signaling. The application of PI3K/AKT inhibitor decreased cell viability mediated by FABP6-knockdown additionally. Moreover, FABP6-knockdown reduced peroxisome proliferator-activated receptor γ and retinoid X receptor α levels but increased p-p65 expression. Knockdown of FABP6 also inhibited BC cell motility with focal adhesive complex reduction. Finally, shFABP6 combined with cisplatin suppressed tumor growth in vivo. These results provide evidence that FABP6 may be a potential target in BC cells progression.


Subject(s)
Autophagy/physiology , Cell Cycle/physiology , Cell Movement/physiology , Fatty Acid-Binding Proteins/metabolism , Gastrointestinal Hormones/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Cell Survival/physiology , Down-Regulation/physiology , Gene Expression Regulation, Neoplastic/physiology , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology
9.
Int J Mol Sci ; 23(4)2022 Feb 21.
Article in English | MEDLINE | ID: mdl-35216486

ABSTRACT

The passage number of cells refers to the number of subculturing processes that the cells have undergone. The effect of passage number on morphological and phenotypical characteristics of cells is of great importance. Advanced glycation end products have also been associated with cell functionality and characteristics. Murine monocyte RAW 264.7 cells differentiate into osteoclasts upon receptor activation caused by nuclear factor-kappa-Β ligand (RANKL) treatment. This study aims to identify the role of passage number on intracellular advanced glycation end products (AGEs) formation and osteoclastogenic differentiation of RAW 264.7 cells. Western blotting was performed to check intracellular AGE formation along with fluorometric analysis using a microplate reader. Tartrate-resistant acid phosphatase (TRAP) staining was performed to check osteoclastogenic differentiation, and qPCR was realized to check the responsible mRNA expression. Immunofluorescence was used to check the morphological changes. Intracellular AGE formation was increased with passaging, and the higher passage number inhibited multinucleated osteoclastogenic differentiation. Osteoclastogenic gene expression also showed a reducing trend in higher passages, along with a significant reduction in F-actin ring size and number. Lower passages should be used to avoid the effects of cell subculturing in in vitro osteoclastogenesis study using RAW 264.7 cells.


Subject(s)
Down-Regulation/physiology , Glycation End Products, Advanced/metabolism , Osteogenesis/physiology , RANK Ligand/metabolism , Actins/metabolism , Animals , Bone Resorption/metabolism , Cell Differentiation/physiology , Cell Line , Mice , NF-kappa B/metabolism , Osteoclasts/metabolism , RAW 264.7 Cells , Signal Transduction/physiology , Tartrate-Resistant Acid Phosphatase/metabolism
10.
Int J Mol Sci ; 23(4)2022 Feb 14.
Article in English | MEDLINE | ID: mdl-35216190

ABSTRACT

Endometrial cancer (EC) is the most frequent gynaecologic cancer in postmenopausal women. We used 2D-DIGE and mass spectrometry to identify candidate biomarkers in endometrial cancer, analysing the serum protein contents of 10 patients versus 10 control subjects. Using gel-based proteomics, we identified 24 candidate biomarkers, considering only spots with a fold change in volume percentage ≥ 1.5 or intensity change ≤ 0.6, which were significantly different between cases and controls (p < 0.05). We used Western blotting analysis both in the serum and tissue of 43 patients for data validation. Among the identified proteins, we selected Suprabasin (SBSN), an oncogene previously associated with poor prognosis in different cancers. SBSN principal isoforms were subjected to Western blotting analysis in serum and surgery-excised tissue: both isoforms were downregulated in the tissue. However, in serum, isoform 1 was upregulated, while isoform 2 was downregulated. Data-mining on the TCGA and GTEx projects, using the GEPIA2.0 interface, indicated a diminished SBSN expression in the Uterine Corpus Endometrial Cancer (UCEC) database compared to normal tissue, confirming proteomic results. These results suggest that SBSN, specifically isoform 2, in tissue or serum, could be a potential novel biomarker in endometrial cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Endometrial Neoplasms/metabolism , Proteome/metabolism , Adult , Antigens, Differentiation/metabolism , Down-Regulation/physiology , Endometrium/metabolism , Female , Humans , Middle Aged , Oncogenes/physiology , Protein Isoforms/metabolism , Proteomics/methods , Two-Dimensional Difference Gel Electrophoresis/methods , Up-Regulation/physiology
11.
Toxicol Appl Pharmacol ; 436: 115849, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34974052

ABSTRACT

Nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) is a Group 1 human carcinogen, as classified by the International Agency for Research of Cancer (IARC), and plays a significant role in lung carcinogenesis. However, its carcinogenic mechanism has not yet been fully elucidated. In this study, we performed colony formation assays, soft-agar assays, and tumor growth in nude mice to show that 100 mg/L NNK facilitates the malignant transformation of human bronchial epithelial Beas-2B cells. Transcriptome sequencing showed that insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1), a post-transcriptional regulator, was differentially expressed in NNK-induced malignant transformed Beas-2B cells (2B-NNK cells). Small interfering RNA (SiRNA) was used to downregulate the expression of the IGF2BP1 gene. The reduction in protein expression, cell proliferation rate, and colony-forming ability and the increase in the apoptosis rate of Beas-2B cells transfected with the SiRNA indicated a role for IGF2BP1 in NNK-induced malignant transformation. IGF2BP1 is an N6-methyladenosine (m6A) regulatory factor, but it is not known whether its association with m6A mediates the malignant transformation of cells. Therefore, we measured the overall levels of m6A in Beas-2B cells. We found that the overall m6A level was lower in 2B-NNK cells, and knocking down IGF2BP1, the overall level of m6A was restored. Hence, we concluded that IGF2BP1 is involved in the NNK-induced malignant transformation of Beas-2B cells, possibly via m6A modification. This study therefore contributes novel insights into the environmental pathogenesis of lung cancer and the gene regulatory mechanisms of chemical carcinogenesis.


Subject(s)
Bronchi/drug effects , Butanones/pharmacology , Cell Transformation, Neoplastic/genetics , Epithelial Cells/drug effects , Methyltransferases/metabolism , Nicotiana/adverse effects , Nitrosamines/pharmacology , RNA-Binding Proteins/genetics , Adult , Aged , Animals , Apoptosis/drug effects , Apoptosis/genetics , Carcinogens/pharmacology , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Transformation, Neoplastic/chemically induced , Down-Regulation/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Lung/drug effects , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Transfection/methods
12.
Arch Biochem Biophys ; 715: 109047, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34619102

ABSTRACT

OBJECTIVE: Sepsis is a leading cause of morbidity and mortality after surgery. We aimed to explore the role of long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) sponging microRNA-26a-5p in sepsis-induced myocardial injury by regulating regulator of calcineurin 2 (Rcan2). METHODS: HL-1 cells were incubated with lipopolysaccharide (LPS) to induce in vitro cardiomyocyte injury models, which were then treated with silenced MALAT1 vector, miR-26a-5p mimic or Rcan2 overexpression vector. Next, inflammatory factor level and apoptosis of cells were determined. The in vivo mouse models were constructed by intraperitoneal injection of LPS. The modeled mice were injected with relative oligonucleotides and the pathology, apoptosis, and inflammation in mouse myocardial tissues were assessed. Expression of MALAT1, miR-26a-5p and Rcan2 in vivo and in vitro was evaluated. RESULTS: MALAT1 and Rcan2 were upregulated while miR-26a-5p was downregulated in LPS-treated HL-1 cells and mice. MALAT1 silencing or miR-26a-5p upregulation suppressed LPS-induced inflammation and apoptosis of cardiomyocytes in cellular and animal models. These effects of elevated miR-26a-5p could be reversed by upregulating Rcan2, and MALAT1 knockdown-induced ameliorative impacts could be reversed by miR-26a-5p downregulation. CONCLUSION: MALAT1 silencing elevated miR-26a-5p to ameliorate LPS-induced myocardial injury by reducing Rcan2. Our research may provide novel biomarkers for the treatment of sepsis.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism , Myocardial Ischemia/physiopathology , RNA, Long Noncoding/metabolism , Sepsis/physiopathology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/physiology , Down-Regulation/drug effects , Down-Regulation/physiology , Inflammation/chemically induced , Inflammation/physiopathology , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Myocardial Ischemia/etiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Sepsis/complications , Up-Regulation/drug effects , Up-Regulation/physiology
13.
J Bacteriol ; 204(1): e0037821, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34694902

ABSTRACT

Salmonella enterica serovar Typhimurium invades the intestinal epithelium and induces inflammatory diarrhea using the Salmonella pathogenicity island 1 (SPI1) type III secretion system (T3SS). Expression of the SPI1 T3SS is controlled by three AraC-like regulators, HilD, HilC, and RtsA, which form a feed-forward regulatory loop that leads to activation of hilA, encoding the main transcriptional regulator of the T3SS structural genes. This complex system is affected by numerous regulatory proteins and environmental signals, many of which act at the level of hilD mRNA translation or HilD protein function. Here, we show that the sRNA MicC blocks translation of the hilD mRNA by base pairing near the ribosome binding site. MicC does not induce degradation of the hilD message. Our data indicate that micC is transcriptionally activated by SlyA, and SlyA feeds into the SPI1 regulatory network solely through MicC. Transcription of micC is negatively regulated by the OmpR/EnvZ two-component system, but this regulation is dependent on SlyA. OmpR/EnvZ control SPI1 expression partially through MicC but also affect expression through other pathways, including an EnvZ-dependent, OmpR-independent mechanism. MicC-mediated regulation plays a role during infection, as evidenced by an SPI1 T3SS-dependent increase in Salmonella fitness in the intestine in the micC deletion mutant. These results further elucidate the complex regulatory network controlling SPI1 expression and add to the list of sRNAs that control this primary virulence factor. IMPORTANCE The Salmonella pathogenicity island 1 (SPI1) type III secretion system (T3SS) is the primary virulence factor required for causing intestinal disease and initiating systemic infection. The system is regulated in response to a large variety of environmental and physiological factors such that the T3SS is expressed at only the appropriate time and place in the host during infection. Here, we show how the sRNA MicC affects expression of the system. This work adds to our detailed mechanistic studies aimed at a complete understanding of the regulatory circuit.


Subject(s)
Bacterial Proteins/metabolism , Down-Regulation/physiology , RNA, Bacterial/metabolism , Salmonella typhimurium/metabolism , Transcription Factors/metabolism , Type III Secretion Systems/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Down-Regulation/genetics , Gene Expression Regulation, Bacterial/physiology , Host Factor 1 Protein , RNA, Bacterial/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Salmonella typhimurium/genetics , Transcription Factors/genetics , Type III Secretion Systems/genetics
14.
Can J Physiol Pharmacol ; 100(2): 142-150, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34559978

ABSTRACT

The endothelial-mesenchymal transition (EndMT) participates in the progression of diabetic retinopathy (DR), but cell-intrinsic factors modulating this process remain elusive. In this study, we explored the role of lysophosphatidic acid (LPA) - producing enzyme, acylglycerol kinase (AGK), in the EndMT of human retinal microvascular endothelial cells (HRECs) under high-glucose (HG) conditions. We found that AGK was significantly elevated in HG-treated cells. In addition, AGK knockdown reversed the HG-induced EndMT in HRECs, which was evidenced by the increased endothelial markers (CD31 and VE-cadherin) and decreased mesenchymal markers (FSP1 and α-SMA). Furthermore, downregulation of AGK inhibited the HG-induced activation of transforming growth factor ß (TGF-ß)/Notch pathways, whereas exogenous TGF-ß1 (10 ng/mL) impeded the inhibitory effects of AGK knockdown on HG-induced EndMT in HRECs. Additionally, the silencing of AGK abolished the HG-induced upregulation of LPA and its receptor, LPA receptor 1 (LPAR1), and overexpression of LPAR1 further rescued the AGK knockdown-mediated inhibition of the EndMT process. In conclusion, we demonstrate that downregulation of AGK suppresses HG-induced EndMT in HRECs through regulating the LPAR1/TGF-ß/Notch signaling pathway, indicating that AGK might be a potential therapeutic target for the treatment of DR.


Subject(s)
Down-Regulation/genetics , Down-Regulation/physiology , Endothelial Cells/physiology , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Glucose/adverse effects , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Lysophosphatidic Acid/genetics , Receptors, Notch/metabolism , Retinal Vessels/cytology , Signal Transduction/genetics , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Cells, Cultured , Gene Expression Regulation/genetics , Humans , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Notch/genetics , Transforming Growth Factor beta/genetics
15.
Eur J Histochem ; 65(s1)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34814650

ABSTRACT

Evidence suggests that transient receptor potential (TRP) ion channels dysfunction significantly contributes to the physiopathology of metabolic and neurological disorders. Dysregulation in functions and expression in genes encoding the TRP channels cause several inherited diseases in humans (the so-called 'TRP channelopathies'), which affect the cardiovascular, renal, skeletal, and nervous systems. This study aimed to evaluate the expression of ion channels in the forebrain of rats with diet-induced obesity (DIO). DIO rats were studied after 17 weeks under a hypercaloric diet (high-fat diet, HFD) and were compared to the control rats with a standard diet (CHOW). To determine the systemic effects of HFD exposure, we examined food intake, fat mass content, fasting glycemia, insulin levels, cholesterol, and triglycerides. qRT-PCR, Western blot, and immunochemistry analysis were performed in the frontal cortex (FC) and hippocampus (HIP). After 17 weeks of HFD, DIO rats increased their body weight significantly compared to the CHOW rats. In DIO rats, TRPC1 and TRPC6 were upregulated in the HIP, while they were downregulated in the FC. In the case of TRPM2 expression, instead was increased both in the HIP and in the FC. These could be related to the increase of proteins and nucleic acid oxidation. TRPV1 and TRPV2 gene expression showed no differences both in the FC and HIP. In general, qRT-PCR analyses were confirmed by Western blot analysis. Immunohistochemical procedures highlighted the expression of the channels in the cell body of neurons and axons, particularly for the TRPC1 and TRPC6. The alterations of TRP channel expression could be related to the activation of glial cells or the neurodegenerative process presented in the brain of the DIO rat highlighted with post synaptic protein (PSD 95) alterations. The availability of suitable animal models may be useful for studying possible pharmacological treatments to counter obesity-induced brain injury. The identified changes in DIO rats may represent the first insight to characterize the neuronal alterations occurring in obesity. Further investigations are necessary to characterize the role of TRP channels in the regulation of synaptic plasticity and obesity-related cognitive decline.


Subject(s)
Frontal Lobe/metabolism , Hippocampus/metabolism , Obesity/physiopathology , Transient Receptor Potential Channels/metabolism , Animals , Axons/metabolism , Diet, High-Fat , Down-Regulation/physiology , Frontal Lobe/pathology , Gene Expression/physiology , Hippocampus/pathology , Male , Obesity/pathology , Oxidative Stress/physiology , Rats, Wistar , Up-Regulation/physiology
16.
Exp Eye Res ; 213: 108836, 2021 12.
Article in English | MEDLINE | ID: mdl-34774487

ABSTRACT

Retinal vascular development is a very tightly regulated and organized process of vessel formation and regression to generate the mature vasculature system. Claudin-3 has been found to be required for the normal development of the neural retina and its vessels in zebrafish in our recent study. In this study, we investigated whether Claudin-3 played a role in the development of mouse retinal vasculature. Immunofluorescent staining was performed to detect the expression and localization of Claudin-3 in the mouse retina. Intravitreal injection of a recombinant adeno-associated virus (AAV) expressing a short hairpin RNA targeting Claudin-3 mRNA was performed to down-regulate Claudin-3 expression in retina in neonatal (Postnatal Day 3, P3) C57BL/6J mice. Retinal vessels were examined by isolectin B4 immunofluorescent staining on the whole-mount retinas and frozen retinal sections at P10. The apoptotic retinal ganglion cells (RGCs) were measured by TdT-mediated dUTP nick-end labelling (TUNEL) staining. Vascular endothelial growth factor A (VEGF-A) expression was detected by immunofluorescent staining. The protein levels of Claudin-3, VEGF-A and B cell lymphoma 2 (Bcl-2) were evaluated by Western blot at P7, P10 and P14. We found that Claudin-3 mainly expressed in the RGCs and progressively increased during the retinal development. The AAV-mediated downregulation of Claudin-3 at P3 impeded the development of retinal deep vascularization of P10 mouse, but without effect on the development of the retinal superficial plexus. Claudin-3 knockdown increased RGC apoptosis and reduced the expression of VEGF-A and Bcl-2 in the retinas. These results suggested that the downregulation of Claudin-3 induced RGC apoptosis and impeded the mouse retinal vascular development by downregulating the levels of VEGF-A and Bcl-2.


Subject(s)
Claudin-3/metabolism , Dependovirus/genetics , Neovascularization, Physiologic/physiology , Retinal Vessels/physiology , Animals , Animals, Newborn , Apoptosis , Blotting, Western , Down-Regulation/physiology , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Proto-Oncogene Proteins c-bcl-2/metabolism , Real-Time Polymerase Chain Reaction , Retina/metabolism , Retinal Ganglion Cells/pathology , Vascular Endothelial Growth Factor A/metabolism
17.
Life Sci ; 287: 120131, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34767806

ABSTRACT

AIMS: Potential anti-obesity effects of quinizarin, a plant anthraquinone, were investigated using 3 T3-L1 preadipocyte cells and high-fat diet (HD)-induced obese mice. MAIN METHOD: Cell viability was determined using the MTT assay. Triglyceride (TG) and lipid accumulation were determined using a TG assay kit and Oil Red O staining, respectively. Adipogenic, lipogenic, and lipolytic gene and protein expression was measured by RT-PCR or Western blot. Serum biochemical indices, including cholesterol and blood glucose, in HD-fed obese mice were determined using corresponding assay kits. Histological analysis was performed with haematoxylin and eosin (H&E) staining. RESULTS: Quinizarin (0-10 µM) significantly reduced intracellular TG and lipid droplets during the differentiation of preadipocytes. Quinizarin significantly suppressed the expression of adipocyte differentiation marker proteins, such as CCAAT/enhancer-binding protein ß (C/EBP-ß), C/EBP-α, PPAR-γ, and aP2, and lipogenic marker proteins, including SREBP1c, SREBP2, fatty acid synthase (FAS), and acetyl-CoA carboxylase 1 (ACC1), reduced ACC2 expression and increased carnitine palmitoyltransferase 1 (CPT1) expression. Oral administration of quinizarin (15-30 mg/kg/day) to HD-fed mice for 6 weeks reduced the body weight gain and size of liver adipocytes and epididymal fat tissues, with significant reductions in liver TG and serum total cholesterol, blood glucose, LDL, and HDL levels. SIGNIFICANCE: The results of this study indicated that quinizarin exerts anti-obesity effects by inhibiting both adipogenesis and lipogenesis and stimulating lipolysis in vitro and in vivo mainly by downregulating the SREBP signalling pathway; thus, it might be a potent candidate as a health-beneficial food or therapeutic agent to prevent or treat obesity.


Subject(s)
Adipocytes/metabolism , Anthraquinones/pharmacology , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , Cell Differentiation/physiology , Lipogenesis/physiology , Sterol Regulatory Element Binding Proteins/antagonists & inhibitors , 3T3-L1 Cells , Adipocytes/drug effects , Adipogenesis/drug effects , Adipogenesis/physiology , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Lipogenesis/drug effects , Male , Mice , Mice, Inbred C57BL , Sterol Regulatory Element Binding Proteins/metabolism
18.
Int J Mol Sci ; 22(21)2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34769029

ABSTRACT

Although microRNAs (miRNAs or miRs) have been studied in the peripheral nervous system, their function in Schwann cells remains elusive. In this study, we performed a microRNA array analysis of cyclic adenosine monophosphate (cAMP)-induced differentiated primary Schwann cells. KEGG pathway enrichment analysis of the target genes showed that upregulated miRNAs (mR212-5p, miR335, miR20b-5p, miR146b-3p, and miR363-5p) were related to the calcium signaling pathway, regulation of actin cytoskeleton, retrograde endocannabinoid signaling, and central carbon metabolism in cancer. Several key factors, such as purinergic receptors (P2X), guanine nucleotide-binding protein G(olf) subunit alpha (GNAL), P2RX5, P2RX3, platelet-derived growth factor receptor alpha (PDGFRA), and inositol 1,4,5-trisphosphate receptor type 2 (ITPR2; calcium signaling pathway) are potential targets of miRNAs regulating cAMP. Our analysis revealed that miRNAs were differentially expressed in cAMP-treated Schwann cells; miRNA363-5p was upregulated and directly targeted the P2X purinoceptor 4 (P2RX4)-UTR, reducing the luciferase activity of P2RX4. The expression of miRNA363-5p was inhibited and the expression of P2RX4 was upregulated in sciatic nerve injury. In contrast, miRNA363-5p expression was upregulated and P2RX4 expression was downregulated during postnatal development. Of note, a P2RX4 antagonist counteracted myelin degradation after nerve injury and increased pERK and c-Jun expression. Interestingly, a P2RX4 antagonist increased the levels of miRNA363-5p. This study suggests that a double-negative feedback loop between miRNA363-5p and P2RX4 contributes to the dedifferentiation and migration of Schwann cells after nerve injury.


Subject(s)
MicroRNAs/metabolism , Peripheral Nerve Injuries/metabolism , Receptors, Purinergic P2X4/metabolism , Schwann Cells/metabolism , Animals , Cells, Cultured , Down-Regulation/physiology , Female , Mice , Mice, Inbred C57BL , Myelin Sheath/metabolism , Phenotype , Pregnancy , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Up-Regulation/physiology
19.
J Neuroinflammation ; 18(1): 246, 2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34711216

ABSTRACT

BACKGROUND: Cognitive deficits are common in patients with sepsis. Previous studies in sepsis-associated encephalopathy (SAE) implicated the C-X-C chemokine receptor type (CXCR) 5. The present study used a mouse model of SAE to examine whether CXCR5 down-regulation could attenuate cognitive deficits. METHODS: Sepsis was induced in adult male C57BL/6 J and CXCR5-/- mice by cecal ligation and puncture (CLP). At 14-18 days after surgery, animals were tested in a Morris water maze, followed by a fear conditioning test. Transmission electron microscopy of hippocampal sections was used to assess levels of autophagy. Primary microglial cultures challenged with lipopolysaccharide (LPS) were used to examine the effects of short interfering RNA targeting CXCR5, and to investigate the possible involvement of the p38MAPK/NF-κB/STAT3 signaling pathway. RESULTS: CLP impaired learning and memory and up-regulated CXCR5 in hippocampal microglia. CLP activated hippocampal autophagy, as reflected by increases in numbers of autophagic vacuoles, conversion of microtubule-associated protein 1 light chain 3 (LC3) from form I to form II, accumulation of beclin-1 and autophagy-related gene-5, and a decrease in p62 expression. CLP also shifted microglial polarization to the M1 phenotype, and increased levels of IL-1ß, IL-6 and phosphorylated p38MAPK. CXCR5 knockout further enhanced autophagy but partially reversed all the other CLP-induced effects, including cognitive deficits. Similar effects on autophagy and cytokine expression were observed after knocking down CXCR5 in LPS-challenged primary microglial cultures; this knockdown also partially reversed LPS-induced up-regulation of phosphorylated NF-κB and STAT3. The p38MAPK agonist P79350 partially reversed the effects of CXCR5 knockdown in microglial cultures. CONCLUSIONS: CXCR5 may act via p38MAPK/NF-κB/STAT3 signaling to inhibit hippocampal autophagy during sepsis and thereby contribute to cognitive dysfunction. Down-regulating CXCR5 can restore autophagy and mitigate the proinflammatory microenvironment in the hippocampus.


Subject(s)
Cognitive Dysfunction/metabolism , NF-kappa B/metabolism , Receptors, CXCR5/deficiency , STAT3 Transcription Factor/metabolism , Sepsis-Associated Encephalopathy/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Autophagy/physiology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/prevention & control , Down-Regulation/physiology , Male , Maze Learning/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , NF-kappa B/genetics , Receptors, CXCR5/genetics , STAT3 Transcription Factor/genetics , Sepsis-Associated Encephalopathy/genetics , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/genetics
20.
Medicine (Baltimore) ; 100(42): e27489, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34678879

ABSTRACT

ABSTRACT: Tissue inhibitor of metalloproteinases 2 (TIMP2) is a member of the TIMP gene family. Accumulated evidence indicates that TIMP2 plays a significant role in various tumor processes including cell growth, apoptosis, invasion, and metastasis. However, the expression patterns and exact roles of TIMP2 had not been elucidated in breast cancer. In our research, we evaluated the expression and prognostic value of TIMP2 in breast cancer through analyzing various databases including Oncomine, bc-GenExMiner, PrognoScan, UCSC Xena, Kaplan-Meier Plotter, and PPI network. The results showed that TIMP2 was down-regulated in various breast cancer subtypes. Additionally, TIMP2 was significantly associated with age, estrogen receptor status, basal-like group, triple-negative breast cancer, PAM50 subtypes, and RSSPC subtypes. Also, the expression of TIMP2 was related to overall survival with different clinical characteristics. We analyzed the co-expressed genes with TIMP2 and interaction information with other proteins. These results disclosed that TIMP2 might serve as a potential target and prognostic biomarker in breast cancer. However, additional research is required to demonstrate our findings and motivate the clinical importance of TIMP2 in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Age Factors , Biomarkers, Tumor , Computational Biology , Down-Regulation/physiology , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Prognosis , Receptors, Estrogen/biosynthesis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL