Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Mol Cancer ; 23(1): 39, 2024 02 21.
Article in English | MEDLINE | ID: mdl-38378518

ABSTRACT

BACKGROUND: Focal adhesion signaling involving receptor tyrosine kinases (RTK) and integrins co-controls cancer cell survival and therapy resistance. However, co-dependencies between these receptors and therapeutically exploitable vulnerabilities remain largely elusive in HPV-negative head and neck squamous cell carcinoma (HNSCC). METHODS: The cytotoxic and radiochemosensitizing potential of targeting 10 RTK and ß1 integrin was determined in up to 20 3D matrix-grown HNSCC cell models followed by drug screening and patient-derived organoid validation. RNA sequencing and protein-based biochemical assays were performed for molecular characterization. Bioinformatically identified transcriptomic signatures were applied to patient cohorts. RESULTS: Fibroblast growth factor receptor (FGFR 1-4) targeting exhibited the strongest cytotoxic and radiosensitizing effects as monotherapy and combined with ß1 integrin inhibition, exceeding the efficacy of the other RTK studied. Pharmacological pan-FGFR inhibition elicited responses ranging from cytotoxicity/radiochemosensitization to resistance/radiation protection. RNA sequence analysis revealed a mesenchymal-to-epithelial transition (MET) in sensitive cell models, whereas resistant cell models exhibited a partial epithelial-to-mesenchymal transition (EMT). Accordingly, inhibition of EMT-associated kinases such as EGFR caused reduced adaptive resistance and enhanced (radio)sensitization to FGFR inhibition cell model- and organoid-dependently. Transferring the EMT-associated transcriptomic profiles to HNSCC patient cohorts not only demonstrated their prognostic value but also provided a conclusive validation of the presence of EGFR-related vulnerabilities that can be strategically exploited for therapeutic interventions. CONCLUSIONS: This study demonstrates that pan-FGFR inhibition elicits a beneficial radiochemosensitizing and a detrimental radioprotective potential in HNSCC cell models. Adaptive EMT-associated resistance appears to be of clinical importance, and we provide effective molecular approaches to exploit this therapeutically.


Subject(s)
Antineoplastic Agents , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Integrin beta1/genetics , Cell Line, Tumor , Receptor Protein-Tyrosine Kinases/genetics , Antineoplastic Agents/therapeutic use , ErbB Receptors/metabolism , Phenotype , Epithelial-Mesenchymal Transition/genetics
3.
J Cell Commun Signal ; 17(1): 209-215, 2023 Mar.
Article in English | MEDLINE | ID: mdl-35838944

ABSTRACT

Interleukin-17A (IL-17A) is one of the member of IL-17 family consisting of other five members (IL-17B to IL-17F). The Gamma delta (γδ) T cells and T helper 17 (Th17) cells are the major producers of IL-17A. Aberrant signaling by IL-17A has been implicated in the pathogenesis of several autoimmune diseases including idiopathic pulmonary fibrosis, acute lung injury, chronic airway diseases, and cancer. Activation of the IL-17A/IL-17 receptor A (IL-17RA) system regulates phosphoinositide 3-kinase/AKT serine/threonine kinase/mammalian target of rapamycin (PI3K/AKT/mTOR), mitogen-activated protein kinases (MAPKs) and activation of nuclear factor-κB (NF-κB) mediated signaling pathways. The IL-17RA activation orchestrates multiple downstream signaling cascades resulting in the release of pro-inflammatory cytokines such as interleukins (IL)-1ß, IL-6, and IL-8, chemokines (C-X-C motif) and promotes neutrophil-mediated immune response. Considering the biomedical importance of IL-17A, we developed a pathway resource of signaling events mediated by IL-17A/IL-17RA in this study. The curation of literature data pertaining to the IL-17A system was performed manually by the NetPath criteria. Using data mined from the published literature, we describe an integrated pathway reaction map of IL-17A/IL-17RA consisting of 114 proteins and 68 reactions. That includes detailed information on IL-17A/IL-17RA mediated signaling events of 9 activation/inhibition events, 17 catalysis events, 3 molecular association events, 68 gene regulation events, 109 protein expression events, and 6 protein translocation events. The IL-17A signaling pathway map data is made freely accessible through the WikiPathways Database ( https://www.wikipathways.org/index.php/Pathway : WP5242).

4.
Article in English | MEDLINE | ID: mdl-35642119

ABSTRACT

AIM: This study aimed to assess the role of Tight junction proteins (TJPs) and claudins in smokers with and without COPD compared to healthy individuals. BACKGROUND: Chronic obstructive pulmonary disease (COPD) is a complex chronic respiratory disease, including various inflammatory mediators. The prime etiological element in the development of COPD is cigarette smoking. The lung airway epithelium comprises beneficial immunological barriers to draw in insults, such as environmental particulates, cigarette smoke, etc. Tight junctions (TJ) connected by transmembrane proteins determine epithelial permeability. Cigarette smoke is indicated to defect TJ integrity. The possible involvement of the airway epithelium in the pathogenesis of COPD has recently become apparent; however, its detailed mechanisms remain elusive. The integrity of airway epithelium is crucial for airway homeostasis; defective airway barrier activity contributes to COPD. OBJECTIVE: In the present study, the objective was to investigate mRNA expression levels of TJP's like TJP-1, TJP-2, TJP-3, Tight junction-associated proteins-1, claudin-1, claudin-3, claudin-4, claudin-7, claudin-10, claudin-15, claudin-19, and claudin-25 from blood samples of smokers with COPD and compared them with smokers without COPD and healthy individuals. METHODS: The mRNA expressions were evaluated by the quantitative PCR method. RESULTS: The gene expressions of these TJPs were significantly down-regulated, specifically in COPD patients with a history of smoking (Smokers with COPD). Besides, FEV% was also established for these patients. Similarly, smokers with COPD showed a significant increase in the expression levels of transcription factors, like ZEB-1, ZEB-2, PDGFA, and HDGF, compared to COPD patients without a history of smoking (smokers without COPD) and the healthy subjects. CONCLUSION: In conclusion, cigarette smoke disrupts TJ of the human airway epithelium, and the transcriptional factors counteract this smoke-induced COPD. Thus, TJPs may serve as protective elements for airway epithelial homeostasis during COPD.


Subject(s)
Pulmonary Disease, Chronic Obstructive , Tight Junction Proteins , Humans , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Smokers , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Claudins/genetics , Claudins/metabolism , Nicotiana , Blood Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Nat Immunol ; 23(4): 518-531, 2022 04.
Article in English | MEDLINE | ID: mdl-35354953

ABSTRACT

Internal organs heal injuries with new connective tissue, but the cellular and molecular events of this process remain obscure. By tagging extracellular matrix around the mesothelium lining in mouse peritoneum, liver and cecum, here we show that preexisting matrix was transferred across organs into wounds in various injury models. Using proteomics, genetic lineage-tracing and selective injury in juxtaposed organs, we found that the tissue of origin for the transferred matrix likely dictated the scarring or regeneration of the healing tissue. Single-cell RNA sequencing and genetic and chemical screens indicated that the preexisting matrix was transferred by neutrophils dependent on the HSF-integrin AM/B2-kindlin3 cascade. Pharmacologic inhibition of this axis prevented matrix transfer and the formation of peritoneal adhesions. Matrix transfer was thus an early event of wound repair and provides a therapeutic window to dampen scaring across a range of conditions.


Subject(s)
Neutrophils , Peritoneum , Animals , Epithelium , Extracellular Matrix , Mice , Peritoneum/injuries , Wound Healing
6.
J Proteome Res ; 19(8): 2950-2963, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32618472

ABSTRACT

Bleomycin (BLM)-induced pulmonary fibrosis is characterized by inflammation in the alveoli, subsequent deposition of extracellular matrix (ECM) and myofibroblasts, and an impaired fibrinolytic system. Here, we describe major hematological changes, the IL-17A-mediated p53-fibrinolytic pathway, and the high throughput hits of liquid chromatography-mass spectrometry (LC-MS) analysis during the progression of pulmonary fibrosis and the therapeutic potential of curcumin against disease progression. C57BL/6 mice were exposed to BLM, followed by curcumin intervention after 24 and 48 h. Mice were sacrificed after 7 days to validate the hematological parameters, molecular pathways, and proteomics. Various techniques such as western blotting, immunofluorescence, reverse transcriptase polymerase chain reaction (RT-PCR), hematoxylin and eosin staining, Masson's trichrome staining, and immunohistochemistry were used to validate the proposed theory. LC-MS analysis was performed using a Q-Orbitrap mass spectrometer. The Schrödinger approach was used to perform the in silico molecular docking studies. BLM-exposed mice exhibited gradual weight loss and altered lung morphology; however, these were reversed by curcumin treatment. Significant changes in the hematological parameters confirmed the severity of BLM exposure in the mice, and expression of IL-17A-mediated p53-fibrinolytic system components and alveolar epithelial cell (AEC) apoptosis further confirmed the pathophysiology of pulmonary fibrosis. Differentially expressed proteins were characterized and mapped using the proteomics approach. A strong interaction of curcumin is observed with p53, uPA, and PAI-I proteins. The key role of IL-17A-mediated inflammation in the impairment of the p53-fibrinolytic system and AEC apoptosis was confirmed during BLM-induced pulmonary fibrosis. Therapeutic efficacy of curcumin exhibited a protective role against the progression of pulmonary fibrosis, which promises potent therapeutic modality to target the IL-17A-mediated p53-fibrinolytic system during pulmonary fibrosis.


Subject(s)
Curcumin , Pulmonary Fibrosis , Animals , Bleomycin/toxicity , Curcumin/pharmacology , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/genetics , Lung , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Proteomics , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy
7.
Inflammation ; 42(4): 1160-1169, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31011925

ABSTRACT

Acute lung injury (ALI) is characterized by acute inflammation and tissue injury results in dysfunction of the alveolar epithelial membrane. If the epithelial injury is severe, a fibroproliferative phase of ALI can develop. During this phase, the activated fibroblast and myofibroblasts synthesize excessive collagenous extracellular matrix that leads to a condition called pulmonary fibrosis. Lung injury can be caused by several ways; however, the present review focus on bleomycin (BLM)-mediated changes in the pathology of lungs. BLM is a chemotherapeutic agent and has toxic effects on lungs, which leads to oxidative damage and elaboration of inflammatory cytokines. In response to the injury, the inflammatory cytokines will be activated to defend the system from injury. These cytokines along with growth factors stimulate the proliferation of myofibroblasts and secretion of pathologic extracellular matrix. During BLM injury, the pro-inflammatory cytokine such as IL-17A will be up-regulated and mediates the inflammation in the alveolar epithelial cell and also brings about recruitment of certain inflammatory cells in the alveolar surface. These cytokines probably help in up-regulating the expression of p53 and fibrinolytic system molecules during the alveolar epithelial cells apoptosis. Here, our key concern is to provide the adequate knowledge about IL-17A-mediated p53 fibrinolytic system and their pathogenic progression to pulmonary fibrosis. The present review focuses mainly on IL-17A-mediated p53-fibrinolytic aspects and how curcumin is involved in the regulation of pathogenic progression of ALI and pulmonary fibrosis.


Subject(s)
Acute Lung Injury/pathology , Curcumin/pharmacology , Inflammation/metabolism , Interleukin-17/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Alveolar Epithelial Cells/pathology , Animals , Bleomycin/pharmacology , Humans , Pulmonary Fibrosis/etiology , Tumor Suppressor Protein p53/metabolism
8.
Mol Biol Rep ; 45(6): 2835-2841, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30250995

ABSTRACT

COPD is a chronic airway inflammatory disease characterized mainly by neutrophil airway infiltrations. The neutrophil airway inflammation is mainly mediated through a key player like the pro-inflammatory cytokine IL-17A which is involved in the modulation of p53-fibrinolytic system. This study was undertaken to examine the molecular changes for the expressions of IL-17A and p53-fibrinolytic system in smokers with or without COPD. Blood and serum samples were collected from ten patients of smokers having COPD and ten samples from smokers without COPD and ten healthy control subjects. Western blot analyses were performed to evaluate the expressions of IL-17A, p53 and PAI-1. Apoptosis was assessed by immunoblot for cleaved caspase-3. In addition, FEV% was also determined of these patients. qRT-PCR was done to detect the gene expression study from the blood samples on p53-fibrinolytic components. A significant difference was found in the expression levels of IL-17A in smokers with COPD patient when compared to smokers without COPD and the control subjects. Similarly the smokers with COPD showed significant increase in the fibrinolytic component PAI-1 as well as in expression levels of p53 when compared to smokers without COPD and normal subjects. Increased cleaved caspase-3 may also promote apoptosis.The expression pattern of the IL-17A in chronic obstructive pulmonary distress syndrome samples was increased as compared of those of normal samples, and their main role in the regulation of and p53-fibrinolytic system makes these components as a predictive prominent component in smokers with COPD.


Subject(s)
Interleukin-17/biosynthesis , Pulmonary Disease, Chronic Obstructive/blood , Smoking/blood , Tumor Suppressor Protein p53/biosynthesis , Adult , Aged , Apoptosis/physiology , Case-Control Studies , Female , Fibrinolysis , Humans , Interleukin-17/blood , Interleukin-17/genetics , Male , Middle Aged , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/blood , Plasminogen Activator Inhibitor 1/genetics , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/genetics , Smoking/adverse effects , Smoking/genetics , Tumor Suppressor Protein p53/blood , Tumor Suppressor Protein p53/genetics , Urokinase-Type Plasminogen Activator/blood
9.
Lung ; 196(5): 609-616, 2018 10.
Article in English | MEDLINE | ID: mdl-30121847

ABSTRACT

Acute respiratory distress syndrome (ARDS) is the most advanced form of acute lung injury (ALI). This is characterized by bilateral pulmonary infiltrates and severe hypoxemia. According to Berlin definition of ARDS, this is defined based on the timings, radiographic changes, edema formation, and severity on the PaO2/FiO2 ratio. During ARDS, the loss of integrity of the epithelium causes the septic shock. The degree of epithelial injury is the major prognostic marker of ARDS. In addition to this, inflammatory cell migration, fibro-proliferation, and activation of apoptosis also play an important role in the pathophysiology of ARDS. The alveolar epithelial cell is the prime target during injury where this cell either undergo apoptosis or epithelial-mesenchymal transition (EMT). Injury to the AECs triggers the changes in the DNA fragmentation and activation of certain apoptotic markers such as caspases at the same time some cells undergo biochemical changes and loses its epithelial morphology as well epithelial biomarkers and gain mesenchymal biomarkers and morphology. In both the cases, the fibrinolytic system plays an important role in maintaining the integrity of the disease process efficiently. This review highlights the research evidence of apoptosis and EMT in lung development, injury and its prognosis in ARDS thereby to develop an effective strategy for the treatment of ARDS.


Subject(s)
Apoptosis , Epithelial-Mesenchymal Transition , Fibrinolysis , Inflammation/immunology , Respiratory Distress Syndrome/immunology , Respiratory Mucosa/immunology , Acute Lung Injury/immunology , Acute Lung Injury/metabolism , Cytokines/immunology , Humans , Inflammation/metabolism , Pulmonary Alveoli/cytology , Respiratory Distress Syndrome/metabolism , Respiratory Mucosa/metabolism
10.
Mol Biol Rep ; 45(4): 645-650, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29808357

ABSTRACT

Pro-inflammatory cytokine IL-17A modulates the expression of Akt in bleomycin (BLM) administered alveolar basal epithelial cells, the mechanism behind which remains unclear. This investigation was carried out to assess IL-17A mediated down-regulation of Akt expression and the pivotal role of curcumin as a regulatory molecule. Alveolar basal epithelial cells were treated with BLM and IL-17A and curcumin was administered as an intervention to regulate the BLM-induced oxidative damage. Cell proliferation was evaluated by clonogenic assay. Akt phosphorylation and total Akt expressions were studied using western blot analysis. Cell proliferation reduced upon treatment with BLM and this phenomenon was reversed in cells upon administration with curcumin. Administrations of BLM and IL-17A to the alveolar basal epithelial cells showed significant down-regulation of Akt expression which was reversed by treatment with curcumin. BLM and IL-17A mediated inflammation was intervened effectively with curcumin. Results of this study suggest the probable use of curcumin as an anti-inflammatory therapeutic for lung injury.


Subject(s)
Curcumin/pharmacology , Interleukin-17/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Apoptosis/drug effects , Bleomycin/pharmacology , Cell Proliferation/drug effects , Curcumin/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Interleukin-17/physiology , Lung/drug effects , Lung Injury/drug therapy , Transcriptional Activation/drug effects , Up-Regulation
11.
J Cell Biochem ; 119(9): 7285-7299, 2018 09.
Article in English | MEDLINE | ID: mdl-29775223

ABSTRACT

Bleomycin (BLM) induced cellular damage causes inflammation in the alveolar compartment and impairment of fibrinolytic system leads to alveolar epithelial cell apoptosis. Here, we describe novel inflammatory pathway associated with p53-fibrinolytic system and apoptosis of alveolar epithelial cells and pharmacological efficiency of curcumin against this action. In the present study we used C57BL/6 mice. The specific dose and time interval of curcumin were analyzed to assess the intervention. Experiments were designed to investigate the IL-17A mediated modulation in the alveolar epithelial cell apoptosis and injury. Various techniques such as Western blot, RT-PCR, Immunohistochemistry were used for this study. We observed that the BLM-induced lung injury and its progression were successfully regulated by the effective dose and time intervention of curcumin. There was also decreased expression of chemokines, p53, and fibrinolytic components such as PAI-1 and increased uPA, uPAR expression, and decreased alveolar epithelial cell apoptosis, which indicates the IL-17A mediated novel inflammatory pathway. It is confirmed that the IL-17A involved in the modulation of p53-fibrinolytic system and epithelial cell apoptosis in BLM induced mice. The cross-talk between the inflammatory, fibrinolytic, and apoptotic pathways were resolved by curcumin intervention. This pathway and intervention could serve as a modern therapy to resolve the complications to cure the lung injury and its progression.


Subject(s)
Acute Lung Injury/chemically induced , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibiotics, Antineoplastic/adverse effects , Bleomycin/adverse effects , Curcumin/pharmacology , Interleukin-17/metabolism , Tumor Suppressor Protein p53/metabolism , Acute Lung Injury/prevention & control , Alveolar Epithelial Cells/metabolism , Analysis of Variance , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Apoptosis/drug effects , Caspase 3/metabolism , Curcumin/administration & dosage , Curcumin/therapeutic use , Disease Models, Animal , Down-Regulation , Fibrosis/metabolism , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Time Factors
12.
J Cell Biochem ; 119(2): 2222-2230, 2018 02.
Article in English | MEDLINE | ID: mdl-28902433

ABSTRACT

Bleomycin-mediated inflammatory pathway is known to play an important role in the up regulation of oxidative stress. IL-17A is a pro-inflammatory cytokine involved in the modulation of fibrosis. The complex underlying mechanism for the said phenomenon remains unclear. This newly defined investigation was designed to understand the changes associated with 1L-17A mediated up-regulation of p53 and PAI-1 expression and the role of curcumin in attenuating this process. A549 cells were treated with bleomycin (BLM) and IL-17A to induce the inflammatory response in vitro. Curcumin, a known anti-inflammatory bioactive compound was administered as an intervention. Cytotoxicity in the treatment groups was assessed using Methyl thiazolyl tetrazolium (MTT) assay. Cell migration was evaluated using scratch assay. Protein expressions were studied using Western blot analysis for the downstream effector molecules of IL-17A mediated inflammatory pathways. In MTT assay, BLM treatment showed cytotoxicty upto 88% at a concentration of 1000 µM after 48 h of treatment. Cell migration assay results revealed that curcumin blocked the migration of cells to the area of the scratch. BLM treatment to the cells significantly induced the expression of pro-inflammatory cytokine IL-17A, which in turn modulated p53-PAI-1 expression. Bioactive compound curcumin showed anti-inflammatory and anti-apoptotic activity. Curcumin also regulated the BLM and IL-17A mediated changes in p53-PAI-1 expression. Curcumin has the ability to regulate inflammatory cytokines during BLM-induced injury and their effect on p53-PAI-1 expression. It can be used as a potential anti-inflammatory and anti-fibrinolytic component for intervening the epithelial cell damage.Very little information is provided till date on the inflammatory mechanism controlling the fibrinolytic system in acute lung injury (ALI). Damage to alveolar epithelial cells during ALI is important in the development of pulmonary fibrosis (PF). Most forms of ALI are characterized by defective alveolar fibrinolysis, inflammation, and fibrotic lesions. Recent reports show that alveolar epithelial cells express uPA, uPAR, and p53-mediated changes inhibit epithelial cell viability contributing to ALI. Thus, the roles of pulmonary epithelial cells in the inflammatory cascades activated after noninfectious injury, and the key signaling mediators of this process were actively investigated in this study. This investigation revealed that curcumin is an effective inhibitor of BLM-induced inflammation, apoptosis, and migration of basal alveolar epithelial cells. These results throw an insight into the possibility of developing curcumin as a novel therapeutic for ALI.


Subject(s)
Alveolar Epithelial Cells/cytology , Bleomycin/adverse effects , Interleukin-17/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Tumor Suppressor Protein p53/metabolism , A549 Cells , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/metabolism , Apoptosis/drug effects , Cell Movement/drug effects , Gene Expression Regulation/drug effects , Humans
13.
Appl Physiol Nutr Metab ; 43(4): 371-380, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29099999

ABSTRACT

Given the association between subclinical hypothyroidism and metabolic syndrome, we wanted to explore if high-fat, simple-carbohydrate (HFSC) diet affects hypothalamus-pituitary-thyroid axis. One-month-old male C57BL/6J mice were fed with control (C) and HFSC (T) feed (n = 18 each), respectively, for 5 months. There was a significant increase in triiodothyronine in the T group (13.5%) compared with the age-matched C group by the fifth month. Thyroid-stimulating hormone was significantly higher (1 month: 1.9-fold; 3 months: 2.66-fold; 5 months: 3.5-fold) from the first to fifth months in the T group compared with age-matched C group. Thyrotropin-releasing hormone (TRH) gene expression showed significant decrease (1 month: 83.2%; 5 months: 40.7%) in the T group compared with the age-matched C group. TRHR1 showed significant decrease in the T group compared with the age-matched C group throughout the study (1 month: 82.8%; 3 months: 45.7%; 5 months: 75.2%). However, TRHR2 showed dynamic change during the study. Initially there was significant (1 month: 0.104-fold) downregulation, followed by significant upregulation (3 months: 3.6-fold) and downregulation (0.73-fold) by the fifth month in the T group compared with the age-matched C group. There was marked depletion of functional follicular cells and colloid substance in the thyroid glands of the T group by the fifth month compared with the C group. Leptin receptors ObRa (1 month: 48.25%; 5 months: 88%) and ObRb (1 month: 46.9%; 5 months: 63.3%) were significantly downregulated in the T group compared with the age-matched C group in the first and fifth months of feeding the respective diets. The expression of p-STAT3, a transcription factor known to have a role in energy balance, intermediate metabolism, and leptin signalling was seen to decrease significantly (6.25-fold) in the hypothalamus of the T group compared with the age-matched C group. In conclusion, HFSC feed disrupts the hypothalamus-pituitary-thyroid axis in male C57BL/6J mice.


Subject(s)
Diet, High-Fat , Dietary Carbohydrates , Hypothalamo-Hypophyseal System/metabolism , Thyroid Gland/metabolism , Animals , Hypothalamo-Hypophyseal System/physiopathology , Male , Mice, Inbred C57BL , Phosphorylation , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Receptors, Thyrotropin-Releasing Hormone/genetics , Receptors, Thyrotropin-Releasing Hormone/metabolism , STAT3 Transcription Factor/metabolism , Thyroid Gland/physiopathology , Thyroid Gland/ultrastructure , Thyrotropin/blood , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/metabolism , Time Factors , Triiodothyronine/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...