84 results on '"Ezhilarasan, Devaraj"'
Search Results
2. Thyromimetics and MASLD: Unveiling the Novel Molecules Beyond Resmetirom.
- Author
-
Ezhilarasan D
- Abstract
Background: Resmetirom, the first FDA-approved drug for nonalcoholic steatohepatitis (NASH) with fibrosis in obese patients, when combined with lifestyle modifications, improves NASH resolution and reduces fibrosis by at least one stage. Low thyroid hormone (T
3 ) levels are linked to a higher risk of developing metabolic dysfunction-associated steatotic liver disease (MASLD). Epidemiological studies have confirmed the positive correlation between hypothyroidism and MASLD. Unraveling the molecular mechanisms of T3 signaling pathways in MASLD will enhance the prospects of identifying effective and specific targets. Therefore, this review discusses the significant role of thyroid hormones in the homeostasis of fat metabolism and describes the possible molecular mechanisms of thyromimetics in the treatment of MASLD., Methods: A comprehensive search in PubMed and EMBASE was conducted using the keywords "thyromimetics and liver diseases," "thyroid hormone and liver diseases," "hypothyroidism and liver diseases," "T3 , T4 and liver disease," and "resmetirom and liver disease." Relevant papers published before October 2024 were included., Results: T3 treatment enhances mitochondrial respiration, biogenesis, β-oxidation, and mitophagy, reducing liver lipid accumulation. However, T3 treatment causes cardiotoxicity through thyroid hormone receptor (THR)α agonistic activity. To address this, molecules with high THRβ agonistic but lower THRα activity have been developed. Besides resmetirom, other THRβ agonists like TG68, CS27109, MB07811, and KB-141 show promising results in experimental studies. These molecules upregulate THRβ target genes, activate genes for fatty acid β-oxidation in mitochondria and fatty acid breakdown in peroxisomes, downregulate the genes involved in de novo lipogenesis, reduce inflammation by downregulating NF-κB/JNK/STAT3 signaling pathways, and accelerate fibrosis resolution by downregulating the expressions of fibrosis marker genes in NASH liver tissue., Conclusion: Future clinical studies should thoroughly investigate THRβ agonists, including TG68, CS27109, MB07811, and KB-141., (© 2025 Journal of Gastroenterology and Hepatology Foundation and John Wiley & Sons Australia, Ltd.)- Published
- 2025
- Full Text
- View/download PDF
3. Selecting Preclinical Animal Models in Hepatology Research: A Call for Uniform Guidelines.
- Author
-
Ezhilarasan D and Najimi M
- Published
- 2025
- Full Text
- View/download PDF
4. Four years after the ranitidine withdrawal by FDA: Is it time for India to act?
- Author
-
Ezhilarasan D
- Published
- 2025
- Full Text
- View/download PDF
5. Coleus vettiveroides Root Extract Protects Against Thioacetamide-Induced Chronic Liver Injury by Inhibiting NF-κB Signaling Pathway.
- Author
-
Mohamed Azar KAH, Ezhilarasan D, Karthick M, Shree Harini K, and Kumar V
- Abstract
The roots of Coleus vettiveroides (CV) have been traditionally used in Indian medicinal systems such as Ayurveda and Siddha for its antioxidant, anti-inflammatory, and antidiabetic effects. This study examines the antifibrotic potential of CV ethanolic root extract (CVERE) against thioacetamide (TAA)-induced liver fibrosis in Wistar rats. TAA was administered via i.p., thrice weekly for 11 weeks to induce liver fibrosis in rats. In separate groups, rats were administered with TAA and were concurrently treated with CVERE 125 mg/kg, CVERE 250 mg/kg, and silymarin (SIL) 100 mg/kg. Liver marker enzymes of hepatotoxicity, oxidative stress markers, proinflammatory marker gene expression (TNF-α, NF-κB, COX, and ILs), fibrotic marker gene expression (collagen I and III), immune histochemical expression of fibrosis marker proteins, and histopathologic changes were analyzed. TAA administration led to a significant (p < 0.001) increase in the serum level of hepatotoxic marker enzymes. The TAA-treated group showed higher levels (p < 0.001) of MDA and reduced activities of SOD and CAT in the liver. TAA administration increased CYP2E1 expression, proinflammatory, and fibrotic marker gene expressions in rat liver. The histopathology of the liver confirms TAA-induced architectural distortion and fibrotic changes. CVERE and SIL simultaneous treatments significantly protected against TAA-induced oxidative stress, inflammation, and liver fibrosis. In conclusion, CVERE inhibited TAA-induced liver fibrosis through downregulation of TAA metabolic activation, redox imbalance, and inflammation through repression of the NF-κB pathway., (© 2024 Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
6. Preclinical liver toxicity models: Advantages, limitations and recommendations.
- Author
-
Ezhilarasan D, Karthikeyan S, Najimi M, Vijayalakshmi P, Bhavani G, and Jansi Rani M
- Abstract
Experimental animal models are crucial for elucidating the pathophysiology of liver injuries and for assessing new hepatoprotective agents. Drugs and chemicals such as acetaminophen, isoniazid, valproic acid, ethanol, carbon tetrachloride (CCl
4 ), dimethylnitrosamine (DMN), and thioacetamide (TAA) are metabolized by the CYP2E1 enzyme, producing hepatotoxic metabolites that lead to both acute and chronic liver injuries. In experimental settings, acetaminophen (centrilobular necrosis), carbamazepine (centrilobular necrosis and inflammation), sodium valproate (necrosis, hydropic degeneration and mild inflammation), methotrexate (sinusoidal congestion and inflammation), and TAA (centrilobular necrosis and inflammation) are commonly used to induce various types of acute liver injuries. Repeated and intermittent low-dose administration of CCl4 , TAA, and DMN activates quiescent hepatic stellate cells, transdifferentiating them into myofibroblasts, which results in abnormal extracellular matrix production and fibrosis induction, more rapidly with DMN and CCL4 than TAA (DMN > CCl4 > TAA). Regarding toxicity and mortality, CCl4 is more toxic than DMN and TAA (CCl4 > DMN > TAA). Models used to induce metabolic dysfunction-associated liver disease (MAFLD) vary, but MAFLD's multifactorial nature driven by factors like obesity, fatty liver, dyslipidaemia, type II diabetes, hypertension, and cardiovascular disease makes it challenging to replicate human metabolic dysfunction-associated steatohepatitis accurately. From an experimental point of view, the degree and pattern of liver injury are influenced by various factors, including the type of hepatotoxic agent, exposure duration, route of exposure, dosage, frequency of administration, and the animal model utilized. Therefore, there is a pressing need for standardized protocols and regulatory guidelines to streamline the selection of animal models in preclinical studies., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
7. Beyond resmetirom approval for NAFLD: what has to be done?
- Author
-
Ezhilarasan D
- Published
- 2024
- Full Text
- View/download PDF
8. SNRPE, an Oncofetal Protein: can be a Diagnostic Marker and Therapeutic Target for Oral Cancers?
- Author
-
Sanjay S, Sharmila M, and Ezhilarasan D
- Subjects
- Humans, Mouth Neoplasms pathology, Mouth Neoplasms diagnosis, Biomarkers, Tumor analysis
- Published
- 2024
- Full Text
- View/download PDF
9. Oral submucosal fibrosis: An updated molecular mechanism on pathogenesis and treatment modalities.
- Author
-
Kumar VJ, Ezhilarasan D, and Veeraiyan DN
- Subjects
- Humans, Oral Submucous Fibrosis therapy, Oral Submucous Fibrosis pathology, Oral Submucous Fibrosis etiology
- Published
- 2024
- Full Text
- View/download PDF
10. Hepatocellular Interactions of Potential Nutraceuticals in the Management of Inflammatory NAFLD.
- Author
-
Ezhilarasan D and Langeswaran K
- Subjects
- Humans, Antioxidants pharmacology, Antioxidants therapeutic use, Animals, Non-alcoholic Fatty Liver Disease drug therapy, Non-alcoholic Fatty Liver Disease metabolism, Non-alcoholic Fatty Liver Disease pathology, Dietary Supplements, Inflammation drug therapy, Inflammation metabolism, Inflammation pathology
- Abstract
Numerous studies highlight the potential of natural antioxidants, such as those found in foods and plants, to prevent or treat nonalcoholic fatty liver disease (NAFLD). Inflammation is a key factor in the progression from high-fat diet-induced NAFLD to nonalcoholic steatohepatitis (NASH). Injured liver cells and immune cells release inflammatory cytokines, activating hepatic stellate cells. These cells acquire a profibrogenic phenotype, leading to extracellular matrix accumulation and fibrosis. Persistent fibrosis can progress to cirrhosis. Fatty infiltration, oxidative stress, and inflammation exacerbate fatty liver diseases. Thus, many plant-derived antioxidants, like silymarin, silibinin, curcumin, resveratrol, berberine, and quercetin, have been extensively studied in experimental models and clinical patients with NAFLD. Experimentally, these compounds have shown beneficial effects in reducing lipid accumulation, oxidative stress, and inflammatory markers by modulating the ERK, NF-κB, AMPKα, and PPARγ pathways. They also help decrease metabolic endotoxemia, intestinal permeability, and gut inflammation. Clinically, silymarin and silibinin have been found to reduce transaminase levels, while resveratrol and curcumin help alleviate inflammation in NAFLD patients. However, these phytocompounds exhibit poor water solubility, leading to low oral bioavailability and hindering their biological efficacy. Additionally, inconclusive clinical results highlight the need for further trials with larger populations, longer durations, and standardized protocols., (© 2024 John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
11. Quiescent hepatic stellate cell activation in liver fibrosis: Have we found the right trigger yet?
- Author
-
Ezhilarasan D and Najimi M
- Subjects
- Humans, Hepatic Stellate Cells, Liver Cirrhosis pathology
- Abstract
Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
- Published
- 2024
- Full Text
- View/download PDF
12. Deciphering the molecular pathways of saroglitazar: A dual PPAR α/γ agonist for managing metabolic NAFLD.
- Author
-
Ezhilarasan D
- Subjects
- Humans, Animals, Phenylpropionates therapeutic use, Phenylpropionates pharmacology, Insulin Resistance, Pyrroles, Non-alcoholic Fatty Liver Disease drug therapy, Non-alcoholic Fatty Liver Disease metabolism, PPAR alpha agonists, PPAR alpha metabolism, PPAR gamma agonists, PPAR gamma metabolism
- Abstract
Saroglitazar (SARO), a dual peroxisome proliferator activated receptor (PPAR)-α/γ agonist, has been used to treat metabolic diseases such as insulin resistance and diabetic dyslipidemia in patients with non-alcoholic fatty liver disease (NAFLD). SARO, administered at a dose of 4 mg/day, has been consistently studied in clinical trials with different time points ranging from 4 to 24 weeks with NAFLD patients. Due to its PPAR-γ agonistic action, SARO prevents adipose tissue-mediated fatty acid delivery to the liver by increasing insulin sensitivity and regulating adiponectin and leptin levels in adipose tissue. In hepatocytes, SARO induces fatty acid β-oxidation in mitochondria and transcriptionally activates lipid metabolizing genes in peroxisomes. SARO inhibits insulin resistance, thereby preventing the activation of sterol regulatory element-binding proteins -1c and carbohydrate response element binding protein in hepatocytes through its PPAR-α agonistic action. SARO treatment reduces lipotoxicity-mediated oxidative stress by activating the nuclear factor erythroid 2-related factor 2 and transcriptionally expressing the antioxidants from the antioxidant response element in the nucleus through its PPAR-γ agonistic action. SARO provides a PPAR-α/γ-mediated anti-inflammatory effect by preventing the phosphorylation of mitogen-activated protein kinases (JNK and ERK) and nuclear factor kappa B in hepatocytes. Additionally, SARO interferes with transforming growth factor-β/Smad downstream signaling, thereby reducing liver fibrosis progression through its PPAR-α/γ agonistic actions. Thus, SARO improves insulin resistance and dyslipidemia in NAFLD, reduces lipid accumulation in the liver, and thereby prevents mitochondrial toxicity, oxidative stress, inflammation, and fibrosis progression. This review summarizes the possible molecular mechanism of SARO in the NAFLD., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. Thymoquinone protects thioacetamide-induced chronic liver injury by inhibiting TGF-β1/Smad3 axis in rats.
- Author
-
Raghunandhakumar S, Ezhilarasan D, and Shree Harini K
- Subjects
- Rats, Animals, Thioacetamide toxicity, Antioxidants metabolism, Liver metabolism, Liver Cirrhosis chemically induced, Liver Cirrhosis drug therapy, Liver Cirrhosis prevention & control, Transforming Growth Factor beta metabolism, Inflammation metabolism, Oxidative Stress, Transforming Growth Factor beta1 metabolism, Liver Neoplasms metabolism, Benzoquinones
- Abstract
Chronic liver injury due to various etiological factors results in excess secretion and accumulation of extracellular matrix proteins, leading to scarring of liver tissue and ultimately to hepatic fibrosis. If left untreated, fibrosis might progress to cirrhosis and even hepatocellular carcinoma. Thymoquinone (TQ), an active compound of Nigella sativa, has been reported to exhibit antioxidant, anti-inflammatory and anticancer activities. Therefore, the effect of TQ against thioacetamide (TAA)-induced liver fibrosis was assessed in rats. Fibrosis was induced with intraperitoneal administration of TAA (250 mg/kg b.w.) twice a week for 5 weeks. TQ (20 mg/kg b.w.) and silymarin (50 mg/kg b.w.) were orally administered daily for 5 weeks separately in TAA administered groups. Liver dysfunction was reported by elevated liver enzymes, increased oxidative stress, inflammation and fibrosis upon TAA administration. Our study demonstrated that TQ inhibited the elevation of liver marker enzymes in serum. TQ administration significantly increased antioxidant markers, such as superoxide dismutase, catalase, glutathione, glutathione peroxidase and glutathione reductase in the liver tissue of rats. Further, TQ significantly attenuated liver fibrosis, as illustrated by the downregulation of TAA-induced interleukin-β, tumour necrosis factor-α, inducible nitric oxide synthase and fibrosis markers like transforming growth factor-β (TGF-β), α-smooth muscle actin, collagen-1, Smad3 and 7. Therefore, these findings suggest that TQ has a promising hepatoprotective property, as indicated by its potential to effectively suppress TAA-induced liver fibrosis in rats by inhibiting oxidative stress and inflammation via TGF-β/Smad signaling., (© 2024 Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
14. Boldine protects against carbon tetrachloride-induced chronic liver injury by regulating NF-κB signaling pathway.
- Author
-
Ezhilarasan D, Shree Harini K, Karthick M, and Lavanya P
- Subjects
- Rats, Animals, Antioxidants pharmacology, Antioxidants metabolism, Carbon Tetrachloride toxicity, Actins metabolism, Actins pharmacology, Tumor Necrosis Factor-alpha metabolism, Cyclooxygenase 2 metabolism, Liver metabolism, Signal Transduction, Oxidative Stress, Inflammation metabolism, Anti-Inflammatory Agents pharmacology, NF-kappa B metabolism, Chemical and Drug Induced Liver Injury metabolism, Aporphines
- Abstract
Sustained liver injuries predominantly promote oxidative stress and inflammation that lead to the progression of chronic liver disease (CLD), including fibrosis, cirrhosis, and hepatocellular carcinoma. Boldine, an alkaloid isolated from Peumus boldus, has been shown to have antioxidant and anti-inflammatory effects. Currently, there is no definitive treatment option available for CLD. Therefore, we investigated the hepatoprotective effect of boldine against carbon tetrachloride (CCl
4 )-induced chronic liver injury in rats. CCl4 (2 mL/kg., b.w., i.p.) was administered twice weekly for 5 weeks to induce chronic liver injury in rats. Separate groups of rats were given boldine (20 mg/kg b.w., and 40 mg/kg b.w.) and silymarin (100 mg/kg b.w.) orally, daily. Serum transaminases, lipid peroxidation, and antioxidant levels were measured, and nuclear factor-κB (NF-κB), tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (cox-2), interleukin-1 β (IL-1β), and α-smooth muscle actin (α-SMA) gene and protein expressions were evaluated. CCl4 administration increased liver marker enzymes of hepatotoxicity in serum and oxidative stress markers, inflammatory genes and α-smooth muscle actin expression in liver tissue. Boldine concurrent treatment suppressed CCl4 -induced elevation of transaminase levels in serum, restored enzymic and non-enzymic antioxidants, and downregulated NF-κB, TNF-α, Cox-2 and IL-1β expressions, thereby suppressing hepatic inflammation. Boldine administration also repressed α-SMA expression. The results of this study demonstrate the antioxidant, anti-inflammatory, and antifibrotic properties of boldine, and it can be a potential therapeutic candidate in the treatment of CLD., (© 2024 Wiley Periodicals LLC.)- Published
- 2024
- Full Text
- View/download PDF
15. Coleus vettiveroides ethanolic root extract induces cytotoxicity by intrinsic apoptosis in HepG2 cells.
- Author
-
Mohamed Azar KAH, Ezhilarasan D, and Shree Harini K
- Subjects
- Humans, Hep G2 Cells, Reactive Oxygen Species metabolism, Apoptosis, Ethanol, Carcinoma, Hepatocellular drug therapy, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular pathology, Liver Neoplasms metabolism, Coleus metabolism
- Abstract
Hepatocellular carcinoma (HCC) contributes to more than 80% of all primary cancers globally and ranks fourth in cancer-related deaths, due to the lack of an effective, definite therapeutic drug. Coleus vettiveroides (CV) has been used in Indian traditional medicine to treat diabetes, liver ailments, skin diseases, leukoderma, and leprosy. This study investigates the anticancer effect of CV ethanolic root extract in HepG2 cells. HepG2 cells were treated with CV extract, and its cytotoxicity was analyzed by MTT assay. AO/EB staining, propidium iodide staining, DCFH-DA assay, phalloidine staining, flow cytometry, and qPCR studies were performed for ROS expression, apoptosis and cell cycle analysis. The phytochemical analysis confirmed the presence of quercetin and galangin in CV root extract. The results showed that CV inhibited the proliferation of HepG2 cells, with altered cellular and nuclear morphology. CV was also found to increase intracellular ROS levels and oxidative stress markers in HepG2 cells. CV significantly altered the actin microfilament distribution in HepG2 cells and caused cell cycle arrest at the sub G
0 -G1 phase. CV also induced mitochondria-mediated apoptosis, as evidenced by increased expression of p53, Bax, cytochrome C, Apaf-1, PARP, caspase-3 and caspase-9, and downregulated Bcl-2 expression. Therefore, CV exerts its anticancer effect by inducing mitochondrial dysfunction, oxidative stress, cytoskeletal disorganization, cell cycle arrest, and mitochondria-mediated apoptosis, and it could be a potent therapeutic option for HCC., (© 2023 John Wiley & Sons Ltd.)- Published
- 2024
- Full Text
- View/download PDF
16. Lagerstroemia speciosa Pers. (Lythraceae) Ethanolic Extract Attenuates Isoniazid-Induced Oxidative Stress and Hepatic Inflammation in Rats.
- Author
-
Rohit Singh T, Ezhilarasan D, Karthick M, and Shree Harini K
- Abstract
Background Drug-induced liver injury is a common cause of acute liver failure. Isoniazid (INH) is used as a first-line treatment for tuberculosis. Clinical and experimental studies have reported abnormal liver function after INH therapy. Lagerstroemia speciosa Pers., commonly known as banaba, has been traditionally used to treat various ailments including diabetes and obesity due to its antioxidant and anti-inflammatory properties. Aim To investigate the hepatoprotective effect of ethanolic banaba leaf extract (EBLE) against INH-induced hepatotoxicity in rats. Materials and methods A total of 30 male Wistar albino rats (150 - 200 g) were divided into five groups (n = 6). Group I rats were served as a control and were administered dimethyl sulfoxide for the first 30 days and water for the next 30 consecutive days. Group II rats were administered INH (50 mg/kg, p.o.) once in the first 30 consecutive days and sacrificed at Day 30. Group III rats were administered INH for 30 consecutive days and left without treatment for the next 30 days. In Groups IV and V, rats were post-treated orally with EBLE 250 and 500 mg/kg, p.o. (0.3 ml/rat) for 30 days after INH administration. At the end of Day 60, the remaining group of animals were sacrificed. The blood and liver tissues were collected. The marker enzymes of hepatotoxicity, oxidative stress markers, inflammatory markers, and histopathology were analyzed. Results INH administration induced significant elevation of marker enzymes (aspartate transaminase, alanine transaminase, alkaline phosphatase, lactate dehydrogenase, bilirubin, gamma-glutamyl transpeptidase) of hepatotoxicity in the serum. This treatment also increased lipid peroxidation and proinflammatory marker expression (tumor necrosis factor-alpha, transforming growth factor-beta, and nuclear factor kappa B (NF-κB) except inhibitor of NF-κB) and decreased antioxidants such superoxide dismutase, catalase, and glutathione in the liver tissue. All these abnormalities were significantly mitigated after treatment with EBLE. Conclusion The results of this study suggest that EBLE can be used for INH-induced hepatotoxicity., Competing Interests: The authors have declared that no competing interests exist., (Copyright © 2024, Rohit Singh et al.)
- Published
- 2024
- Full Text
- View/download PDF
17. Ethyl gallate concurrent administration protects against acetaminophen-induced acute liver injury in mice: An in vivo and in silico approach.
- Author
-
Ezhilarasan D, Shree Harini K, Karthick M, and Selvaraj C
- Subjects
- Mice, Animals, Acetaminophen toxicity, Liver, Gallic Acid metabolism, Gallic Acid pharmacology, Anti-Inflammatory Agents pharmacology, Oxidative Stress, Antioxidants pharmacology, Antioxidants therapeutic use, Antioxidants metabolism, Chemical and Drug Induced Liver Injury drug therapy, Chemical and Drug Induced Liver Injury pathology
- Abstract
Acetaminophen (APAP) in high doses causes acute liver injury and acute liver failure. Ethyl gallate (EG) is a natural polyphenol, possessing antioxidant, anti-inflammatory, and anti-microbial properties. Therefore, in this study, we evaluated the protective role of EG against APAP-induced acute liver injury in mice. Acute liver injury was induced by a single dose of APAP (400 mg/kg., i.p.). In separate groups, EG (10 mg/kg), EG (20 mg/kg), and N-acetylcysteine (NAC; 1200 mg/kg., i.p.) were administered concurrently with APAP. The mice were sacrificed after 24 h of treatment. Liver marker enzymes of hepatotoxicity, antioxidant markers, inflammatory markers, and histopathological studies were done. APAP administration caused a significant elevation of marker enzymes of hepatotoxicity and lipid peroxidation. APAP administration also decreased enzymic and nonenzymic antioxidants. Acute APAP intoxication induced nuclear factor κ B, tumor necrosis factor-α, interleukin-1, p65, and p52 and downregulated IκB gene expressions. Our histopathological studies have confirmed the presence of centrilobular necrosis, 24 h after APAP intoxication. All the above abnormalities were significantly inhibited in groups of mice that were concurrently administered with APAP + EG and APAP + NAC. Our in silico analysis further confirms that hydroxyl groups of EG interact with the above inflammatory proteins at the 3,4,5-trihydroxybenzoic acid region. These effects of EG against APAP-induced acute liver injury could be attributed to its antioxidative, free radical scavenging, and anti-inflammatory potentials. Therefore, this study suggests that EG can be an efficient therapeutic approach to protect the liver from APAP intoxication., (© 2023 John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
18. Molecular insights on intracellular Wnt/β-catenin signaling in alcoholic liver disease.
- Author
-
Shree Harini K, Ezhilarasan D, and Mani U
- Subjects
- Humans, beta Catenin, Ethanol, Liver Cirrhosis, Carcinoma, Hepatocellular, Liver Neoplasms, Liver Diseases, Alcoholic, Fatty Liver
- Abstract
Alcoholic liver disease (ALD) is one of the most common health problems worldwide, especially in developing countries caused by chronic consumption of alcohol on a daily basis. The ALD spectrum is initiated with the early stages of alcoholic fatty liver (steatosis), progressing to alcoholic steatohepatitis, followed by the later stages of fibrosis and in some cases, cirrhosis and hepatocellular carcinoma (HCC). The Wnt/β-catenin signaling required for healthy liver development, function, and regeneration is found to be aberrated in ALD, attributed to its progression. This review is to elucidate the association of Wnt/β-catenin signaling with various stages of ALD progression. Alcohol causes downregulation of Wnt/β-catenin signaling components and thereby suppressing the pathway. Reports have been published that aberrated Wnt/β-catenin signaling, especially the absence of β-catenin, results in decreased alcohol metabolism, causing steatosis followed by steatohepatitis via lipid accumulation, lipid peroxidation, liver injury, increased oxidative stress and apoptosis of hepatocytes, contributing to the advancement of ALD. Contrastingly, the progression of later stages of ALD like fibrosis and HCC depends on the increased activation of Wnt/β-catenin signaling and its components. Existing studies reveal the varied expression of Wnt/β-catenin signaling in ALD. However, the dual role of the Wnt/β-catenin pathway in earlier and later stages of ALD is not clear. Therefore, studies on the Wnt/β-catenin pathway and its components in various manifestations of ALD might provide insight in targeting the Wnt/β-catenin pathway in ALD treatment., (© 2024 John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
19. Anticancer Potential of Farnesol Against Human Osteosarcoma Saos-2 Cells and Human Colorectal Carcinoma HCT-116 Cells.
- Author
-
Fathima Hinaz ZH, Pragya S, Ezhilarasan D, and Shree Harini K
- Abstract
Introduction: Increased colorectal carcinoma (CRC) and osteosarcoma prevalence, low survival rate, poor prognosis, and the limitations of existing anticancer therapies like side effects of drugs, non-specificity, short half-life, etc., pose a need for novel anticancer drugs. Farnesol, an organic sesquiterpene compound, found in the essential oils of various plants has been shown to possess antioxidant, anti-inflammatory, and anticancer properties. However, the anticancer effect of farnesol against CRC and osteosarcoma has not yet been adequately elucidated., Aim: The aim of the study was to analyze the anticancer effects of farnesol against human osteosarcoma and CRC cell lines., Materials and Methods: Human osteosarcoma (Saos-2) and colorectal carcinoma (HCT-116) cell lines were procured and cultured at 37
o C and 5% CO2 . The cells were treated with 10, 20, 40, 60, 80, and 100 µM/ml and 20, 40, 60, 80, 100, and 120 µM/ml of farnesol for 24 hours, respectively. 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide assay was performed to assess the cytotoxicity of farnesol on Saos-2 and HCT-116 cells. Acridine orange/ethidium bromide staining was carried out to analyze apoptosis. 4',6-diamidino-2-phenylindole staining was done to observe the nuclear changes. Dichloro-dihydro-fluorescein diacetate staining was performed to assess the farnesol-induced reactive oxygen species (ROS)-mediated cell death., Results: Farnesol reduced the viability and proliferation of Saos-2 and HCT-116 cells in a dose-dependent manner. Farnesol was able to alter the cellular and nuclear morphology of Saos-2 and HCT-116 cells, promoting cell death. Farnesol-induced apoptosis in human osteosarcoma and colorectal carcinoma cell lines. Early apoptosis was observed in farnesol-treated HCT-116 cells. Additionally, ROS-mediated apoptotic cell death was reported in Saos-2 cells., Conclusion: Farnesol has the potential to induce cytotoxicity against human osteosarcoma and CRC cell lines., Competing Interests: The authors have declared that no competing interests exist., (Copyright © 2023, Fathima Hinaz et al.)- Published
- 2023
- Full Text
- View/download PDF
20. Boldine Treatment Induces Cytotoxicity in Human Colorectal Carcinoma and Osteosarcoma Cells.
- Author
-
Chandan P, Dev A, Ezhilarasan D, and Shree Harini K
- Abstract
Introduction Cancer continues to be a significant health issue worldwide, with colorectal cancer (CRC) standing out as one of the most prevalent forms of cancer on a global scale. The lifetime risk of developing CRC is about one in 23 (4.3%) for men and one in 25 (4.0%) for women. Moreover, children and adolescents are frequently reported with osteosarcoma with a low five-year survival rate (69% and 67%, respectively). Aim The aim of the study was to analyze the cytotoxic effects of boldine against human CRC (HCT-116) and osteosarcoma cell lines (Saos-2). Materials and methods HCT-116 and Saos-2 cell lines were subjected to different concentrations of boldine treatment (5, 10, 20, 30, 40, and 50 μg/mL) and (10, 20, 40, 60, and 80 µg/mL), respectively, for 24 hours. The cytotoxicity was analyzed by MTT assay, AO/EB staining, DCFH-DA assay, and scratch assay. Results The MTT assay, microscopic analysis, and staining showed that boldine had dose-dependent cytotoxic effects against HCT-116 and Saos-2 cell lines by inhibiting their proliferation, viability, and migration, and inducing ROS-mediated apoptosis. Conclusion The study concluded that boldine had a concentration-dependent cytotoxic effect on human CRC and osteosarcoma cell lines., Competing Interests: The authors have declared that no competing interests exist., (Copyright © 2023, Chandan et al.)
- Published
- 2023
- Full Text
- View/download PDF
21. Concurrent administration of farnesol protects acetaminophen-induced acute hepatic necrosis in mice.
- Author
-
Gayatri Devi R and Ezhilarasan D
- Subjects
- Mice, Animals, Antioxidants metabolism, Farnesol pharmacology, Farnesol metabolism, NF-kappa B metabolism, Acetylcysteine pharmacology, Liver metabolism, Glutathione metabolism, Necrosis, Transaminases metabolism, Transaminases pharmacology, Alanine Transaminase, Acetaminophen toxicity, Chemical and Drug Induced Liver Injury drug therapy, Chemical and Drug Induced Liver Injury prevention & control, Chemical and Drug Induced Liver Injury metabolism
- Abstract
Acetaminophen (APAP) is known to cause acute liver injury and acute liver failure in Western countries. This study investigates the protective role of farnesol (FAR) (C
15 H26 O), a natural sesquiterpene alcohol in essential oils, against APAP-induced acute liver necrosis in mice. Mice were injected with a single dose of APAP (300 mg/kg) via an intraperitoneal route. Different groups of mice were concurrently treated with a single dose of FAR 25 mg/kg, FAR 50 mg/kg, and N-acetylcysteine. APAP administration caused a significant increase in transaminase activities and malondialdehyde (MDA) levels in the serum and liver tissue, respectively, with a concomitant decrease in intracellular antioxidants, including reduced glutathione (GSH) in the liver tissue. APAP intoxication upregulated proinflammatory cytokines such as tumor necrosis factor-α, interleukin-1β (IL-1β), IL-6, nuclear factor-κB (NF-κB), and IκB kinase β in the liver tissue. FAR and N-acetylcysteine (NAC) administrations concurrently with APAP prevented serum transaminase increase in serum and MDA levels in the liver tissue. A high dose of FAR and NAC treatments significantly inhibited GSH and other antioxidant depletion. FAR and NAC treatments also downregulated the expression of proinflammatory markers. FAR treatments protects against APAP-induced acute liver injury and offers antioxidant and anti-inflammatory effects by inhibiting the NF-κB pathway involved in the transcription of genes responsible for inflammatory cytokine synthesis., (© 2023 Wiley Periodicals LLC.)- Published
- 2023
- Full Text
- View/download PDF
22. Nelumbo nucifera Leaf Extract Induces Cytotoxicity in Osteosarcoma Saos-2 Cells.
- Author
-
Britina G, Ezhilarasan D, and Shree Harini K
- Abstract
Background Osteosarcoma is the eighth most common cancer and its prevalence in children makes it a global concern. Existing medications and treatments like high-dose methotrexate possess harmful side effects. Therefore, novel herbal drugs like Nelumbo nucifera are of utmost importance. Aim To analyze a novel anticancer herbal drug, Nelumbo nucifera leaf extract for its cytotoxic potential against osteosarcoma. Materials and method Nelumbo nucifera leaf extract was prepared. Saos-2 Cells (human osteosarcoma cell line) were treated with Nelumbo nucifera leaf extract (25, 50, 75, 100, 125, and 150 µg/ml) for 24 hours which were then subjected to MTT assay, morphological analysis and DAPI staining. Results The results suggested that Nelumbo nucifera leaf extract had a concentration-dependent cytotoxic effect on Saos-2 cell line. The extract significantly reduced the number of viable cells, inhibited proliferation and induced morphological changes in Saos-2 cells. Conclusion Nelumbo nucifera has the potential to induce cytotoxicity against osteosarcoma cell lines and hence, this study provides a novel therapeutic regimen for the treatment of osteosarcoma., Competing Interests: The authors have declared that no competing interests exist., (Copyright © 2023, Britina et al.)
- Published
- 2023
- Full Text
- View/download PDF
23. Coleus vettiveroides ethanolic root extract protects against thioacetamide-induced acute liver injury in rats.
- Author
-
Mohamed Azar KAH, Ezhilarasan D, Shree Harini K, Karthick M, and Uthirappan M
- Abstract
Acute liver injury is caused by various factors, including oxidative stress and inflammation. Coleus vettiveroides, an ayurvedic medicinal plant, is known to possess antioxidant, antibacterial, and antidiabetic properties. In this current study, we investigated the protective effect of C. vettiveroides ethanolic root extract (CVERE) against thioacetamide (TAA)-induced acute liver injury in rats. A single dose of TAA (300 mg/kg, b.w., i.p.) was administered to induce acute liver injury. The treatment groups of rats were concurrently treated with CVERE (125 and 250 mg/kg, b.w., p.o.) and silymarin (100 mg/kg, b.w., p.o.), respectively. After 24 h of the experimental period, TAA-induced liver injury was confirmed by increased activity of serum transaminases and malondialdehyde levels in liver tissue, decreased levels of antioxidants, upregulated expression of the inflammatory marker gene, and altered liver morphology. Whereas CVERE simultaneous treatment inhibited hepatic injury and prevented the elevation of serum aspartate and alanine transaminases, alkaline phosphatase, and lactate dehydrogenase activities. CVERE attenuated TAA-induced oxidative stress by suppressing lipid peroxidation and restoring antioxidants such as superoxide dismutase, catalase, and reduced glutathione. Further, CVERE treatment was found to inhibit nuclear factor κB-mediated inflammatory signaling, as indicated by downregulated pro-inflammatory cytokines including tumor necrosis factor-α and interleukin-1β. Our findings suggest that CVERE prevents TAA-induced acute liver injury by targeting oxidative stress and inflammation., (© 2023 John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
24. Wnt/beta-catenin signaling and its modulators in nonalcoholic fatty liver diseases.
- Author
-
Shree Harini K and Ezhilarasan D
- Subjects
- Humans, Wnt Signaling Pathway physiology, beta Catenin, Glycogen Synthase Kinase 3 beta metabolism, Disease Progression, Liver pathology, Liver Cirrhosis pathology, Fibrosis, Non-alcoholic Fatty Liver Disease pathology, Carcinoma, Hepatocellular pathology, Liver Neoplasms pathology
- Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global health concern associated with significant morbidity and mortality. NAFLD is a spectrum of diseases originating from simple steatosis, progressing through nonalcoholic steatohepatitis (NASH), fibrosis, and cirrhosis that may lead to hepatocellular carcinoma (HCC). The pathogenesis of NAFLD is mediated by the triglyceride accumulation followed by proinflammatory cytokines expression leading to inflammation, oxidative stress, and mitochondrial dysfunction denoted as "two-hit hypothesis", advancing with a "third hit" of insufficient hepatocyte proliferation, leading to the increase in hepatic progenitor cells contributing to fibrosis and HCC. Wnt/β-catenin signaling is responsible for normal liver development, regeneration, hepatic metabolic zonation, ammonia and drug detoxification, hepatobiliary development, etc., maintaining the overall liver homeostasis. The key regulators of canonical Wnt signaling such as LRP6, Wnt1, Wnt3a, β-catenin, GSK-3β, and APC are abnormally regulated in NAFLD. Many experimental studies have shown the aberrated Wnt/β-catenin signaling during the NAFLD progression and NASH to hepatic fibrosis and HCC. Therefore, in this review, we have emphasized the role of Wnt/β-catenin signaling and its modulators that can potentially aid in the inhibition of NAFLD., (Copyright © 2022. Published by Elsevier B.V.)
- Published
- 2023
- Full Text
- View/download PDF
25. Antitubercular drugs induced liver injury: an updated insight into molecular mechanisms.
- Author
-
Ezhilarasan D
- Subjects
- Humans, Antitubercular Agents adverse effects, Isoniazid, Pyrazinamide therapeutic use, Rifampin, Chemical and Drug Induced Liver Injury, Chronic drug therapy, Chemical and Drug Induced Liver Injury
- Abstract
Tuberculosis (TB) remains a major global health burden. Antitubercular drugs (ATDs) such as isoniazid (INH), rifampicin (RIF), pyrazinamide (PZA), and ethambutol are used as first-line therapy in TB patients. Drug-induced liver injury is one of the common side effects that leads to the discontinuation of ATDs in TB patients. Therefore, this review discusses the molecular pathogenesis of ATDs induced liver injury. The biotransformation of INH, RIF, and PZA in the liver liberates several reactive intermediates, leading to peroxidation of the hepatocellular membrane and oxidative stress. INH + RIF administration decreased the expression of bile acid transporters such as the bile salt export pump and multidrug resistance-associated protein 2 and induced liver injury by sirtuin 1 and farnesoid X receptor pathway. INH inhibits the nuclear translocation of Nrf2 by interfering with its nuclear importer, karyopherin β1, thereby inducing apoptosis. INF + RIF treatments alter Bcl-2 and Bax homeostasis, mitochondrial membrane potential, and cytochrome c release, thereby triggering apoptosis. RIF administration enhances the expression of genes involved in fatty acid synthesis and hepatocyte fatty acid uptake (CD36). RIF induces the expression of peroxisome proliferator-activated receptor -γ and its downstream proteins and perilipin-2 by activating the pregnane X receptor in the liver to increase fatty infiltration into the liver. ATDs administration induces oxidative stress, inflammation, apoptosis, cholestasis, and lipid accumulation in the liver. However, ATDs toxic potentials are not elaborately studied at the molecular level in clinical samples. Therefore, future studies are warranted to explore ATDs induced liver injuries at the molecular level in clinical samples whenever possible.
- Published
- 2023
- Full Text
- View/download PDF
26. Reply to "Wnt/beta-catenin signaling inhibitors and nonalcoholic fatty liver disease: Potential therapeutic implications".
- Author
-
Shree Harini K and Ezhilarasan D
- Subjects
- Humans, beta Catenin, Wnt Signaling Pathway, Non-alcoholic Fatty Liver Disease drug therapy
- Published
- 2023
- Full Text
- View/download PDF
27. Unraveling the pathophysiologic role of galectin-3 in chronically injured liver.
- Author
-
Ezhilarasan D
- Subjects
- Humans, Fibrosis, Hepatic Stellate Cells metabolism, Inflammation pathology, Liver Cirrhosis pathology, Galectin 3 genetics, Liver injuries, Liver pathology
- Abstract
Galectin-3 (Gal-3) previously referred to as S-type lectins, is a soluble protein that specifically binds to β-galactoside carbohydrates with high specificity. Gal-3 plays a pivotal role in a variety of pathophysiological processes such as cell proliferation, inflammation, differentiation, angiogenesis, transformation and apoptosis, pre-mRNA splicing, metabolic syndromes, fibrosis, and host defense. The role of Gal-3 has also been implicated in liver diseases. Gal-3 is activated upon a hepatotoxic insult to the liver and its level has been shown to be upregulated in fatty liver diseases, inflammation, nonalcoholic steatohepatitis, fibrosis, cholangitis, cirrhosis, and hepatocellular carcinoma (HCC). Gal-3 directly interacts with the NOD-like receptor family, pyrin domain containing 3, and activates the inflammasome in macrophages of the liver. In the chronically injured liver, Gal-3 secreted by injured hepatocytes and immune cells, activates hepatic stellate cells (HSCs) in a paracrine fashion to acquire a myofibroblast like collagen-producing phenotype. Activated HSCs in the fibrotic liver secrete Gal-3 which acts via autocrine signaling to exacerbate extracellular matrix synthesis and fibrogenesis. In the stromal microenvironment, Gal-3 activates cancer cell proliferation, migration, invasiveness, and metastasis. Clinically, increased serum levels and Gal-3 expression were observed in the liver tissue of nonalcoholic steatohepatitis, fibrotic/cirrhotic, and HCC patients. The pathological role of Gal-3 has been experimentally and clinically reported in the progression of chronic liver disease. Therefore, this review discusses the pathological role of Gal-3 in the progression of chronic liver diseases., (© 2023 Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
28. Molecular mechanisms in thioacetamide-induced acute and chronic liver injury models.
- Author
-
Ezhilarasan D
- Subjects
- Animals, Liver Cirrhosis chemically induced, Liver Cirrhosis metabolism, Hepatocytes metabolism, Hepatic Stellate Cells metabolism, Hepatic Stellate Cells pathology, Transforming Growth Factor beta, Thioacetamide toxicity, Thioacetamide metabolism, Liver metabolism
- Abstract
Thioacetamide (TAA) undergoes bioactivation in the liver by the CYP450 2E1 enzyme, resulting in the formation of TAA-S-oxide and TAA-S-dioxide. TAA-S-dioxide induces oxidative stress via lipid peroxidation of the hepatocellular membrane. A single TAA dose (50-300 mg/kg) administration initiates hepatocellular necrosis around the pericentral region after its covalent binding to macromolecules in the liver. Intermittent TAA administration (150-300 mg/kg, weekly thrice, for 11-16 weeks) activates transforming growth factor (TGF)-β/smad3 downstream signaling in injured hepatocytes, causing hepatic stellate cells (HSCs) to acquire myofibroblast like phenotype. The activated HSCs synthesize a variety of extracellular matrix, leading to liver fibrosis, cirrhosis, and portal hypertension. The TAA induced liver injury varies depending on the animal model, dosage, frequency, and routes of administration. However, TAA induces hepatotoxicity in a reproducible manner, and it is an ideal model to evaluate the antioxidant, cytoprotective, and antifibrotic compounds in experimental animals., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
29. Personalized mRNA cancer vaccines with immune checkpoint inhibitors: A promising therapeutic approach in oral cancer patients.
- Author
-
Gheena S and Ezhilarasan D
- Subjects
- Humans, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, RNA, Messenger, Immunotherapy, Cancer Vaccines therapeutic use, Neoplasms drug therapy, Mouth Neoplasms drug therapy, Mouth Neoplasms genetics
- Abstract
Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
- Published
- 2023
- Full Text
- View/download PDF
30. Deciphering the possible reciprocal loop between hepatic stellate cells and cancer cells in the tumor microenvironment of the liver.
- Author
-
Ezhilarasan D and Najimi M
- Subjects
- Humans, Hepatic Stellate Cells metabolism, Hepatic Stellate Cells pathology, Tumor Microenvironment, Endothelial Cells, Liver Neoplasms pathology, Carcinoma, Hepatocellular pathology
- Abstract
Activated hepatic stellate cells (HSCs)/myofibroblasts are the important sources of cancer-associated fibroblasts in the liver tumor microenvironment (TME). The crosstalk between activated HSCs and tumor cells mediates HCC progression, metastasis, tumor cell survival, angiogenesis and chemoresistance. In TME, HCC cells secrete various soluble factors responsible for the phenotypic activation of quiescent HSCs. Tumor cells use activated HSC-derived extracellular matrix (ECM) for migration and invasion. Further, in liver TME, activated HSCs and sinusoidal endothelial cells engage in a crosstalk that causes the secretion of angiogenesis and metastasis-related growth factors and cytokines. Activated HSCs and immune cells crosstalk to decrease immune surveillance in the liver TME by increasing the population of T regulatory cells and M2 macrophages or myeloid-derived suppressor cells. Thus, HSCs play a vital role in liver TME cell interactions. Therefore, a deep understanding of HSCs activation and their crosstalk with cancer and immune cells in TME may lead to the development of novel therapeutic strategies to target HCC., Competing Interests: Declaration of Competing Interest The authors report no declarations of interest., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
31. Intercellular communication among liver cells in the perisinusoidal space of the injured liver: Pathophysiology and therapeutic directions.
- Author
-
Ezhilarasan D and Najimi M
- Subjects
- Humans, Carcinoma, Hepatocellular pathology, Endothelial Cells metabolism, Hepatocytes metabolism, Liver Cirrhosis pathology, Liver Neoplasms pathology, Transforming Growth Factor beta1 metabolism, Tumor Microenvironment, Cell Communication, Hepatic Stellate Cells metabolism, Liver metabolism, Liver physiopathology
- Abstract
Hepatic stellate cells (HSCs) in the perisinusoidal space are surrounded by hepatocytes, liver sinusoidal endothelial cells, Kupffer cells, and other resident immune cells. In the normal liver, HSCs communicate with these cells to maintain normal liver functions. However, after chronic liver injury, injured hepatocytes release several proinflammatory mediators, reactive oxygen species, and damage-associated molecular patterns into the perisinusoidal space. Consequently, such alteration activates quiescent HSCs to acquire a myofibroblast-like phenotype and express high amounts of transforming growth factor-β1, angiopoietins, vascular endothelial growth factors, interleukins 6 and 8, fibril forming collagens, laminin, and E-cadherin. These phenotypic and functional transdifferentiation lead to hepatic fibrosis with a typical abnormal extracellular matrix synthesis and disorganization of the perisinusoidal space of the injured liver. Those changes provide a favorable environment that regulates tumor cell proliferation, migration, adhesion, and survival in the perisinusoidal space. Such tumor cells by releasing transforming growth factor-β1 and other cytokines, will, in turn, activate and deeply interact with HSCs via a bidirectional loop. Furthermore, hepatocellular carcinoma-derived mediators convert HSCs and macrophages into protumorigenic cell populations. Thus, the perisinusoidal space serves as a critical hub for activating HSCs and their interactions with other cell types, which cause a variety of liver diseases such as hepatic inflammation, fibrosis, cirrhosis, and their complications, such as portal hypertension and hepatocellular carcinoma. Therefore, targeting the crosstalk between activated HSCs and tumor cells/immune cells in the tumor microenvironment may also support a promising therapeutic strategy., (© 2022 Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
32. Letter to Editor on: "Ginger ( Zingiber officinale roscoe) extract could upregulate the renal expression of NRF2 and TNFα and prevents ethanol-induced toxicity in rat kidney" by Akbari et al.
- Author
-
Elumalai P, Ezhilarasan D, and Raghunandhakumar S
- Abstract
Competing Interests: The author declares that he has no conflicts of interest to disclose.
- Published
- 2023
- Full Text
- View/download PDF
33. Restoring the anti-tumor property of PTEN: A promising oral cancer treatment.
- Author
-
Shree Harini K, Ezhilarasan D, and Elumalai P
- Subjects
- Humans, PTEN Phosphohydrolase genetics, Mouth Neoplasms drug therapy
- Abstract
Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
- Published
- 2022
- Full Text
- View/download PDF
34. Emerging applications of CRISPR/Cas9 gene editing technology in reversing drug resistance in oral squamous cell carcinoma.
- Author
-
Elumalai P and Ezhilarasan D
- Subjects
- CRISPR-Cas Systems genetics, Drug Resistance, Gene Editing, Humans, Squamous Cell Carcinoma of Head and Neck genetics, Technology, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell genetics, Head and Neck Neoplasms genetics, Mouth Neoplasms drug therapy, Mouth Neoplasms genetics
- Abstract
Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
- Published
- 2022
- Full Text
- View/download PDF
35. Nano-based targeted drug delivery for lung cancer: therapeutic avenues and challenges.
- Author
-
Ezhilarasan D, Lakshmi T, and Mallineni SK
- Subjects
- Humans, Drug Delivery Systems, Epidermal Growth Factor therapeutic use, Paclitaxel therapeutic use, Pharmaceutical Preparations, Drug Carriers therapeutic use, Cell Line, Tumor, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Antineoplastic Agents metabolism, Lung Neoplasms drug therapy, Lung Neoplasms metabolism, Nanoparticles
- Abstract
Most anticancer drugs often fail in clinical trials due to poor solubility, poor bioavailability, lack of targeted delivery and several off-target effects. Polymeric nanoparticles such as poly(lactide), poly(lactic- co -glycolic acid), ALB-loading paclitaxel (Abraxane
® ABI-007), lomustine-loaded chitosan, gelatin (decorated with EGF receptor-targeted biotinylated EGF) and so on offer controlled and sustained drug-release properties, biocompatibility and promising anticancer effects. EGF, folic acid, transferrin, sigma and urokinase plasminogen activator receptors-targeting nano preparations improve bioavailability and accumulate drugs on the lung tumor cell surface. However, route of administration, size, pharmacokinetic properties, immune clearance and so on hamper nanomedicines' clinical uses. This review focuses on the benefits, avenues and challenges of nanoparticle-based drug-delivery systems for lung cancer treatment.- Published
- 2022
- Full Text
- View/download PDF
36. Valproic acid induced liver injury: An insight into molecular toxicological mechanism.
- Author
-
Ezhilarasan D and Mani U
- Subjects
- Adenosine Triphosphate metabolism, Ammonia, Anticonvulsants toxicity, Carnitine O-Palmitoyltransferase metabolism, Coenzyme A metabolism, DNA, Mitochondrial metabolism, Glutathione metabolism, Humans, Liver metabolism, Peroxisome Proliferator-Activated Receptors metabolism, Protons, Transaminases metabolism, Triglycerides metabolism, Valproic Acid toxicity, Chemical and Drug Induced Liver Injury metabolism, Chemical and Drug Induced Liver Injury, Chronic drug therapy, Chemical and Drug Induced Liver Injury, Chronic metabolism, Chemical and Drug Induced Liver Injury, Chronic pathology
- Abstract
Valproic acid (VPA) is an anti-seizure drug that causes idiosyncratic liver injury. 2-propyl-4-pentenoic acid (Δ
4 VPA), a metabolite of VPA, has been implicated in VPA-induced hepatotoxicity. This review summarizes the pathogenesis involved in VPA-induced liver injury. The VPA induce liver injury mainly by i) liberation of Δ4 VPA metabolites; ii) decrease in glutathione stores and antioxidants, resulting in oxidative stress; iii) inhibition of fatty acid β-oxidation, inducing mitochondrial DNA depletion and hypermethylation; a decrease in proton leak; oxidative phosphorylation impairment and ATP synthesis decrease; iv) induction of fatty liver via inhibition of carnitine palmitoyltransferase I, enhancing nuclear receptor peroxisome proliferator-activated receptor-gamma and acyl-CoA thioesterase 1, and inducing long-chain fatty acid uptake and triglyceride synthesis. VPA administration aggravates liver injury in individuals with metabolic syndromes. Therapeutic drug monitoring, routine serum levels of transaminases, ammonia, and lipid parameters during VPA therapy may thus be beneficial in improving the safety profile or preventing the progression of DILI., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 Elsevier B.V. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
37. Novel mTOR inhibitors: Promising therapeutic interventions in oral cancer therapy.
- Author
-
Ezhilarasan D and Shree Harini K
- Subjects
- Humans, Phosphoinositide-3 Kinase Inhibitors, Protein Kinase Inhibitors therapeutic use, TOR Serine-Threonine Kinases, MTOR Inhibitors, Mouth Neoplasms drug therapy
- Published
- 2022
- Full Text
- View/download PDF
38. Porphyromonas gingivalis and dental stem cells crosstalk amplify inflammation and bone loss in the periodontitis niche.
- Author
-
Ezhilarasan D and Varghese SS
- Subjects
- Alkaline Phosphatase metabolism, Collagen Type I, Core Binding Factor Alpha 1 Subunit, Cytokines metabolism, Humans, Inflammation, Osteocalcin, Osteogenesis, Osteopontin, Porphyromonas gingivalis metabolism, Stem Cells metabolism, Dental Caries, Periodontal Diseases, Periodontitis
- Abstract
Periodontitis is the sixth most prevalent disease, and almost 3.5 billion people are affected globally by dental caries and periodontal diseases. The microbial shift from a symbiotic microbiota to a dysbiotic microbiota in the oral cavity generally initiates periodontal disease. Pathogens in the periodontal microenvironment interact with stem cells to modulate their regenerative potential. Therefore, this review focuses on the interaction between microbes and stem cells in periodontitis conditions. Microbes direct dental stem cells to secrete a variety of pro-inflammatory cytokines and chemokines, which increase the inflammatory burden in the damaged periodontal tissue, which further aggravates periodontitis. Microbial interaction also decreases the osteogenic differentiation potential of dental stem cells by downregulating alkaline phosphatase, runt-related transcription factor 2, type 1 collagen, osteocalcin, osteopontin, and so on. Microbe and stem cell interaction amplifies pro-inflammatory cytokine signaling in the periodontitis niche, decreasing the osteogenic commitment of dental stem cells. A clear understanding of microbial stem cell interactions is crucial in designing regenerative therapies using stem cells in the management of periodontitis., (© 2022 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
39. Lagerstroemia speciosa (L.) Pers., ethanolic leaves extract attenuates dapsone-induced liver inflammation in rats.
- Author
-
Rohit Singh T and Ezhilarasan D
- Subjects
- Animals, Antioxidants pharmacology, Dapsone toxicity, Ethanol toxicity, Inflammation chemically induced, Inflammation drug therapy, Lipid Peroxidation, Liver, Oxidative Stress, Plant Extracts pharmacology, Rats, Chemical and Drug Induced Liver Injury etiology, Chemical and Drug Induced Liver Injury prevention & control, Lagerstroemia, Silymarin pharmacology
- Abstract
Drug-induced liver injury is a common cause of acute liver failure. Dapsone is increasingly used in combination with rifampicin for the treatment of leprosy and also for several dermatological disorders. Clinically, abnormal liver function and focal bile duct destruction were reported after dapsone therapy. Lagerstroemia speciosa Pers., commonly known as Banaba has been traditionally used to treat various ailments including diabetes and obesity due to its antioxidant and anti-inflammatory efficacies. This study investigated the hepatoprotective effect of ethanolic banaba leaves extract (EBLE) against dapsone-induced hepatotoxicity in rats. Dapsone (30 mg/kg, i.p.) was administered twice daily for 30 days. In separate groups, rats were post-treated orally with EBLE (250 and 500 mg/kg) and silymarin (100 mg/kg) once daily for 30 days after dapsone administration. The marker enzymes of hepatotoxicity, oxidative stress markers, inflammatory markers and histopathology of liver were done. HPTLC analysis confirmed the presence of 12.87 µg of corosolic acid per mg of EBLE. Dapsone administration-induced significant ( p < 0.001) elevation of marker enzymes of hepatotoxicity in serum. This treatment also increased lipid peroxidation ( p < 0.001) and pro-inflammatory markers (tumor necrosis factor-alpha, transforming growth factor-beta, and nuclear factor kappa-B) expressions ( p < 0.001) and decreased antioxidants ( p < 0.001) such superoxide dismutase, catalase and glutathione in the liver tissue. All these abnormalities were significantly ( p < 0.001) mitigated after EBLE (500 mg/kg) and silymarin post-treatments. The results of this study suggest that silymarin and EBLE can be used for dapsone-induced hepatotoxicity.
- Published
- 2022
- Full Text
- View/download PDF
40. Syringic acid and silymarin concurrent administration inhibits sodium valproate-induced liver injury in rats.
- Author
-
Gheena S, Ezhilarasan D, Shree Harini K, and Rajeshkumar S
- Subjects
- Animals, Anti-Inflammatory Agents pharmacology, Antioxidants metabolism, Antioxidants pharmacology, Gallic Acid analogs & derivatives, Humans, Liver, Oxidative Stress, Rats, Rats, Wistar, Valproic Acid metabolism, Chemical and Drug Induced Liver Injury etiology, Chemical and Drug Induced Liver Injury metabolism, Chemical and Drug Induced Liver Injury prevention & control, Chemical and Drug Induced Liver Injury, Chronic metabolism, Silymarin pharmacology
- Abstract
Sodium valproate (SV) is a well-known anti-epileptic drug, also used to control convulsions, bipolar disorders and migraines. SV has been shown to induce liver toxicity in clinical subjects. Syringic acid (SA), a natural polyphenolic compound has potential antioxidant, anti-inflammatory and several beneficial effects. Therefore, in this study, we evaluated hepatoprotective effect of SA against SV-induced liver injury in rats. Wistar rats were treated with SV orally at a dose of 500 mg/kg, once daily, for 14 days. Another three groups of rats were administered with SV and concurrently treated with SA (40 and 80 mg/kg) and silymarin (SIL) (100 mg/kg) for 14 days. SV administration for 14 days caused significant (p < .001) elevation of liver transaminases and ALP in serum. Liver MDA level was significantly (p < .001) increased with a concomitant decrease (p < .001) in enzymic antioxidants activities in SV administered rats. SV administration also caused the upregulation of proinflammatory markers such as tumor necrosis factor α, c-Jun N-terminal kinase, nuclear factor kappa B, cyclooxygenase-2 and Interleukin 6 expressions in liver tissue. Histopathological studies also revealed the presence of inflammatory cell infiltration and hepatocellular necrosis upon SV administration. At both doses, concurrent administration of SA and SIL significantly (p < .001) inhibited the liver transaminase activities in serum, oxidative stress, and proinflammatory markers expression in liver tissue. Our current results suggest that SA can be a promising herbal drug that can inhibit SV-induced hepatotoxicity when administered together due its potential anti-inflammatory effects., (© 2022 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
41. Osteopontin: A reliable diagnostic marker and an attractive therapeutic target in oral cancers.
- Author
-
Ezhilarasan D, Shree Harini K, and Lakshmi T
- Subjects
- Biomarkers, Tumor, Humans, Mouth Neoplasms diagnosis, Osteopontin
- Published
- 2022
- Full Text
- View/download PDF
42. Novel fibroblast growth factor receptor inhibitors: Potential therapeutic approach in oral cancer treatment.
- Author
-
Shree Harini K, Ezhilarasan D, and Lakshmi T
- Subjects
- Humans, Protein Kinase Inhibitors, Receptor, Fibroblast Growth Factor, Type 1 metabolism, Receptors, Fibroblast Growth Factor metabolism, Mouth Neoplasms drug therapy
- Published
- 2022
- Full Text
- View/download PDF
43. Promising autophagy inhibitors: Therapeutic implications in oral cancer.
- Author
-
Shree Harini K and Ezhilarasan D
- Subjects
- Apoptosis, Cell Line, Tumor, Humans, Autophagy, Mouth Neoplasms drug therapy
- Published
- 2022
- Full Text
- View/download PDF
44. Patient derived tumour organoids: An emerging strategy in oral cancer research and therapeutics.
- Author
-
Shree Harini K, Ezhilarasan D, and Lakshmi T
- Subjects
- Humans, Mouth Neoplasms pathology, Mouth Neoplasms therapy, Organoids pathology
- Published
- 2022
- Full Text
- View/download PDF
45. A Molecular Insight into the Role of Antioxidants in Nonalcoholic Fatty Liver Diseases.
- Author
-
Ezhilarasan D and Lakshmi T
- Subjects
- Antioxidants metabolism, Antioxidants pharmacology, Antioxidants therapeutic use, Humans, Liver metabolism, NF-kappa B metabolism, Resveratrol pharmacology, Resveratrol therapeutic use, Silybin therapeutic use, Liver Neoplasms pathology, Non-alcoholic Fatty Liver Disease pathology, Pentoxifylline therapeutic use, Silymarin
- Abstract
Nonalcoholic fatty liver disease (NAFLD) defines fat accumulation in the liver, and it is commonly associated with metabolic syndromes like diabetes and obesity. Progressive NAFLD leads to nonalcoholic steatohepatitis (NASH) and ultimately causes cirrhosis and hepatocellular carcinoma, and NASH is currently a frequent cause of liver transplantation. Oxidative stress is often contributed to the progression of NAFLD, and hence, antioxidants such as silymarin, silybin, or silibinin, pentoxifylline, resveratrol, and vitamins A, C, and E are used in clinical trials against NAFLD. Silymarin induces the peroxisome proliferator-activated receptor α (PPAR α ), a fatty acid sensor, which promotes the transcription of genes that are required for the enzymes involved in lipid oxidation in hepatocytes. Silybin inhibits sterol regulatory element-binding protein 1 and carbohydrate response element-binding protein to downregulate the expression of genes responsible for de novo lipogenesis by activating AMP-activated protein kinase phosphorylation. Pentoxifylline inhibits TNF- α expression and endoplasmic reticulum stress-mediated inflammatory nuclear factor kappa B (NF- κ B) activation. Thus, it prevents NAFLD to NASH progression. Resveratrol inhibits methylation at Nrf-2 promoters and NF- κ B activity via SIRT1 activation in NAFLD conditions. However, clinically, resveratrol has not shown promising beneficial effects. Vitamin C is beneficial in NAFLD patients. Vitamin E is not effectively regressing hepatic fibrosis. Hence, its combination with antifibrotic agents is used as an adjuvant to produce a synergistic antifibrotic effect. However, to date, none of these antioxidants have been used as a definite therapeutic agent in NAFLD patients. Further, these antioxidants should be studied in NAFLD patients with larger populations and multiple endpoints in the future., Competing Interests: The authors declare that there is no conflict of interest regarding the publication of this article., (Copyright © 2022 Devaraj Ezhilarasan and Thangavelu Lakshmi.)
- Published
- 2022
- Full Text
- View/download PDF
46. Telescreening as an alternate modality for early detection of oral cancer.
- Author
-
DagliNamrata, Lakshmi T, Rushabh D, Dineshkumar T, and Ezhilarasan D
- Subjects
- Early Detection of Cancer, Humans, Mass Screening, Mouth Neoplasms diagnosis, Telemedicine
- Published
- 2022
- Full Text
- View/download PDF
47. The ambiguous role of sirtuins in head and neck squamous cell carcinoma.
- Author
-
Ezhilarasan D, Lakshmi T, Subha M, Deepak Nallasamy V, and Raghunandhakumar S
- Subjects
- Cell Proliferation, Humans, Squamous Cell Carcinoma of Head and Neck, Head and Neck Neoplasms, Mouth Neoplasms, Sirtuins metabolism
- Abstract
Oral cancer is one of the most leading cancer responsible for significant morbidity and mortality. The sirtuins (SIRTs) are a family of class III histone deacetylases and are known to regulate a variety of molecular signaling associated with different cancer types including oral malignancies. SIRT1 acts as bifunctional in a variety of cancer. In oral cancer, SIRT1 seems to work as a tumor suppressor. The carcinogenic potential of SIRT1 is also reported in oral cancer, and hence, its role is still ambiguous. SIRT2 is also said to play a dual-faced role in different types of cancers. However, in oral cancer, SIRT2 is not studied and its role remains obscure. SIRT3 expression was positively correlated with oral malignancies. However, studies also showed the anti-cancer role of SIRT3 in oral cancer. SIRT7 loss was observed in oral cancer cells, while its overexpression caused the suppression of oral cancer cells proliferation, migration, and invasiveness. The role of other SIRTs in oral cancer was studied meagerly or reports not available. To date, only the roles of SIRT1, SIRT3, and SIRT7 have been reported in oral malignancies. Therefore, understanding the regulatory mechanisms employed by sirtuins to modulate oral cancer is important for developing potential anti-cancer therapeutic strategies., (© 2021 Wiley Periodicals LLC.)
- Published
- 2022
- Full Text
- View/download PDF
48. Targeting head and neck cancer epigenetics with CRISPR-dCas9: An emerging therapeutic approach.
- Author
-
Elumalai P, Ezhilarasan D, and Lakshmi T
- Subjects
- Epigenesis, Genetic, Gene Editing, Humans, Clustered Regularly Interspaced Short Palindromic Repeats, Head and Neck Neoplasms genetics, Head and Neck Neoplasms therapy
- Published
- 2022
- Full Text
- View/download PDF
49. Role of stress management in oral malignant and premalignant conditions.
- Author
-
Namrata D, Lakshmi T, Rushabh D, and Ezhilarasan D
- Subjects
- Humans, Mouth Neoplasms pathology, Mouth Neoplasms therapy, Precancerous Conditions pathology, Precancerous Conditions therapy
- Published
- 2022
- Full Text
- View/download PDF
50. Molecular docking analysis of syringic acid with proteins in inflammatory cascade.
- Author
-
Sukumaran G, Ezhilarasan D, Ramani P, and Merlin RJ
- Abstract
Syringic Acid (SA) is a dimethoxybenzene derived from plants. Dietary SA possesses anti-obesity, anti-inflammatory and anti-steatotic effects and is of interest as a potential therapeutic medication in the treatment of obesity, diabetes, diabetic cataracts and asthma. It has anti-tumorigenic effect against hepatocellular carcinoma, lung carcinoma and oral mucosal carcinoma. It is also believed to have a protective effect on Acetaminophen induced damage in Wistar rats. Therefore, it is of interest to document the molecular docking analysis of syringic acid with proteins in inflammatory cascade such as TNF α, NFκB, P50, P65 and IKB for further consideration in drug discovery., Competing Interests: None to declare, (© 2022 Biomedical Informatics.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.