- Home
- A-Z Publications
- Current Molecular Pharmacology
- Issue Home
Current Molecular Pharmacology - Current Issue
Volume 17, Issue 1, 2024
-
-
RBM3 Accelerates Wound Healing of Skin in Diabetes through ERK1/2 Signaling
Authors: Jianguo Feng, Menghong Long, Xin Zhao, Pijun Yan, Yunxiao Lin, Maohua Wang and Wenhua HuangBackgroundWith the increasing risk of infections and other serious complications, the underlying molecular mechanism of wound healing impairment in diabetes deserves attention. Cold shock proteins (CSPs), including CIRP and RBM3 are highly expressed in the skin; however, it is unknown whether CSPs are involved in the wound-healing impairment of diabetic skin.
ObjectivesThe objective of this study is to investigate the effects of RBM3 on skin wound healing in diabetes.
MethodsIn vitro experiments, western blot assay was used to test the levels of proteins in HaCaT cells treated with different concentrations of glucose. RBM3 was over-expressed in HaCaT cells using lentivirus particles. Cell viability was analyzed by Cell-Counting Kit-8 assay and colony formation assay. The migration of HaCaT cells at different concentrations of glucose was evaluated by wound healing assay. In vivo experiments, the mouse model of diabetes was established by intraperitoneal injection of streptozotocin. Four weeks later, the mice were anesthetized by intraperitoneal injection of pentobarbital sodium for skin tissue collection or wound healing experiments. RBM3 knockout mice were established by removing exons 2–6 using the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 technique and then used in skin wound healing experiments with or without diabetic stress.
ResultsIn this study, the expression of RBM3, rather than CIRP, was altered in the skin of diabetic specimens, and the RBM3’s overexpression accelerated the cell viability and proliferation of HaCaT cells under high glucose conditions. RBM3 deficiency caused delayed wound healing in RBM3 knockout in diabetic conditions. Moreover. RBM3 enhanced the ERK1/2 signaling pathway, and its inhibitor FR180204 blocked the beneficial effect of RBM3 overexpression on skin wound healing in diabetes.
ConclusionRBM3 activated the ERK1/2 signal to facilitate skin wound healing in diabetes, offering a novel therapeutic target for its treatment.
-
- Pharmacology
-
-
-
Targeting Mutant-p53 for Cancer Treatment: Are We There Yet?
BackgroundMutations in the TP53 gene are the most common among genetic alterations in human cancers, resulting in the formation of mutant p53 protein (mutp53). Mutp53 promotes proliferation, migration, invasion, and metastasis in cancer cells. Not only does the initiation of oncogenesis ensue due to mutp53, but resistance towards chemotherapy and radiotherapy in cancer cells also occurs. This review aims to summarise and discuss the oncogenesis of mutant p53 in cancer cells and introduce the various mutant p53 inhibitors currently being evaluated at the pre-clinical and clinical stages. Compounds that induce the wild-type conformation on the targeted p53 missense mutation, restore or enhance the DNA binding of mutant p53, and inhibit cancer cells' growth are highlighted. In addition, the progression and development of the mutant p53 inhibitors in clinical trials are updated.
ConclusionThe progress of developing a cancer treatment that may successfully and efficiently target mutant p53 is on the verge of development. Mutant p53 proteins not only initiate oncogenesis but also cause resistance in cancer cells to certain chemo or radiotherapies, further endorse cancer cell survival and promote migration as well as metastasis of cancerous cells. With this regard, many mutant p53 inhibitors have been developed, some of which are currently being evaluated at the pre-clinical level and have been identified and discussed. To date, APR-246 is the most prominent one that has progressed to the Phase III clinical trial.
-
-
-
-
Maprotiline Prompts an Antitumour Effect by Inhibiting PD-L1 Expression in Mice with Melanoma
Authors: Lirui Liang, Yang Li, Yang Jiao, Chunjing Zhang, Mingguang Shao, Hanyu Jiang, Zunge Wu, Haoqi Chen, Jiaming Guo, Huijie Jia and Tiesuo ZhaoBackgroundResearch has revealed that the expression of PD-L1 is significantly upregulated in tumour cells and that the binding of programmed cell death protein 1 (PD-1) to programmed cell death 1 ligand 1 (PD-L1) inhibits the response of T cells, thereby suppressing tumour immunity. Therefore, blocking PD-L1/PD-1 signalling has become an important target in clinical immunotherapy. Some old drugs, namely, non-anticancer drugs, have also been found to have antitumour effects, and maprotiline is one of them. Maprotiline is a tetracyclic antidepressant that has been widely used to treat depression. However, it has not yet been reported whether maprotiline can exert an antitumour effect on melanoma.
ObjectiveThis study aimed to investigate the antitumour efficacy of maprotiline in mice with melanoma.
MethodsIn this study, female C57BL/6 mice were used to establish a tumour-bearing animal model. After treatment with maprotiline, the survival rate of mice was recorded daily. The expression of relevant proteins was detected by Western blotting, the proportion of immune cells was detected by flow cytometry, and the infiltration of immune cells in tumour tissue was detected by immunofluorescence staining.
ResultsMaprotiline was found to inhibit the proliferation and migration of B16 cells while increasing cell apoptosis. Importantly, treatment with maprotiline decreased the expression of PD-L1 and increased the proportion of CD4+ T cells, CD8+ T cells, and NK cells in the spleen. It also increased the infiltration of CD4+ and CD8+ T cells in tumour tissue.
ConclusionOur research findings suggest that maprotiline enhances the antitumour immune response in mouse melanoma by inhibiting PD-L1 expression. This study may discover a new PD-L1 inhibitor, providing a novel therapeutic option for the clinical treatment of tumours.
-
-
-
Targeting FGFR3 is a Useful Therapeutic Strategy for Rheumatoid Arthritis Treatment
Authors: Shan-Fu Yu, Tien-Tsai Cheng, Gong-Kai Huang, Chung-Yuan Hsu, Ying-Hsien Kao and Yueh-Hua ChungBackgroundRheumatoid arthritis (RA) is a systemic inflammatory disease in which TNF-α plays an important role. Fibroblast growth factor receptor 3 (FGFR3) is reportedly involved in RA by regulating the expression of inflammatory cytokines.
ObjectiveThis study examined the expression profile of FGFR3 in human synovial biopsy tissues and evaluated its gene-silencing effects on behaviors of synovial cells.
MethodsImmunohistochemical staining was used to measure FGFR3 expression in human RA joint tissues. Cell proliferation, migration, and apoptosis assays were used to monitor behavioral changes in cultured synovial SW-982 cells with siRNA-mediated FGFR3 gene silencing. Immunofluorescent staining and western blotting were used to detect molecular changes in the FGFR3 gene-silenced cells.
ResultsFGFR3 up-regulation was noted in both cytoplasms and nuclei of synovial cells in human RA joints. FGFR3 siRNA delivery experiments corroborated that FGFR3 knockdown decreased proliferation and migration, and triggered apoptosis of synovial cells. The FGFR3 gene knockdown enhanced constitutive expression of epithelial marker E-cadherin and conversely suppressed expression of epithelial-mesenchymal transition (EMT) markers, including Snail, fibronectin, and vimentin. In addition, FGFR3 silencing significantly reduced the constitutive expressions of TNF-α, transcription factor NF-κΒ, and downstream COX-2 protein and collagenolytic enzyme MMP-9. MAPK inhibition markedly suppressed constitutive levels of NF-κΒ, COX-2, and MMP-9.
ConclusionGenetic interference of FGFR3 could modulate the expression of inflammatory mediators and EMT markers in the synovial cells. Targeting the FGFR3/MAPK signal axis may be considered a useful therapeutic strategy to ameliorate the development of RA.
-
-
-
Co-treatment of Astragaloside IV with Vitamin D in Diabetic Peripheral Neuropathic Rats: Protective Effects and Potential Mechanisms
Authors: Fengyan Tang, Bo Zhao, Li Zhang, Faisal Raza, Hajra Zafar, Shao Zhong, Lin Li, Wenhua Zhu, Lingna Fang, Bing Lu, Liwen Shen, Ping Guo, Nengxing Yu and Quanmin LiObjective:The potential mechanism underlying the protective effect of Astragaloside IV (AS-IV) co-treatment with 1, 25-dihydroxy-vitamin D (Vit-D) on neuropathy in diabetic high-fat rats was investigated.
Methods:The rat diabetic hyperlipidemia (DH) model was established via streptozotocin and a high-fat diet (HFD). After co-treatment (of AS-IV and Vit-D at respective doses of 50 mg/kg via oral gavage and 30000 IU/kg via intramuscular injection), blood glucose levels, markers of inflammation and oxidative stress, as well as apoptosis and histopathology were evaluated with appropriate techniques.
Results:Co-treatment could effectively reduce blood glucose levels substantially (p< 0.01), improve weight loss, and decrease oral glucose tolerance. Reduced respective sensory and motor nerve conduction velocities in rats were substantially improved (p<0.01) after co-treatment. Also, we observed obvious improvement in DH-induced injured nerve fiber myelin structure and other organ pathologies in co-treated rats. Besides, we observed up-regulated expressions of peroxisomal-proliferator activated receptor-alpha (PPAR-α) and Vit-D receptors (VDR) (p< 0.01) through the western blotting technique. Using the same technique, we also discovered reduced levels of interleukin (IL)1 beta, IL-6, and tumor necrosis factor-alpha, coupled with increased IL-10 and superoxide dismutase levels (p< 0.01). Importantly, co-treatment could effectively exert anti-oxidative and anti-inflammatory effects. Also, co-treatment resulted in the up-regulation of PPAR-α and VDR expressions, inhibition of the renin–angiotensin–aldosterone system, and promotion of β-cell sensitivity to insulin.
Conclusion:The combined application of AS-IV and Vit-D exhibited health effects such as anti-oxidation, regulation of inflammatory factors, and promotion of cell repair, which may be considered as the mechanisms underlying treatment of diabetic peripheral neuropathy and improvement in biochemical indicators.
-
- Pharmacology
-
-
-
A Promising Breakthrough: The Potential of VORASIDENIB in the Treatment of Low-grade Glioma
Authors: Alice Bombino, Marcello Magnani and Alfredo ContiBackground:This commentary explores the potential of Vorasidenib, also known as AG-881. This emerging small-molecule inhibitor has garnered substantial attention within the realm of oncology due to its unique mechanism of action and potential therapeutic applications.
Introduction:Gliomas are common malignant brain tumors characterized by diffuse brain infiltration. World Health Organization grade II and grade III diffuse gliomas are considered lower-grade gliomas (LGGs) and have isocitrate dehydrogenase (IDH) mutations. LGGs are challenging due to their infiltrative nature, making them capable of progressing into higher-grade malignancies. Vorasidenib is a novel therapeutic agent targeting mutant IDH1/2, sparking interest in the field.
Mechanism of Action:Vorasidenib inhibits mutant IDH1/2 through a unique mechanism, reducing the production of the oncometabolite 2-hydroxyglutarate (2-HG). This alteration affects key enzymes and DNA methylation, impacting tumor growth and invasion.
Preclinical Evidence:Preclinical studies show vorasidenib's efficacy in inhibiting mutant IDH1/2 and 2-HG production in glioma models. It suppresses tumor growth, making it a potential treatment option.
Clinical Evidence:Early clinical trials demonstrate vorasidenib's clinical activity in non-enhancing gliomas. It reduces 2-hydroxyglutarate levels and tumor cell proliferation, with an objective response rate and prolonged progression-free survival. The drug's safety profile is favorable.
Challenges and Future Directions:Challenges include identifying predictive biomarkers and optimizing sequencing or combinations with existing therapies. Further research is needed to establish long-term effectiveness, evaluate side effects, and explore combinations with immunotherapy.
Conclusion:Vorasidenib significantly advances LGG treatment, targeting a prevalent mutation and slowing tumor growth. Promising preclinical and clinical evidence and manageable side effects suggest its potential impact on LGG management. However, more research, including large trials, is needed to confirm its efficacy and role in treatment.
-
-
-
Grp94 Inhibitor HCP1 Suppressed the Replication of SVA in BHK-21 Cells and PK-15 Cells
Authors: Shuo Wang, XiaoLing Cui, Ren Hui, Wen Yao, BaoXiang Zhao, Jun Li and JunYing MiaoBackgroundGlucoregulatory protein 94 (Grp94) is necessary for the post-viral life cycle and plays a quality control role in viral proteins, but the role of Grp94 in regulating viral replication in host cells is not well known. Therefore, finding a compound that can regulate Grp94 will help us to study the mechanism of viral replication. Previously, we synthesized a coumarin pyrazoline derivative HCP1 that is an effective inhibitor of Grp94. We suppose that HCP1 may inhibit viral replication.
ObjectiveThis study aimed to investigate the effect of HCP1 on the replication ability of Senecavirus A (SVA), so as to provide a target and a leading compound for revealing the pathogenic mechanism of the virus and developing antiviral drugs.
MethodsRat cell lines BHK-21 and porcine cell lines PK-15 were infected with SVA, and the infected cells were treated with different concentrations of HCP1. The cell viability (CCK-8), virus titer (TCID50), autophagy level, and Grp94 expression were measured.
ResultsThe results showed that a low concentration of HCP1 decreased viral titer and viral load in BHK-21 and PK-15 cells, and 5μM HCP1 significantly decreased the expression of SVA VP2 protein. In addition, SVA infection can lead to an increased level of autophagy, and HCP1 can inhibit host cell autophagy caused by SVA infection, thereby inhibiting viral replication and infection.
ConclusionThese findings reveal that Grp94 is a key factor in controlling SVA replication, and its inhibitor HCP1 suppresses SVA replication by inhibiting the increase of Grp94 protein level and autophagy induced by SVA. This study will contribute to the development of a new class of small-molecule antiviral drugs.
-
-
-
Differential Kat3 Coactivator Usage Regulates Brain Metabolism and Neuronal Differentiation
IntroductionOur previous work has demonstrated significant effects on the oxidative stress response, mitochondrial function, and oxidative phosphorylation in the livers and intestines of p300 S89A knockin (S89AKI) mice. We now show that this mutation is also associated with brain metabolic defects and neuronal differentiation.
Methodsp300 S89A edited P19 cells, and S89AKI mice demonstrated metabolic and neuronal differentiation defects based on proteomic, cell biological and PET imaging studies.
ResultsThe metabolic and differentiation defects associated with the p300 S89A knockin mutation could be corrected both in vitro and in vivo utilizing the small molecule CBP/beta-catenin antagonist ICG-001.
ConclusionRebalancing the equilibrium between CBP/β-catenin versus p300/β-catenin associated transcription, utilizing the small molecule CBP/beta-catenin antagonist ICG-001, enhances mitochondrial oxidative phosphorylation, metabolic function, and neuronal differentiation and may be able to ameliorate the cognitive decline seen in neurodegenerative disorders, including Alzheimer’s Disease.
-
-
-
Antiarrhythmic Potential of Epicardial Botulinum Toxin Injection for Suppression of Postoperative Atrial Fibrillation
More LessFollowing heart surgery, postoperative atrial fibrillation (AF) is the most prevalent kind of secondary AF and the most frequent adverse event. Postoperative AF is related to a number of unfavorable cardiac outcomes, such as heart failure, stroke, and death. However, the pharmacological treatment for postoperative AF is only relatively efficient and is frequently linked to detrimental complications, including symptomatic bradycardia with atrioventricular block due to rate control drugs and elevated hemorrhage hazard attributable to the administration of anticoagulants. Ablation procedures also result in the irreversible damage of cardiac anatomic structures, which may have long-term negative implications on heart performance. As a result, there is an unmet demand for treatments that can minimize the incidence of postoperative AF in an effective and safe manner. Botulinum toxin is an established neurotoxin that has progressively gained use in every medical science domain. It hinders the propagation of impulses across nerve fibers without causing immediate damage to the cardiac tissue. The transient feature of botulinum toxin action and the eventual restoration of the autonomic nervous system transmission are undeniably advantageous and may render botulinum toxin a potential and feasible treatment approach for postoperative AF.
-
-
-
Physalin B Reduces Tau Phosphorylation and Cell Apoptosis in HEK293 Cells by Activating FoxO1
Authors: Wei Zhang, Yating Shi, Mingti Lv, Yimin Zhang, Wei Ren, Ruling Shi, Hecheng Wang and Linlin ShanBackgroundPhysalin B (PB) is one of the main active compounds of Solanaceae plants, with a wide range of biological activities. PB reportedly has the potential to treat Alzheimer’s disease (AD).
ObjectiveIn this study, we investigated the effect of PB on Tau phosphorylation and cell apoptosis using Tau-expressing HEK293 cells (HEK293/Tau) as a cellular model.
MethodsThe optimum concentration of PB to treat HEK293/Tau cells was determined using the CCK-8 assay. Additionally, the expression of FoxO1, Tau-5, p-Tau (T231, S262, and S404), ERK, p-ERK, GSK-3β, and p-GSK-3β was detected using western blotting to determine the effect of PB on Tau phosphorylation. The apoptosis rate was detected using flow cytometry, and the expression of Bax and Bcl-2 was detected using western blotting and verified using real-time quantitative polymerase chain reaction (RT-qPCR). Moreover, cells were transfected with FoxO1 siRNA to downregulate FoxO1 expression, and the expression of the above-mentioned proteins was detected to verify the effect of PB on Tau phosphorylation and cell apoptosis.
ResultsAfter 24 h of PB treatment, the phosphorylation levels of Tau at S404, S262, and T231 sites decreased significantly, and the activities of GSK-3β and ERK were inhibited. PB also reduced cell apoptosis by reducing the expression of Bax and increasing the expression of Bcl-2. In addition, PB decreased Tau phosphorylation and cell apoptosis by upregulating FoxO1.
ConclusionThe natural compound PB exhibited a protective effect in the AD cell model by increasing FoxO1 expression and reducing Tau phosphorylation and cell apoptosis.
-
-
-
Hepatic Ischemia-reperfusion Injury: Protective Approaches and Treatment
Authors: Kuldeep Singh, Jeetendra Kumar Gupta, Shivendra Kuma, Anurag, Soumyadip Mukherjee and Aman PatelIschemia and reperfusion damage to the liver is one of the major causes of hepatic dysfunction and liver failure after a liver transplant. The start of hepatic ischemia-reperfusion damage is linked to metabolic acidosis, Kupffer cells, neutrophils, excessive calcium, and changes in the permeability of the mitochondrial membrane. Hypoxia activates Kupffer cells, resulting in the production of reactive oxygen species (ROS). These ROS when accumulated, causes apoptosis and necrosis, as well as activate immune and inflammatory responses that involve many cells and signalling molecules. Numerous antioxidant compounds have been researched to lessen oxidative stress and thus serve as potential compounds to deal the ischemia-reperfusion damage. This article confers a deep understanding of the protective effects of some effective therapies, including hepatoprotective agents, attenuation of an increase in xanthine oxidase activity, and administration of antioxidants like N-acetylcysteine, superoxide dismutase (SOD), and ornithine.
-
-
-
Pathophysiology, Current Therapeutic Options, Vaccine Candidates, and Drug Targets for Human Brucellosis
Authors: Manisha Pritam and Rajnish KumarBrucellosis is an infectious disease caused by different species of Brucella bacteria. It is also known as Malta fever, one of the neglected diseases that can cause infection in both animals and humans. Although human-to-human infection is rare, it can spread through the inhalation of airborne agents, and if left untreated, it can lead to serious health complications. In this review, we aim to highlight the pathophysiology, prevention, epidemiology, mitigation, cure, targets for drug development, and vaccine development against human brucellosis. Human brucellosis is mainly caused by consuming unpasteurized milk or dairy products, uncooked meat, and contact with infected animals. Human brucellosis outbreaks are mainly associated with developing and low- to middle-income countries. Brucella is present all over the world, and only some of the regions are at high risk, including Asia, Africa, Eastern Europe, Mexico, South and Central America, the Caribbean, the Mediterranean Basin, and the Middle East. Because of intracellular survival, inhibition of apoptosis, and immune evasion, Brucella can survive and multiply inside the host cell, which can cause chronic disease. By using proteomics approaches, several new drug targets were reported for human brucellosis that can be used for the development of novel drugs. We can also develop an efficient vaccine against human brucellosis by exploring previously reported vaccine candidates against animal brucellosis. The information provided through this review will facilitate research to control and cure human brucellosis and its complicated symptoms.
-
-
-
CD73 Blockade Alleviated Hepatic Fibrosis via Inhibiting Hepatic Stellate Cells Proliferation and Activation
Authors: Lan Yang, Zhao-Wei Gao, Xia-nan Wu, Chong Liu, Juan Zhang, Hui-Zhong Zhang and Ke DongBackgroundLiver fibrosis is associated with the activation of hepatic stellate cells (HSCs). Inhibition of HSCs activation is a strategy for alleviating hepatic fibrogenesis. CD73 is involved in liver disease development, while the mechanism remains unclear.
ObjectiveThis study aimed to investigate the effect of CD73 targeting inhibition on liver fibrosis.
MethodsIntraperitoneal injection of CCl4 was used to induce liver fibrosis in mice models. Adenosine 5′-(α, β-methylene) diphosphate sodium salt (APCP) was used for CD73 blockade. The siRNA was used to induce CD73 knockdown in HSCs. LX2 and HSC-T6 were used to investigate the role of CD73 in HSCs activation in vitro.
ResultsThe results showed that APCP treatment could alleviate hepatic fibrosis. In fibrotic liver tissues, CD73 exhibited a positive correlation with markers of HSCs activation. Furthermore, APCP treatment and CD73 knockdown could inhibit HSCs (LX2 and HSC-T6) activation and proliferation. By using RNA sequencing of liver tissues from control, CCl4-mice, and APCP-treated mice, 851 genes that were significantly changed in CCl4 mice (vs. control) were reversed by APCP treatment. These genes were mainly enriched in cell division-associated biological processes. Moreover, we found that CD73 might be associated with autophagy in HSCs. In fibrotic liver tissues and HSCs, ATG5 and Beclin1 expression could be downregulated by CD73 knockdown and APCP treatment.
ConclusionThis study demonstrated the effects and mechanism of CD73 in HSCs activation and proliferation, which presents the therapeutical potential of CD73 blockage for liver fibrosis.
-
-
-
The Targeted Therapies for Osteosarcoma via Six Major Pathways
Authors: Shuxing Wang, Quanlei Ren, Guoqing Li, Xiaoxuan Zhao, Xing Zhao and Zhen ZhangOsteosarcoma is the most common primary bone malignancy and has a high tendency of local invasion. Although a lot of studies have focused on chemotherapy and combination chemotherapy regimens in recent years, still, there is no particularly perfect regimen for the treatment of relapsed or metastatic OS, and the prognosis is still relatively poor. As a new therapeutic method, targeted therapy provides a new scheme for patients with osteosarcoma and has a wide application prospect. This article reviews the latest progress of targeted therapy for osteosarcoma, and summarizes the research on the corresponding targets of osteosarcoma through six major pathways. These studies can pave the way for new treatments for osteosarcoma patients who need them.
-
-
-
SGLT2 Inhibitors and Diabetic Kidney Disease: Targeting Multiple and Interrelated Signaling Pathways for Renal Protection
More LessAlmost 20-40% of all patients suffering from diabetes mellitus experience chronic kidney disease, which is related to higher mortality (cardiovascular and all-cause). The implication of several pathophysiological mechanisms (hemodynamic, tubular, metabolic and inflammatory) in the pathogenesis of diabetic kidney disease generates an urgent need to develop multitarget therapeutic strategies to face its development and progression. SGLT2 inhibitors are undoubtedly a practice-changing drug class for individuals who experience type 2 diabetes and diabetic kidney disease. In vitro studies, exploratory research, sub-analyses of large randomized controlled trials, and investigation of several biomarkers have demonstrated that SGLT2 inhibitors achieved multiple beneficial activities, targeting several renal cellular and molecular pathways independent of their antihyperglycemic activity. These mainly include the reduction in intraglomerular pressure through the restoration of TGF, impacts on the renin-angiotensin-aldosterone system, improvement of renal hypoxia, adaptive metabolic alterations in substrate use/energy expenditure, improvement of mitochondrial dysfunction, and reduction of inflammation, oxidative stress and fibrosis. This manuscript thoroughly investigates the possible mechanisms that underlie their salutary renal effects in patients with diabetes, focusing on several pathways involved and the interplay between them. It also explores their upcoming role in ameliorating the evolution of chronic kidney disease in patients with diabetes.
-
-
-
Regulating miRNAs Expression by Resveratrol: Novel Insights based on Molecular Mechanism and Strategies for Cancer Therapy
Resveratrol, a polyphenolic phytoalexin found in a wide range of plants, including grapes, berries, and peanuts, is an extensively researched phytochemical with unique pharmacological capabilities and amazing potential to affect many targets in various cancers. Resveratrol's anti-cancer activities are due to its targeting of a variety of cellular and molecular mechanisms and crucial processes involved in cancer pathogenesis, such as the promotion of growth arrest, stimulation of apoptosis, suppression of cell proliferation, induction of autophagy, regulating oxidative stress and inflammation, and improving the influence of some of the other chemotherapeutic agents. MicroRNAs (miRNAs) are non-coding RNAs that modulate gene expression by degrading mRNA or inhibiting translation. MiRNAs serve critical roles in a wide range of biological activities, and disruption of miRNA expression is strongly linked to cancer progression. Recent research has shown that resveratrol has anti-proliferative and/or pro-apoptotic properties via modulating the miRNA network, which leads to the inhibition of tumor cell proliferation, the activation of apoptosis, or the increase of traditional cancer therapy effectiveness. As a result, employing resveratrol to target miRNAs will be a unique and potential anticancer approach. Here, we discuss the main advances in the modulation of miRNA expression by resveratrol, as well as the several miRNAs that may be influenced by resveratrol in different types of cancer and the significance of this natural drug as a promising strategy in cancer treatment.
-
-
-
Peptides for Dual Targeting of ErbB1 and ErbB2: Blocking EGFR Cell Signaling Transduction Pathways for Cancer Chemotherapy
Cancer is one of the most deadly diseases involving dysregulated cell proliferation. Chemotherapeutic drugs have serious drawbacks of nonspecific toxicity and drug resistance. Tyrosine kinases are a significant class of enzymes of protein kinases. The four members of the trans-membrane family of tyrosine kinase receptors known as the human epidermal growth factor receptors (EGFR), ErbB1/HER1, ErbB2/HER2/neu, ErbB3/HER3, and ErbB4/HER4, are overexpressed in many forms of cancer. These receptors are crucial for cell division, invasion, metastasis, angiogenesis, and uncontrolled activation of cancer cells. In this context, an attractive combination of anticancer drug targets is ErbB1 and ErbB2. Numerous cancer types exhibit overexpression of ErbB1 and ErbB2, which is linked to poor prognosis and causes resistance to ErbB1-targeted therapy. Further, it has been reported in recent years that the use of peptides as anticancer agents have the potential to circumvent the drawbacks of the currently used chemotherapeutic drugs. Among them, short peptides have several advantages when compared to small molecules. The present report reviews the importance of tyrosine kinases as targets for cancer, the role of peptides as therapeutic agents, and the investigations that have been carried out by earlier workers for targeting both ErbB1 and ErbB2 using therapeutic peptides.
-
-
-
E2F1 Reduces Sorafenib’s Sensitivity of Esophageal Carcinoma Cells via Modulating the miR-29c-3p/COL11A1 Signaling Axis
Authors: Zhifeng Ma, Ting Zhu, Haiyong Wang, Bin Wang, Linhai Fu and Guangmao YuObjectiveEsophageal carcinoma (ESCA) is a common malignancy characterized by high morbidity and mortality. Our work managed to dissect the modulatory mechanism of E2F1/miR-29c-3p/COL11A1 in the malignant progression and sensitivity of ESCA cells to sorafenib.
MethodsVia bioinformatics approaches, we identified the target miRNA. Subsequently, CCK-8, cell cycle analysis, and flow cytometry were used to check the biological influences of miR-29c-3p on ESCA cells. TransmiR, mirDIP, miRPathDB, and miRDB databases were used as tools for the prediction of upstream transcription factors and downstream genes of miR-29c-3p. The targeting relationship of genes was detected via RNA immunoprecipitation and chromatin immunoprecipitation, which was further validated by dual-luciferase assay. Finally, in vitro experiments revealed the way E2F1/miR-29c-3p/COL11A1 affected sorafenib’s sensitivity, and in vivo experiments were used to verify the way E2F1 and sorafenib impacted ESCA tumor growth.
ResultsmiR-29c-3p, downregulated in ESCA, could suppress ESCA cell viability, arrest the cell cycle in the G0/G1 phase, and impel apoptosis. E2F1 was found to be upregulated in ESCA and it could abate the transcriptional activity of miR-29c-3p. COL11A1 was found to be a downstream target of miR-29c-3p to enhance cell viability, induce cell cycle arrest in S phase, and constrain apoptosis. Cellular and animal experiments together demonstrated that E2F1 abated the sorafenib’s sensitivity of ESCA cells via miR-29c-3p/COL11A1.
ConclusionE2F1 affected the viability, cell cycle, and apoptosis of ESCA cells by modulating miR-29c-3p/COL11A1, and it attenuated the sensitivity of ESCA cells to sorafenib, shedding new light on the treatment of ESCA.
-