Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
J Adv Res ; 55: 131-144, 2024 Jan.
Article En | MEDLINE | ID: mdl-36801384

INTRODUCTION: Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease all over the world, and no drug is approved for the treatment of NAFLD. Bavachinin (BVC) is proven to possess liver-protecting effect against NAFLD, but its mechanism is still blurry. OBJECTIVES: With the use of Click Chemistry-Activity-Based Protein Profiling (CC-ABPP) technology, this study aims to identify the target of BVC, and investigate the mechanism by which BVC exerts its liver-protecting effect. METHODS: The high fat diet induced hamster NAFLD model is introduced to investigate BVC's lipid-lowering and liver-protecting effects. Then, a small molecular probe ofBVC is designed and synthesized based on theCC-ABPP technology, and BVC's target is fished out. A series of experiments are performed to identify the target, including competitive inhibition assay, surface-plasmon resonance (SPR), cellular thermal shift assay (CETSA), drug affinity responsive target stability (DARTS) assay, and co-immunoprecipitation (Co-IP). Afterward, the pro-regeneration effects of BVC are validated in vitro and in vivo through flow cytometry, immunofluorescence, and the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL). RESULT: In the hamster NAFLD model, BVC shows lipid-lowing effect and improvement on the histology. PCNA is identified as the target of BVC with the method mentioned above, and BVC facilitates the interaction between PCNA and DNA polymerase delta. BVC promotes HepG2 cells proliferation which is inhibited by T2AA, an inhibitor suppresses the interaction between PCNA and DNA polymerase delta. In NAFLD hamsters, BVC enhances PCNA expression and liver regeneration, reduces hepatocyte apoptosis. CONCLUSION: This study suggests that, besides the anti-lipemic effect, BVC binds to the pocket of PCNA facilitating its interaction with DNA polymerase delta and pro-regeneration effect, thereby exerts the protective effect against HFD induced liver injury.


Non-alcoholic Fatty Liver Disease , Humans , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/therapeutic use , DNA Polymerase III/metabolism , Lipid Metabolism/genetics , Lipids/therapeutic use
2.
J Am Soc Nephrol ; 35(1): 22-40, 2024 01 01.
Article En | MEDLINE | ID: mdl-37962623

SIGNIFICANCE STATEMENT: To combat both untoward effects of nephrotoxicity and ototoxicity in cisplatin-treated patients, two potential therapeutic oral anticancer drugs AZD5438 and dabrafenib, a phase-2 clinical trial protein kinase CDK2 inhibitor and an US Food and Drug Administration-approved drug BRAF inhibitor, respectively, were tested in an established mouse AKI model. Both drugs have previously been shown to protect significantly against cisplatin-induced hearing loss in mice. Each drug ameliorated cisplatin-induced increases in the serum biomarkers BUN, creatinine, and neutrophil gelatinase-associated lipocalin. Drugs also improved renal histopathology and inflammation, mitigated cell death by pyroptosis and necroptosis, and significantly enhanced overall survival of cisplatin-treated mice. BACKGROUND: Cisplatin is an effective chemotherapy agent for a wide variety of solid tumors, but its use is dose-limited by serious side effects, including AKI and hearing loss. There are no US Food and Drug Administration-approved drugs to treat both side effects. Recently, two anticancer oral drugs, AZD5438 and dabrafenib, were identified as protective against cisplatin-induced hearing loss in mice. We hypothesize that similar cell stress and death pathways are activated in kidney and inner ear cells when exposed to cisplatin and tested whether these drugs alleviate cisplatin-induced AKI. METHODS: The HK-2 cell line and adult FVB mice were used to measure the protection from cisplatin-induced cell death and AKI by these drugs. Serum markers of kidney injury, BUN, creatinine, and neutrophil gelatinase-associated lipocalin as well as histology of kidneys were analyzed. The levels of markers of kidney cell death, including necroptosis and pyroptosis, pERK, and proliferating cell nuclear antigen, were also examined by Western blotting and immunofluorescence. In addition, CDK2 knockout (KO) mice were used to confirm AZD5438 protective effect is through CDK2 inhibition. RESULTS: The drugs reduced cisplatin-induced cell death in the HK-2 cell line and attenuated cisplatin-induced AKI in mice. The drugs reduced serum kidney injury markers, inhibited cell death, and reduced the levels of pERK and proliferating cell nuclear antigen, all of which correlated with prolonged animal survival. CDK2 KO mice were resistant to cisplatin-induced AKI, and AZD5438 conferred no additional protection in the KO mice. CONCLUSIONS: Cisplatin-induced damage to the inner ear and kidneys shares similar cellular beneficial responses to AZD5438 and dabrafenib, highlighting the potential therapeutic use of these agents to treat both cisplatin-mediated kidney damage and hearing loss.


Acute Kidney Injury , Antineoplastic Agents , Hearing Loss , Humans , Mice , Animals , Cisplatin/toxicity , Lipocalin-2 , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/pharmacology , Proliferating Cell Nuclear Antigen/therapeutic use , Creatinine , Drug Repositioning , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Antineoplastic Agents/toxicity , Hearing Loss/chemically induced , Hearing Loss/drug therapy , Mice, Inbred Strains , Mice, Knockout , Apoptosis
3.
Neurochem Int ; 171: 105641, 2023 Dec.
Article En | MEDLINE | ID: mdl-37952830

Among diseases of the central nervous system (CNS), spinal cord injury (SCI) has a high fatality rate. It has been proven that P2Y G protein-coupled purinergic receptors have a neuroprotective role in apoptosis and regeneration inside the damaged spinal cord. The P2Y12 receptor (P2Y12R) has recently been linked to peripheral neuropathy and stroke. However, the role of P2Y12R after SCI remains unclear. Our study randomly divided C57BL/6J female mice into 3 groups: Sham+DMSO, SCI+DMSO, and SCI+MRS2395. MRS2395 as a P2Y12R inhibitor was intraperitoneally injected at a dose of 1.5 mg/kg once daily for 7 days. We showed that the P2Y12R was markedly activated after injury, and it was double labeled with the microglial and neuron. Behavioral tests were employed to assess motor function recovery. By using immunofluorescence staining, the NeuN expression level was detected. The morphology of neurons was observed by hematoxylin-eosin and Nissl staining. P2Y12R, Bax, GFAP, PCNA and calbindin expression levels were detected using Western blot. Meanwhile, mitochondria and myelin sheath were observed by transmission electron microscopy (TEM). Our findings demonstrated that MRS2395 significantly enhanced motor function induced by SCI and that was used to alleviate apoptosis and astrocyte scarring. NeuN positive cells in the SCI group were lower than in the therapy group, although Bax, GFAP, PCNA and calbindin expression levels were considerably higher. Moreover, following MRS2395 therapy, the histological damage was reversed. A notable improvement in myelin sheath and mitochondrial morphology was seen in the therapy group. Together, our findings indicate that activation of P2Y12R in damaged spinal cord may be a critical event and suggest that inhibition of P2Y12R might be a feasible therapeutic strategy for treating SCI.


Demyelinating Diseases , Spinal Cord Injuries , Rats , Mice , Female , Animals , Rats, Sprague-Dawley , Purinergic P2Y Receptor Antagonists/therapeutic use , Recovery of Function , Dimethyl Sulfoxide/therapeutic use , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/therapeutic use , bcl-2-Associated X Protein/metabolism , Mice, Inbred C57BL , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Spinal Cord/metabolism , Apoptosis , Calbindins
4.
F S Sci ; 4(4): 341-349, 2023 11.
Article En | MEDLINE | ID: mdl-37739343

OBJECTIVE: To investigate the combined effects of Crila and green tea extract, epigallocatechin gallate (EGCG), compared with single treatments, on human uterine fibroid cells. DESIGN: Human uterine leiomyoma (HuLM) cells were treated with different concentrations of Crila, alone or in combination with EGCG, and several experiments were employed. SETTING: A laboratory study. PATIENTSS: N/A. INTERVENTIONS: Crila, EGCG. MAIN OUTCOME MEASURES: Cell proliferation assay, drug synergy using combination index, protein and gene expression analysis of proliferation marker proliferating cell nuclear antigen, and apoptosis marker BAX using western blotting and quantitative polymerase chain reaction, respectively. RESULTS: Results showed that tested Crila concentrations, when combined with 25 and 50 µM EGCG, exerted synergistic growth inhibitory effects on HuLM viability. This inhibitory effect on HuLM cell viability was because of decreased cell proliferation, as shown by a decrease in the proliferation marker proliferating cell nuclear antigen at messenger RNA and protein levels, rather than inducing apoptosis. CONCLUSION: Our study concludes that the utility of natural compounds may provide a safe and cost-effective alternative to currently used short-term hormonal therapies against uterine fibroids.


Leiomyoma , Uterine Neoplasms , Female , Humans , Proliferating Cell Nuclear Antigen/analysis , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/therapeutic use , Uterine Neoplasms/drug therapy , Uterine Neoplasms/metabolism , Cell Line, Tumor , Leiomyoma/drug therapy
5.
Clin Exp Hypertens ; 45(1): 2229538, 2023 Dec 31.
Article En | MEDLINE | ID: mdl-37395230

OBJECTIVE: Neointimal hyperplasia is the primary mechanism underlying atherosclerosis and restenosis after percutaneous coronary intervention. Ketogenic diet (KD) exerts beneficial effects in various diseases, but whether it could serve as non-drug therapy for neointimal hyperplasia remains unknown. This study aimed to investigate the effect of KD on neointimal hyperplasia and the potential mechanisms. METHODS AND RESULTS: Carotid artery balloon-injury model was employed in adult Sprague-Dawley rats to induce neointimal hyperplasia. Then, animals were subjected to either standard rodent chow or KD. For in-vitro experiment, impacts of ß-hydroxybutyrate (ß-HB), the main mediator of KD effects, on platelet-derived growth factor BB (PDGF-BB) induced vascular smooth muscle cell (VSMC) migration and proliferation were determined. Balloon injury induced event intimal hyperplasia and upregulation of protein expression of proliferating cell nuclear antigen (PCNA) and α-smooth muscle actin (α-SMA), and these changes were significantly ameliorated by KD. In addition, ß-HB could markedly inhibit PDGF-BB induced VMSC migration and proliferation, as well as inhibiting expressions of PCNA and α-SMC. Furthermore, KD inhibited balloon-injury induced oxidative stress in carotid artery, indicated by reduced ROS level, malondialdehyde (MDA) and myeloperoxidase (MPO) activities, and increased superoxide dismutase (SOD) activity. We also found balloon-injury induced inflammation in carotid artery was suppressed by KD, indicated by decreased expressions of proinflammatory cytokines IL-1ß and TNF-α, and increased expression of anti-inflammatory cytokine IL-10. CONCLUSION: KD attenuates neointimal hyperplasia through suppressing oxidative stress and inflammation to inhibit VSMC proliferation and migration. KD may represent a promising non-drug therapy for neointimal hyperplasia associated diseases.


Carotid Artery Injuries , Diet, Ketogenic , Rats , Animals , Hyperplasia/complications , Rats, Sprague-Dawley , Becaplermin/metabolism , Becaplermin/pharmacology , Becaplermin/therapeutic use , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/pharmacology , Proliferating Cell Nuclear Antigen/therapeutic use , Neointima/complications , Neointima/drug therapy , Neointima/metabolism , Carotid Artery Injuries/complications , Carotid Artery Injuries/drug therapy , Carotid Artery Injuries/metabolism , Oxidative Stress , Inflammation/complications , Cell Proliferation , Cell Movement , Cells, Cultured
6.
F S Sci ; 4(3): 239-256, 2023 08.
Article En | MEDLINE | ID: mdl-37182601

OBJECTIVE: To investigate the link between EZH2 and Wnt/ß-catenin signaling and its role in uterine fibroids (UFs) pathogenesis and explore the potential effect of natural compound methyl jasmonate (MJ) against UFs. DESIGN: EZH2 overexpression or inhibition was achieved in human uterine leiomyoma (HuLM) cells using EZH2-expressing adenovirus or chemical EZH2 inhibitor (DZNep), respectively. The HuLM and normal uterine smooth muscle cells were treated with 0.1-3 mM of MJ, and several experiments were employed. SETTING: Laboratory study. PATIENTS(S): None. INTERVENTION(S): Methyl jasmonate. MAIN OUTCOME MEASURE(S): Protein expression of EZH2, ß-catenin, and proliferating cell nuclear antigen (PCNA) was measured by Western blot as well as gene expression alterations of Wnt ligands (Wnt5A, Wnt5b, and Wnt9A), WISP1, CTNNB1, and its responsive gene PITX2 using quantitative real-time polymerase chain reaction. The protein and ribonucleic acid (RNA) levels of several markers were measured in MJ-treated or untreated HuLM cells, including EZH2 and ß-catenin, extracellular matrix markers collagen type 1 (COL1A1) and fibronectin (FN), proliferation markers cyclin D1 (CCND1) and PCNA, tumor suppressor marker p21, and apoptotic markers (BAX, cytochrome c, and cleaved caspase 3). RESULT(S): EZH2 overexpression significantly increased the gene expression of several Wnt ligands (PITX2, WISP1, WNT5A, WNT5B, and WNT9A), which increased nuclear translocation of ß-catenin and PCNA and eventually HuLM cell proliferation. EZH2 inhibition blocked Wnt/ß-catenin signaling activation where the aforementioned genes significantly decreased as well as PCNA, cyclin D1, and PITX2 protein expression compared with those in untreated HuLM. Methyl jasmonate showed a potent antiproliferative effect on HuLM cells in a dose- and time-dependent manner. Interestingly, the dose range (0.1-0.5 mM) showed a selective growth inhibitory effect on HuLM cells, not on normal uterine smooth muscle cells. Methyl jasmonate treatment at 0.5 mM for 24 hours significantly decreased both protein and RNA levels of EZH2, ß-catenin, COL1A1, FN, CCND1, PCNA, WISP1, and PITX2 but increased the protein levels of p21, BAX, cytochrome, c and cleaved caspase 3 compared with untreated HuLM. Methyl jasmonate-treated cells exhibited down-regulation in the RNA expression of 36 genes, including CTNNB1, CCND1, Wnt5A, Wnt5B, and Wnt9A, and up-regulation in the expression of 34 genes, including Wnt antagonist genes WIF1, PRICKlE1, and DKK1 compared with control, confirming the quantitative real-time polymerase chain reaction results. CONCLUSION(S): Our studies provide a novel link between EZH2 and the Wnt/ß-catenin signaling pathway in UFs. Targeting EZH2 with MJ interferes with the activation of wnt/ß-catenin signaling in our model. Methyl jasmonate may offer a promising therapeutic option as a nonhormonal and cost-effective treatment against UFs with favorable clinical utility, pending proven safe and efficient in human clinical trials.


Leiomyoma , Uterine Neoplasms , Female , Humans , Wnt Signaling Pathway/genetics , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/therapeutic use , Cyclin D1/metabolism , Cyclin D1/therapeutic use , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/metabolism , Caspase 3/metabolism , Caspase 3/therapeutic use , Ligands , bcl-2-Associated X Protein/therapeutic use , beta Catenin/genetics , beta Catenin/metabolism , beta Catenin/therapeutic use , Leiomyoma/drug therapy , Leiomyoma/genetics , RNA/therapeutic use , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism
7.
Exp Biol Med (Maywood) ; 248(17): 1500-1506, 2023 09.
Article En | MEDLINE | ID: mdl-36941805

Patients with temporomandibular dysfunction (TMD) usually suffer from pathology or malpositioning of the temporomandibular joint (TMJ) disk, leading to the degenerative lesion of condyles. Kartogenin can promote the repair of damaged cartilage. This study aimed to explore whether intra-articular injection of kartogenin could alleviate the TMJ injury induced by type II collagenase. We measured the head withdrawal threshold and found that kartogenin alleviated the pain around TMD induced by type II collagenase. We observed the morphology of the condylar surface and found that kartogenin protected the integration of the condylar surface. We analyzed the density of the subchondral bone and found that kartogenin minimized the damage of TMJ injury to the subchondral bone. We next explored the histological changes and found that kartogenin increased the thickness of the proliferative layer and more collagen formation in the superficial layer. Then, to further ensure whether kartogenin promotes cell proliferation in the condyle, we performed immunohistochemistry of proliferating cell nuclear antigen (PCNA). The ratio of PCNA-positive cells was significantly increased in the kartogenin group. Next, immunofluorescence of TGF-ß1 and SMAD3 was performed to reveal that kartogenin activated the TGF-ß/SMAD pathway in the proliferative layer. In conclusion, kartogenin may have a therapeutic effect on TMJ injury by promoting cell proliferation in cartilage and subchondral bone. Kartogenin may be promising as an intra-articular injection agent to treat TMD.


Cartilage, Articular , Osteoarthritis , Temporomandibular Joint Disorders , Humans , Rats , Animals , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/pharmacology , Proliferating Cell Nuclear Antigen/therapeutic use , Transforming Growth Factor beta/metabolism , Mandibular Condyle , Temporomandibular Joint Disorders/drug therapy , Temporomandibular Joint Disorders/metabolism , Temporomandibular Joint Disorders/pathology , Chondrocytes/metabolism , Cartilage, Articular/metabolism , Osteoarthritis/metabolism , Temporomandibular Joint/metabolism , Temporomandibular Joint/pathology , Collagenases/metabolism , Collagenases/pharmacology , Collagenases/therapeutic use
8.
J Integr Med ; 21(2): 184-193, 2023 03.
Article En | MEDLINE | ID: mdl-36781361

OBJECTIVE: Physical exercise, a common non-drug intervention, is an important strategy in cancer treatment, including hepatocellular carcinoma (HCC). However, the mechanism remains largely unknown. Due to the importance of hypoxia and cancer stemness in the development of HCC, the present study investigated whether the anti-HCC effect of physical exercise is related to its suppression on hypoxia and cancer stemness. METHODS: A physical exercise intervention of swimming (30 min/d, 5 d/week, for 4 weeks) was administered to BALB/c nude mice bearing subcutaneous human HCC tumor. The anti-HCC effect of swimming was assessed in vivo by tumor weight monitoring, hematoxylin and eosin (HE) staining, and immunohistochemistry (IHC) detection of proliferating cell nuclear antigen (PCNA) and Ki67. The expression of stemness transcription factors, including Nanog homeobox (NANOG), octamer-binding transcription factor 4 (OCT-4), v-Myc avian myelocytomatosis viral oncogene homolog (C-MYC) and hypoxia-inducible factor-1α (HIF-1α), was detected using real-time reverse transcription polymerase chain reaction. A hypoxia probe was used to explore the intratumoral hypoxia status. Western blot was used to detect the expression of HIF-1α and proteins related to protein kinase B (Akt)/glycogen synthase kinase-3ß (GSK-3ß)/ß-catenin signaling pathway. The IHC analysis of platelet endothelial cell adhesion molecule-1 (CD31), and the immunofluorescence co-location of CD31 and desmin were used to analyze tumor blood perfusion. SMMC-7721 cells were treated with nude mice serum. The inhibition effect on cancer stemness in vitro was detected using suspension sphere experiments and the expression of stemness transcription factors. The hypoxia status was inferred by measuring the protein and mRNA levels of HIF-1α. Further, the expression of proteins related to Akt/GSK-3ß/ß-catenin signaling pathway was detected. RESULTS: Swimming significantly reduced the body weight and tumor weight in nude mice bearing HCC tumor. HE staining and IHC results showed a lower necrotic area ratio as well as fewer PCNA or Ki67 positive cells in mice receiving the swimming intervention. Swimming potently alleviated the intratumoral hypoxia, attenuated the cancer stemness, and inhibited the Akt/GSK-3ß/ß-catenin signaling pathway. Additionally, the desmin+/CD31+ ratio, rather than the number of CD31+ vessels, was significantly increased in swimming-treated mice. In vitro experiments showed that treating cells with the serum from the swimming intervention mice significantly reduced the formation of SMMC-7721 cell suspension sphere, as well as the mRNA expression level of stemness transcription factors. Consistent with the in vivo results, HIF-1α and Akt/GSK-3ß/ß-catenin signaling pathway were also inhibited in cells treated with serum from swimming group. CONCLUSION: Swimming alleviated hypoxia and attenuated cancer stemness in HCC, through suppression of the Akt/GSK-3ß/ß-catenin signaling pathway. The alleviation of intratumoral hypoxia was related to the increase in blood perfusion in the tumor. Please cite this article as: Xiao CL, Zhong ZP, Lü C, Guo BJ, Chen JJ, Zhao T, Yin ZF, Li B. Physical exercise suppresses hepatocellular carcinoma progression by alleviating hypoxia and attenuating cancer stemness through the Akt/GSK-3ß/ß-catenin pathway. J Integr Med. 2023; 21(2): 184-193.


Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Animals , Mice , Carcinoma, Hepatocellular/drug therapy , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proliferating Cell Nuclear Antigen/therapeutic use , Mice, Nude , Glycogen Synthase Kinase 3 beta/genetics , beta Catenin/genetics , beta Catenin/metabolism , beta Catenin/therapeutic use , Liver Neoplasms/drug therapy , Desmin/therapeutic use , Ki-67 Antigen , Cell Line, Tumor , Hypoxia , RNA, Messenger/therapeutic use , Cell Proliferation
9.
Folia Morphol (Warsz) ; 82(3): 641-655, 2023.
Article En | MEDLINE | ID: mdl-35916379

BACKGROUND: Cellular inflammatory processes, fibrogenesis, and apoptosis are the most characteristic pathologic features of colonic injury and colitis in human and experimental animals. Obestatin, a peptide derived from proghrelin, is reported to have significant protective and curative actions on many gastrointestinal tract inflammatory diseases, including ulcerative colitis. However, its exact protective mechanisms and the associated histopathological changes, are still in need of deeper exploration. This study explores the effect of obestatin on the course of acetic acid (AA)-induced colitis as an antifibrotic, anti-inflammatory, and anti-apoptotic agent in relation to associated tissue stress parameters. MATERIALS AND METHODS: A total of 40 healthy male albino Wistar rats weighing 200-250 g were recruited in this study. The rats were classified into four groups (10 rats each); group I: control, group II: obestatin only treated (16 nmol/kg), group III: colitis induced group (AA 1 mL of 3.5% (v/v), and group IV: AA-induced colitis + obestatin for 14 days. Colonic samples were examined after staining haematoxylin and eosin, Alcian blue, Masson trichrome. The expression of proliferating cell nuclear antigen (PCNA), nuclear factor kappa B (NFkB), and caspase-3 was estimated after immunohistochemical staining. Oxidative stress parameters, antioxidant enzymes, tissue myeloperoxidase (MPO) activity, ghrelin, and fibrogenesis markers were identified by immunoassay and colorimetric techniques. RESULTS: Colonic mucosa of group IV exhibited mucosal healing and regeneration of the surface epithelium with the restoration of the goblet cells' function together with a decline in PCNA, NFkB, and caspase-3 immunoreactivity in comparison to group III. This was accompanied by a reduction of the expression of fibrosis markers, hydroxyproline and fibronectin. In addition, tissue antioxidant status was significantly improved with a marked reduction of tissue MPO. Ghrelin level was significantly increased in comparison to group III. Group IV exhibited significant reduction in the levels of oxidative stress markers, malondialdehyde, total oxidant status with a marked increase in the activity of antioxidant enzymes, superoxide dismutase, catalase, and total cellular total antioxidant capacity. CONCLUSIONS: The concomitant treatment of obestatin inhibits the development of AA-induced colitis. The data signify that it has both curative and protective effects via antifibrotic, antioxidant, and anti-inflammatory activities.


Antioxidants , Colitis , Humans , Male , Rats , Animals , Antioxidants/pharmacology , Ghrelin/pharmacology , Caspase 3 , Proliferating Cell Nuclear Antigen/therapeutic use , Acetic Acid/toxicity , Acetic Acid/therapeutic use , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Rats, Wistar , Anti-Inflammatory Agents/adverse effects
10.
Journal of Integrative Medicine ; (12): 184-193, 2023.
Article En | WPRIM | ID: wpr-971653

OBJECTIVE@#Physical exercise, a common non-drug intervention, is an important strategy in cancer treatment, including hepatocellular carcinoma (HCC). However, the mechanism remains largely unknown. Due to the importance of hypoxia and cancer stemness in the development of HCC, the present study investigated whether the anti-HCC effect of physical exercise is related to its suppression on hypoxia and cancer stemness.@*METHODS@#A physical exercise intervention of swimming (30 min/d, 5 d/week, for 4 weeks) was administered to BALB/c nude mice bearing subcutaneous human HCC tumor. The anti-HCC effect of swimming was assessed in vivo by tumor weight monitoring, hematoxylin and eosin (HE) staining, and immunohistochemistry (IHC) detection of proliferating cell nuclear antigen (PCNA) and Ki67. The expression of stemness transcription factors, including Nanog homeobox (NANOG), octamer-binding transcription factor 4 (OCT-4), v-Myc avian myelocytomatosis viral oncogene homolog (C-MYC) and hypoxia-inducible factor-1α (HIF-1α), was detected using real-time reverse transcription polymerase chain reaction. A hypoxia probe was used to explore the intratumoral hypoxia status. Western blot was used to detect the expression of HIF-1α and proteins related to protein kinase B (Akt)/glycogen synthase kinase-3β (GSK-3β)/β-catenin signaling pathway. The IHC analysis of platelet endothelial cell adhesion molecule-1 (CD31), and the immunofluorescence co-location of CD31 and desmin were used to analyze tumor blood perfusion. SMMC-7721 cells were treated with nude mice serum. The inhibition effect on cancer stemness in vitro was detected using suspension sphere experiments and the expression of stemness transcription factors. The hypoxia status was inferred by measuring the protein and mRNA levels of HIF-1α. Further, the expression of proteins related to Akt/GSK-3β/β-catenin signaling pathway was detected.@*RESULTS@#Swimming significantly reduced the body weight and tumor weight in nude mice bearing HCC tumor. HE staining and IHC results showed a lower necrotic area ratio as well as fewer PCNA or Ki67 positive cells in mice receiving the swimming intervention. Swimming potently alleviated the intratumoral hypoxia, attenuated the cancer stemness, and inhibited the Akt/GSK-3β/β-catenin signaling pathway. Additionally, the desmin+/CD31+ ratio, rather than the number of CD31+ vessels, was significantly increased in swimming-treated mice. In vitro experiments showed that treating cells with the serum from the swimming intervention mice significantly reduced the formation of SMMC-7721 cell suspension sphere, as well as the mRNA expression level of stemness transcription factors. Consistent with the in vivo results, HIF-1α and Akt/GSK-3β/β-catenin signaling pathway were also inhibited in cells treated with serum from swimming group.@*CONCLUSION@#Swimming alleviated hypoxia and attenuated cancer stemness in HCC, through suppression of the Akt/GSK-3β/β-catenin signaling pathway. The alleviation of intratumoral hypoxia was related to the increase in blood perfusion in the tumor. Please cite this article as: Xiao CL, Zhong ZP, Lü C, Guo BJ, Chen JJ, Zhao T, Yin ZF, Li B. Physical exercise suppresses hepatocellular carcinoma progression by alleviating hypoxia and attenuating cancer stemness through the Akt/GSK-3β/β-catenin pathway. J Integr Med. 2023; 21(2): 184-193.


Humans , Animals , Mice , Carcinoma, Hepatocellular/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Proliferating Cell Nuclear Antigen/therapeutic use , Mice, Nude , Glycogen Synthase Kinase 3 beta/genetics , beta Catenin/therapeutic use , Liver Neoplasms/drug therapy , Desmin/therapeutic use , Ki-67 Antigen , Cell Line, Tumor , Hypoxia , RNA, Messenger/therapeutic use , Cell Proliferation
11.
J Food Biochem ; 46(12): e14465, 2022 12.
Article En | MEDLINE | ID: mdl-36226832

The objective of this study was to examine Nerolidol (NER) and Cisplatin (CIS) performed against human laryngeal carcinoma (Hep 2) cells. We evaluated the effect of NER, CIS, and NER + CIS on cell viability, cell migration, oxidative stress, mitochondrial membrane depolarization, nuclear condensation, apoptotic induction, and DNA damage in Hep 2 cells. We used the MTT assay to assess the cytotoxicity effect of NER and CIS on Hep 2 cells in terms of morphological alterations. Present results demonstrated that IC50 values of NER and CIS have potential cytotoxicity against Hep 2 cells. NER effectively inhibited cell viability, increased reactive oxygen species generation, apoptotic induction, and DNA damage in Hep 2 cells. In addition, the docking study evaluated the structural binding interaction of NER with PI3K/Akt and PCNA protein. Furthermore, NER with PI3K/Akt, PCNA has a higher crucial score and affinity. Present results infer that NER could be used to target signaling molecules in anticancer studies. PRACTICAL APPLICATIONS: Nerolidol is a dietary phytochemical with high biological activity that can find in a variety of plants. Many researchers focused on Nerolidol to treat various diseases including cancer. However, there is no studies exist on laryngeal cancer. This study uses Nerolidol and Cisplatin to generate oxidative stress and stimulate apoptosis and DNA damage in human laryngeal cancer cells. Based on present findings, Nerolidol could be a choice of anticancer medication, either alone or in combination against oral squamous cell carcinomas in both in vitro and in vivo experimental systems.


Carcinoma, Squamous Cell , Laryngeal Neoplasms , Humans , Cisplatin/pharmacology , Reactive Oxygen Species/metabolism , Laryngeal Neoplasms/drug therapy , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proliferating Cell Nuclear Antigen/pharmacology , Proliferating Cell Nuclear Antigen/therapeutic use , Cell Line, Tumor , Apoptosis , Carcinoma, Squamous Cell/drug therapy
12.
Sheng Li Xue Bao ; 74(3): 370-380, 2022 Jun 25.
Article Zh | MEDLINE | ID: mdl-35770635

Polycystic ovary syndrome (PCOS) is a common disease caused by complex endocrine and metabolic abnormalities in women of childbearing age. Metformin is the most widely used oral hypoglycemic drug in clinic. In recent years, metformin has been used in the treatment of PCOS, but its mechanism is not clear. In this study, we aimed to investigate the effect of metformin on PCOS and its mechanism through PCOS mouse model. Female C57BL/6J mice aged 4-5 weeks were intragastrically given letrozole (1 mg/kg daily) combined with a high-fat diet (HFD) for 21 days to establish the PCOS model. After modeling, metformin (200 mg/kg daily) was intragastrically administered. One month later, the body weight and oral glucose tolerance test (OGTT) were measured. Hematoxylin eosin (H&E) staining was used to detect the pathological changes of ovary. The serum levels of anti-Mullerian hormone (AMH), follicle-stimulating hormone (FSH), luteinizing hormone (LH), E2 and testosterone (T) were measured by ELISA. The expression of DDX4/MVH was detected by immunohistochemistry. DDX4/MVH and PCNA were co-labeled by immunofluorescence. The protein levels of DDX4/MVH, PCNA, cyclin D2, AMPK and mTOR were detected by Western blot. The results showed that after metformin treatment, the body weights of PCOS mice were gradually returned to normal, glucose tolerance was significantly improved, serum E2 levels were increased, while AMH, LH, T levels and LH/FSH ratio were decreased. Ovarian polycystic lesions were reduced with reduced atresia follicles. Furthermore, the number of proliferative female germline stem cells (FGSCs) and levels of proliferation related proteins (PCNA, cyclin D2) were significantly increased, and the p-mTOR and p-AMPK levels were markedly up-regulated. These results suggest that metformin treatment not only improves hyperandrogenemia, glucose intolerance and polycystic ovarian lesions in PCOS, but also activates the function of FGSCs. The underlying mechanism may be related to the phosphorylation of AMPK and mTOR. These findings provide new evidence to use metformin in the treatment of PCOS and follicular development disorder.


Metformin , Oogonial Stem Cells , Ovarian Cysts , Ovarian Neoplasms , Polycystic Ovary Syndrome , AMP-Activated Protein Kinases , Animals , Cyclin D2 , Female , Follicle Stimulating Hormone/therapeutic use , Humans , Luteinizing Hormone/therapeutic use , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Oogonial Stem Cells/metabolism , Ovarian Cysts/drug therapy , Polycystic Ovary Syndrome/drug therapy , Proliferating Cell Nuclear Antigen/therapeutic use , TOR Serine-Threonine Kinases
13.
Environ Sci Pollut Res Int ; 29(45): 68830-68841, 2022 Sep.
Article En | MEDLINE | ID: mdl-35554805

This study evaluated the protective potentials of Moringa oleifera leaf alcoholic extract (MOLE) against bisphenol A (BPA)-induced stomach ulceration and inflammation in rats. Control rats received olive oil. Second group administered MOLE (200 mg/kg bwt) by oral gavage. Third group was given BPA (50 mg/ kg bwt) for 4 weeks. Fourth group administrated BPA and MOLE simultaneously. Fifth group was given MOLE for 4 weeks then administered BPA and MOLE for another 4 weeks. Bisphenol A induced gastric ulceration and decreased the volume of gastric juice, prostaglandin E2 (PGE2), reduced glutathione (GSH) and interleukin 10 (IL-10) contents, superoxide dismutase (SOD) activity, and proliferating cell nuclear antigen (PCNA) protein in stomach tissues, while increased the titratable acidity, malondialdehyde (MDA), tumor necrosis factor alpha (TNF-α) and interleukin 6 (IL-6) contents, and caspase-3 and NF­κB proteins in stomach tissue. However, MOLE ameliorated BPA-induced gastric ulceration and significantly increased the volume of gastric juice, PGE2, GSH and IL-10 contents, SOD activity, and PCNA protein while significantly decreased titratable acidity, MDA, TNF-α and IL-6 contents, and of NF­κB and caspase-3 proteins in gastric tissue. This study indicated that MOLE protected stomach against BPA-induced gastric injury via its anti-oxidant, anti-apoptotic, and anti-inflammatory activities.


Moringa oleifera , Stomach Ulcer , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/metabolism , Benzhydryl Compounds , Caspase 3 , Dinoprostone/metabolism , Dinoprostone/therapeutic use , Dinoprostone/toxicity , Glutathione/metabolism , Interleukin-10 , Interleukin-6 , Malondialdehyde/metabolism , Moringa oleifera/metabolism , NF-kappa B/metabolism , Olive Oil , Phenols , Plant Extracts/therapeutic use , Proliferating Cell Nuclear Antigen/therapeutic use , Rats , Stomach Ulcer/chemically induced , Stomach Ulcer/drug therapy , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha
14.
Curr Mol Pharmacol ; 15(2): 434-445, 2022.
Article En | MEDLINE | ID: mdl-34061012

BACKGROUND: Tacrolimus is a calcineurin inhibitor widely used for immunological disorders. However, there is significant controversy regarding its effect on the liver. The present study was conducted to evaluate the anticancer effects of tacrolimus on an induced murine hepatocellular carcinoma (HCC) model and its possible hepatotoxicity at standard therapeutic doses. METHODS: Fifty-four male mice were divided into five groups: a control healthy group, control HCC group, tacrolimus-treated group, doxorubicin (DOXO)-treated group, and combined tacrolimus- and DOXO-treated group. The activity of liver enzymes, including alkaline phosphatase, gamma- glutamyl transferase, lactate dehydrogenase, alanine transaminase, and aspartate transaminase, was determined. Serum vascular endothelial growth factor (VEGF) was measured using an enzyme- linked immunosorbent assay. A quantitative real time- polymerase chain reaction (qRTPCR) was conducted to measure the expression of proliferating cell nuclear antigen (PCNA), Bax, and p53 mRNA. Immunohistochemical staining for cyclin D1 and VEGF was performed. RESULTS: Mice that received combined treatment with tacrolimus and DOXO exhibited the best improvement in all parameters when compared with the groups that received DOXO or tacrolimus alone (p < 0.001). CONCLUSION: The combination of DOXO and tacrolimus was more effective in the management of HCC compared with either agent alone. This improvement was detected by the reduction of liver enzymes and the improvement of the histopathological profile. The involved mechanisms included significant apoptosis induction demonstrated by upregulation of bax along with a reduction in angiogenesis demonstrated by downregulation of VEGF. This was accompanied by inhibition of cell cycle progression mediated by upregulated p53 and downregulated PCNA and cyclin D1.


Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cyclin D1/metabolism , Cyclin D1/therapeutic use , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Male , Mice , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/therapeutic use , Tacrolimus/pharmacology , Tacrolimus/therapeutic use , Tumor Suppressor Protein p53 , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/therapeutic use , bcl-2-Associated X Protein/metabolism
15.
Acta Physiologica Sinica ; (6): 370-380, 2022.
Article Zh | WPRIM | ID: wpr-939572

Polycystic ovary syndrome (PCOS) is a common disease caused by complex endocrine and metabolic abnormalities in women of childbearing age. Metformin is the most widely used oral hypoglycemic drug in clinic. In recent years, metformin has been used in the treatment of PCOS, but its mechanism is not clear. In this study, we aimed to investigate the effect of metformin on PCOS and its mechanism through PCOS mouse model. Female C57BL/6J mice aged 4-5 weeks were intragastrically given letrozole (1 mg/kg daily) combined with a high-fat diet (HFD) for 21 days to establish the PCOS model. After modeling, metformin (200 mg/kg daily) was intragastrically administered. One month later, the body weight and oral glucose tolerance test (OGTT) were measured. Hematoxylin eosin (H&E) staining was used to detect the pathological changes of ovary. The serum levels of anti-Mullerian hormone (AMH), follicle-stimulating hormone (FSH), luteinizing hormone (LH), E2 and testosterone (T) were measured by ELISA. The expression of DDX4/MVH was detected by immunohistochemistry. DDX4/MVH and PCNA were co-labeled by immunofluorescence. The protein levels of DDX4/MVH, PCNA, cyclin D2, AMPK and mTOR were detected by Western blot. The results showed that after metformin treatment, the body weights of PCOS mice were gradually returned to normal, glucose tolerance was significantly improved, serum E2 levels were increased, while AMH, LH, T levels and LH/FSH ratio were decreased. Ovarian polycystic lesions were reduced with reduced atresia follicles. Furthermore, the number of proliferative female germline stem cells (FGSCs) and levels of proliferation related proteins (PCNA, cyclin D2) were significantly increased, and the p-mTOR and p-AMPK levels were markedly up-regulated. These results suggest that metformin treatment not only improves hyperandrogenemia, glucose intolerance and polycystic ovarian lesions in PCOS, but also activates the function of FGSCs. The underlying mechanism may be related to the phosphorylation of AMPK and mTOR. These findings provide new evidence to use metformin in the treatment of PCOS and follicular development disorder.


Animals , Female , Humans , Mice , AMP-Activated Protein Kinases , Cyclin D2 , Follicle Stimulating Hormone/therapeutic use , Luteinizing Hormone/therapeutic use , Metformin/pharmacology , Mice, Inbred C57BL , Oogonial Stem Cells/metabolism , Ovarian Cysts/drug therapy , Ovarian Neoplasms , Polycystic Ovary Syndrome/drug therapy , Proliferating Cell Nuclear Antigen/therapeutic use , TOR Serine-Threonine Kinases
16.
J Nat Prod ; 83(4): 1183-1189, 2020 04 24.
Article En | MEDLINE | ID: mdl-32191471

Oleanolic acid (OA) is a natural, biologically active pentacyclic triterpenoid found in Cornus officinalis. Although C. officinalis and OA have antiproliferative actions, the effects and mechanisms of OA in benign prostatic hyperplasia (BPH) are unclear. We examined the effect of OA in an animal model of testosterone-induced BPH. Male rats were injected with testosterone propionate with or without OA. The inhibitory effect of OA on BPH-1 cells was determined in vitro. Rats with BPH exhibited outstanding BPH symptoms, including prostatic enlargement, upregulated dihydrotestosterone and 5α-reductase 2 levels, and histological changes. Compared with the BPH group, the OA group showed fewer pathological alterations and regular androgen events. OA inhibited prostate cell proliferation by downregulating the expression of proliferating cell nuclear antigen (PCNA) and cell cycle markers in BPH-induced animals. This indicated that OA has superior therapeutic effect in the BPH animal model than finasteride. In vitro studies demonstrated upregulation of PCNA and cell cycle proteins, whereas OA clearly reduced this upregulation. Thus, OA may inhibit the development of BPH by targeting cell cycle progression markers. These suggest that OA is a potential agent for BPH treatment.


Oleanolic Acid/pharmacology , Proliferating Cell Nuclear Antigen/therapeutic use , Prostatic Hyperplasia/drug therapy , Testosterone/chemistry , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Humans , Male , Molecular Structure , Oleanolic Acid/chemistry , Proliferating Cell Nuclear Antigen/metabolism , Proliferating Cell Nuclear Antigen/pharmacology , Prostatic Hyperplasia/pathology , Rats , Testosterone/metabolism , Testosterone Propionate/adverse effects
17.
J Nanobiotechnology ; 15(1): 62, 2017 Sep 06.
Article En | MEDLINE | ID: mdl-28877692

BACKGROUND: The parasitic disease malaria remains a major global public health concern and no truly effective vaccine exists. One approach to the development of a malaria vaccine is to target the asexual blood stage that results in clinical symptoms. Most attempts have failed. New antigens such as P27A and P27 have emerged as potential new vaccine candidates. Multiple studies have demonstrated that antigens are more immunogenic and are better correlated with protection when presented on particulate delivery systems. One such particulate delivery system is the self-assembling protein nanoparticle (SAPN) that relies on coiled-coil domains of proteins to form stable nanoparticles. In the past we have used de novo designed amino acid domains to drive the formation of the coiled-coil scaffolds which present the antigenic epitopes on the particle surface. RESULTS: Here we use naturally occurring domains found in the tex1 protein to form the coiled-coil scaffolding of the nanoparticle. Thus, by engineering P27A and a new extended form of the coiled-coil domain P27 onto the N and C terminus of the SAPN protein monomer we have developed a particulate delivery system that effectively displays both antigens on a single particle that uses malaria tex1 sequences to form the nanoparticle scaffold. These particles are immunogenic in a murine model and induce immune responses similar to the ones observed in seropositive individuals in malaria endemic regions. CONCLUSIONS: We demonstrate that our P27/P27A-SAPNs induce an immune response akin to the one in seropositive individuals in Burkina Faso. Since P27 is highly conserved among different Plasmodium species, these novel SAPNs may even provide cross-protection between Plasmodium falciparum and Plasmodium vivax the two major human malaria pathogens. As the SAPNs are also easy to manufacture and store they can be delivered to the population in need without complication thus providing a low cost malaria vaccine.


Antigens, Protozoan/therapeutic use , Malaria Vaccines/therapeutic use , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Nanoparticles/therapeutic use , Plasmodium falciparum/immunology , Proliferating Cell Nuclear Antigen/therapeutic use , Protozoan Proteins/therapeutic use , Amino Acid Sequence , Animals , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Humans , Immunization , Malaria Vaccines/chemistry , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Mice , Mice, Inbred BALB C , Models, Molecular , Nanoparticles/chemistry , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/immunology , Protein Domains , Protein Engineering , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/immunology
19.
Rev. venez. oncol ; 25(1): 2-9, ene.-mar. 2013. tab
Article Es | LILACS | ID: lil-718960

Evaluación molecular de márgenes de resección en pacientes con carcinoma de células escamosas de cavidad oral sometidos a cirugía. 16 pacientes con carcinoma escamoso de cavidad oral, en cualquiera de sus localizaciones, sin tratamientos previos, intervenidos quirúrgicamente en el 2011. La pieza operatoria fue procesada por anatomía patológica a través del método tradicional, realizándose cortes adicionales que incluían: tumor y 0,5 cm de margen no tumoral. Se realizó hematoxilina-eosina y complementó con inmunomarcaje para p53, PCNA, Ki-67, factor de crecimiento epidérmico y receptor de crecimiento endotelial vascular. De los 16 pacientes en estudio la mayoría eran del género masculino, la edad promedio fue cercana a los 60 años, la mayoría eran pacientes consumidores de tabaco y alcohol. La lengua fue la localización más frecuente y los tumores se encontraban en un estadio avanzado (estadio III y IV). Estudio molecular: todos los marcadores evaluados se encontraban positivos en los márgenes de resección en el 93,75% de los pacientes. Los marcadores de proliferación celular como el PCNA y Ki-67 así como el p-53 se encontraban positivos entre 1,5 cm a 2 cm del tumor con un marcaje intenso. Por el contrario, el factor de crecimiento epidérmico el receptor de crecimiento endotelial vascular se encontraban positivos hasta 1,5 cm pero con menor intensidad. En el cáncer oral podemos observar con frecuencia cambios moleculares en el tejido aparentemente sano que rodea el tumor hasta por lo menos 15 mm.


The molecular evaluation of resection margins in patients with squamous cell carcinoma of oral cavity who underwent surgery. Field of cancerization concept. We included 16 patients with oral cavity squamous cell carcinoma in any of their locations,without pre treatment, surgically treated in our hospital in the 2011 year. The surgical specimen was processed by the pathology department of our institution, through the traditional method, additional sectioned including the tumor and at least 0.5 cm margin non tumorigenic. Study was performed hematoxylin eosin and was supplemented with immunostaining for p53, PCNA, Ki-67, epidermal growth factor receptor and vascular endothelial growth factor receptor. The most important features of the 16 patients studied were: The majorities were male, the average age was around 60 years old; most of them were tobacco and alcohol consumers. The tongue was the most frequent location and most of the tumors were in an advanced stage (stage III y IV). In molecular evaluation all the markers were positive in the resection margins in 93.75% of all patients. The cell proliferation markers suchas PCNA and Ki-67 and the p-53 were positive 1.5 cm to2 cm tumor with intense staining. Conversely, epidermal receptor grow factors and vascular endothelial grow factor receptor were positive up to 1.5 cm but with less intensity. In oral cancer can often observe molecular changes in the apparently healthy tissue surrounding the tumor to at least 15 mm.


Humans , Male , Female , Middle Aged , /therapeutic use , Proliferating Cell Nuclear Antigen/therapeutic use , Mouth/injuries , Carcinoma, Squamous Cell/diagnosis , Genes, erbB-1 , Head and Neck Neoplasms/diagnosis , Vascular Endothelial Growth Factor Receptor-1/therapeutic use , Dentistry , Medical Oncology
...