Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 169
Filter
2.
Sci Rep ; 14(1): 6515, 2024 03 18.
Article in English | MEDLINE | ID: mdl-38499634

ABSTRACT

Human pancreatic ductal adenocarcinoma (PDAC) is a highly malignant and lethal tumor of the exocrine pancreas. Cannabinoids extracted from the hemp plant Cannabis sativa have been suggested as a potential therapeutic agent in several human tumors. However, the anti-tumor effect of cannabinoids on human PDAC is not entirely clarified. In this study, the anti-proliferative and apoptotic effect of cannabinoid solution (THC:CBD at 1:6) at a dose of 1, 5, and 10 mg/kg body weight compared to the negative control (sesame oil) and positive control (5-fluorouracil) was investigated in human PDAC xenograft nude mice model. The findings showed that cannabinoids significantly decreased the mitotic cells and mitotic/apoptotic ratio, meanwhile dramatically increased the apoptotic cells. Parallelly, cannabinoids significantly downregulated Ki-67 and PCNA expression levels. Interestingly, cannabinoids upregulated BAX, BAX/BCL-2 ratio, and Caspase-3, meanwhile, downregulated BCL-2 expression level and could not change Caspase-8 expression level. These findings suggest that cannabinoid solution (THC:CBD at 1:6) could inhibit proliferation and induce apoptosis in human PDAC xenograft models. Cannabinoids, including THC:CBD, should be further studied for use as the potent PDCA therapeutic agent in humans.


Subject(s)
Cannabinoids , Cannabis , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Mice , Humans , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Mice, Nude , Heterografts , bcl-2-Associated X Protein , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2
3.
In Vivo ; 38(1): 69-72, 2024.
Article in English | MEDLINE | ID: mdl-38148053

ABSTRACT

BACKGROUND/AIM: Breast-cancer metastasis to the brain is an intractable disease. To discover improved therapy for this disease, we developed a precise non-invasively-imageable orthotopic nude-mouse model, using very-narrow-band-width laser fluorescence excitation. MATERIALS AND METHODS: Female nu/nu nude mice, aged 4-8 weeks, were inoculated through the midline of the skull with triple-negative human MDA-MB-231 breast cancer cells (5×105) expressing red fluorescent protein (RFP). The mice were imaged with the Analytik Jena UVP Biospectrum Advanced at 520 nm excitation with peak emission at 605 nm. RESULTS: Three weeks after injection of MDA-MB-231-RFP cells in the brain, non-invasive fluorescence images of the breast tumor growing on the brain were obtained. The images of the tumor were very bright, with well-defined margins with no detectable skin autofluorescence background. Images obtained at various angles showed that the extent of the tumor margins could be precisely measured. A skin flap over the skull confirmed that the tumor was growing on the surface of the brain which is a frequent occurrence in breast cancer. CONCLUSION: A precise orthotopic model of RFP-expressing breast-cancer metastasis to the brain was developed that could be non-invasively imaged with very-narrow-band-width laser excitation, resulting in an ultra-bright, ultra-low-background signal. The model will be useful in discovering improved therapeutics for this recalcitrant disease.


Subject(s)
Breast Neoplasms , Melanoma , Neoplasms, Second Primary , Skin Neoplasms , Mice , Female , Humans , Animals , Red Fluorescent Protein , Breast Neoplasms/diagnostic imaging , Mice, Nude , Disease Models, Animal , Optical Imaging , Brain/diagnostic imaging , Green Fluorescent Proteins , Cell Line, Tumor
4.
Biomed Pharmacother ; 169: 115922, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-38011786

ABSTRACT

Regenerating family protein 3 A (Reg3A) is highly expressed in a variety of organs and inflammatory tissues, and is closely related to tumorigenesis and cancer progression. However, clinical statistics show that high expression of Reg3A is associated with better prognosis in colorectal cancer (CRC) patients, suggesting a tumor-suppressive effect. The precise action and underlying mechanism of Reg3A in CRC remain controversial. The present study sought to investigate the relationship among Reg3A expression, CRC development, and immune cell alteration in patients using the TCGA, GEPIA, PrognoScan, TIMER and TISIDB databases. Reg3A-overexpressing LoVo cell line (LoVo-Reg3A), a representative of colon adenocarcinoma (COAD), was constructed and the action of Reg3A was assessed in a xenograft nude mouse model. Our bioinformatical analyses revealed that Reg3A upregulation is highly associated with CRC, along with increased frequency of immune cell infiltration. In the xenograft nude mice, Reg3A overexpression offered a tumor-suppressive effect by inhibiting cell proliferation and promoting apoptosis. The result of RNA-seq suggested a positive regulation of leukocytes and an upregulation of T cells in LoVo-Reg3A tumor tissue. CD4+ and CD8+ T cells in tumors, splenic Reg3A-reactive IFN-γ+/CD4+ T cells, and serum TNF-α, IFN-γ and IL-17 were significantly increased by Reg3A overexpression. In the ex vivo co-culture experiment, elevated cytotoxic effect, increased proportion of CD3ε+ T cells, and upregulated expressions of TNF-α, IFN-γ and IL-17 were detected in the PBMCs isolated from LoVo-Reg3A cell-xenografted nude mice. In conclusion, high expression of Reg3A could activate and recruit T cells in COAD leading to the cytotoxic tumor-suppressive effect.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Animals , Humans , Mice , CD8-Positive T-Lymphocytes , Colonic Neoplasms/genetics , Interleukin-17 , Mice, Nude , Tumor Necrosis Factor-alpha
5.
Am J Transl Res ; 15(5): 3410-3423, 2023.
Article in English | MEDLINE | ID: mdl-37303681

ABSTRACT

OBJECTIVE: To investigate the inhibitory effects and mechanisms of triterpenoids from Ganoderma lucidum (G. lucidum triterpenoids) on the growth and metastasis of hepatocellular carcinoma (HCC) both in vitro and in vivo. METHODS: In in-vitro experiments, the inhibitory effects of G. lucidum triterpenoids on human HCC SMMC-7721 cell lines were investigated by observing the proliferation, apoptosis, migration and invasion phenotypes of the cell line and assessing the cell cycles as well as the cell apoptosis and proliferation. In in-vivo experiments, nude mouse SMMC-7721 tumor models were established and divided into control group, treatment group A (low concentration group) and treatment group B (high concentration group) according to the treatment models received. Magnetic resonance imaging (MRI) was performed 3 times on each mouse model to calculate their tumor volumes. The liver and kidney functions of the models were evaluated. Tissues harvested from their solid organs were subjected to HE staining, and the tumor tissues were subjected to HE staining and immunohistochemical staining (E-cad, Ki-67, and Tunel), respectively. RESULTS: i. In in-vitro experiments, G. lucidum triterpenoids could inhibit the growth of human HCC SMMC-7721 cell lines via regulating their proliferation and apoptosis phenotype. ii. In in-vivo experiments, the comparison of tumor volumes of mouse models obtained from the second and third MIR scanning was found to be statistically significant between the control group and treatment group A (P<0.05); and statistically significant differences were also found in the tumor volumes from the second and third MRI scanning between the control group and treatment group B (P<0.05). iii. No significant acute injuries or adverse effects were observed in the liver or kidney of the nude mice. CONCLUSION: G. lucidum triterpenoids could inhibit the growth of tumor cells via blocking their proliferation, accelerating apoptosis, and inhibiting migration and invasion, without marked toxic effects on normal organs and tissues in the body.

6.
J Nutr ; 153(5): 1618-1626, 2023 05.
Article in English | MEDLINE | ID: mdl-37011879

ABSTRACT

BACKGROUND: The ability of soluble dietary fibers (SDFs) to induce the production of IgA, especially in the intestine, is one of the health benefits of SDFs, but the mechanism involved is unclear. OBJECTIVE: This study was designed to identify the relationship between the induction of IgA by SDFs and the cecal short-chain fatty acid (SCFA) content, and to evaluate the importance of T cell-independent IgA production for SDF-induced IgA production. METHODS: We compared 3 SDFs-fructooligosaccharides (FO), indigestible glucan (IG), and polydextrose (PD). Male BALB/cAJcl mice or T cell-deficient BALB/cAJcl-nu/nu (nude) mice were fed diets supplemented with 1 SDF (3% w/w) for 10 wk, and IgA content in their feces, plasma, lung, and submandibular gland was measured. RESULTS: In BALB/cAJcl mice, the consumption of all 3 SDF diets induced fecal IgA production, but the response was stronger in the IG and PD groups than in the FO group. The IgA concentrations of the plasma and lung were also higher in the FO and PD groups, and these groups showed significantly higher cecal acetic and n-butyric acid content. In contrast, in nude mice, the induction of IgA production was identified only in fecal samples of mice fed the 3 SDF diets, although there were significant increases in cecal SCFA content. CONCLUSIONS: The induction of IgA production by SDFs occurred independent of T cells in the intestine, but in the plasma, lung, and submandibular gland it was T-cell dependent. SCFAs generated in the large intestine may influence the systemic immune system, but there is no clear relationship between the generation of SCFAs and intestinal IgA production in response to SDF consumption.


Subject(s)
Fatty Acids, Volatile , Intestines , Mice , Male , Animals , Mice, Nude , Dietary Fiber/pharmacology , Immunoglobulin A
7.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-993149

ABSTRACT

Objective:To evaluate the effects of high mobility group protein box 1 (HMGB1) on clinical prognosis of esophagus squamous cell carcinoma (ESCC) patients treated with chemoradiotherapy and the radiosensitivity of xenograft in nude mice.Methods:A total of 90 endoscopic biopsy specimens were obtained from ESCC patients treated with chemoradiotherapy. The expression level of HMGB1 was determined by immunohistochemical staining. High expression level was defined when staining was observed on ≥50% of the tumor cells. All patients were divided into the high expression group ( n=48) and low expression group ( n=42), and their survival information was retrospectively analyzed. Cell transfection was performed with the plasmid carrying human HMGB1-shRNA to knockdown HMGB1 expression in ECA109 cells and xenograft mouse models were established. The tumor volume and mass were calculated after irradiation with a dose of 15 Gy. The cell apoptosis in xenograft tissues were detected. Survival analysis was performed using Kaplan-Meier method. Univariate prognostic analysis was conducted by log-rank test. Intergroup comparison was performed by analysis of variance (ANOVA). Results:The expression level of HMGB1 was significantly associated with gross tumor volume, longest diameter of tumor, T staging and distant metastasis ( χ2=9.663, 5.625, 4.068, 7.146, all P<0.05). In the low expression group, the overall survival (OS) ( χ2=4.826, P=0.028), progression-free survival (PFS) ( χ2=4.390, P=0.036) were longer compared with that in the high expression group. Further analysis of HMGB1-high expression patients showed that the radiation dose and the combination of chemoradiotherapy did not significantly affect the OS or PFS of ESCC patients. We observed that knockdown of HMGB1 slowed the growth rate of xenograft, decreased the tumor volume and increased the apoptosis rate after irradiation. Conclusions:ESCC patients with high expression level of HMGB1 obtain poor prognosis after chemoradiotherapy, which can be enhanced by increasing the sensitivity to radiotherapy and chemotherapy. HMGB1 knockdown can effectively increase the radiosensitivity of xenograft in ESCC nude mice.

8.
Mol Ther Oncolytics ; 27: 239-255, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36458203

ABSTRACT

Historically, the clinical utility of oncolytic virotherapy as a treatment for a wide range of cancer types was first demonstrated by three pilot human clinical trials conducted in Japan in the 1970s and 1980s using a wild-type Urabe mumps virus (MuV) clinical isolate. Using a sample of the actual original oncolytic Urabe MuV clinical trial virus stock (MuV-U-Japan) used in these Japanese clinical trials, we found that MuV-U-Japan consisted of a wide variety of very closely related Urabe MuVs that differed by an average of only three amino acids. Two MuV-U-Japan isolates, MuV-UA and MuV-UC, potently killed a panel of established human breast cancer cell lines in vitro, significantly extended survival of nude mice with human triple-negative breast cancer (TNBC) MDA-MB-231 tumor xenografts in vivo, and demonstrated significant killing activity against breast cancer patient-derived xenograft (PDX) cell lines grown as 3D organoids, including PDXs from patients resistant to anthracycline- and taxane-based chemotherapy. We also report success in developing a large-scale MuV-U production and purification process suitable for supporting Investigational New Drug applications for clinical trials. This study demonstrates the suitability of the MuV-UC virus for translation to modern clinical trials for treating patients with TNBC.

9.
Int J Mol Sci ; 23(21)2022 Nov 02.
Article in English | MEDLINE | ID: mdl-36362174

ABSTRACT

From previous studies of negatively correlating the expression of human METCAM/MUC18 with the pathology of nasopharyngeal carcinoma (NPC), we have suggested that human METCAM/MUC18 (huMETCAM/MUC18) might play a tumor suppressor role in the development of nasopharyngeal carcinoma. To scrutinize this hypothesis, we investigated the effects of huMETCAM/MUC18's over-expression on in vitro cellular behavior and on the in vivo tumorigenesis of one NPC cell line (NPC-TW01). HuMETCAM/MUC18 cDNA was first transfected into the NPC-TW01 cell line, which was established from NPC type I, and many G418-resistant clones were obtained. Then, two NPC-TW01 clones, which expressed high and medium levels of huMETCAM/MUC18, respectively, and one empty vector (control) clone were used to test the effects of huMETCAM/MUC18's over-expression on in vitro behaviors and on in vivo tumorigenesis (via subcutaneous injection) in athymic nude mice (Balb/cAnN.Cg-Foxnlnu/Cr1Nar1). The time course of tumor proliferation and the final tumor weights were determined. Tumor sections were used for the histology and immunohistochemistry (IHC) studies. Tumor lysates were used for determining the expression levels of huMETCAM/MUC18 and various downstream key effectors. HuMETCAM/MUC18's over-expression reduced in vitro motility and invasiveness and altered growth behaviors in 3D basement membrane culture assays, and it decreased the in vivo tumorigenicity of the NPC-TW01 cells. The tumor cells from a high-expressing clone were clustered and confined in small areas, whereas those from a vector control clone were more spread out, suggesting that the tumor cells from the high-expressing clone appeared to stay dormant in micro-clusters. Expression levels of the proliferation index, an index of the metabolic switch to aerobic glycolysis, angiogenesis indexes, and survival pathway indexes were reduced, whereas the pro-apoptosis index increased in the corresponding tumors. The over-expression of huMETCAM/MUC18 in the NPC-TW01 cells decreased the epithelial-to-mesenchymal transition and the in vitro and in vitro tumorigenesis, suggesting that it plays a tumor suppressor role in the development of type I NPC, perhaps by increasing apoptosis and decreasing angiogenesis, proliferation, and the metabolic switch to aerobic glycolysis.


Subject(s)
Carcinogenesis , Nasopharyngeal Neoplasms , Mice , Animals , Humans , Nasopharyngeal Carcinoma/genetics , Cell Line, Tumor , Mice, Nude , CD146 Antigen/genetics , Carcinogenesis/genetics , Cell Proliferation , Neovascularization, Pathologic/genetics , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Cell Movement/genetics , Gene Expression Regulation, Neoplastic
10.
Front Immunol ; 13: 1007579, 2022.
Article in English | MEDLINE | ID: mdl-36341323

ABSTRACT

Animal models play an indispensable role in the study of human diseases. However, animal models of different diseases do not fully mimic the complex internal environment of humans. Immunodeficient mice are deficient in certain genes and do not express these or show reduced expression in some of their cells, facilitating the establishment of humanized mice and simulation of the human environment in vivo. Here, we summarize the developments in immunodeficient mice, from the initial nude mice lacking T lymphocytes to NOD/SCID rgnull mice lacking T, B, and NK cell populations. We describe existing humanized immune system mouse models based on immunodeficient mice in which human cells or tissues have been transplanted to establish a human immune system, including humanized-peripheral blood mononuclear cells (Hu-PBMCs), humanized hematopoietic stem cells (Hu-HSCs), and humanized bone marrow, liver, thymus (Hu-BLT) mouse models. The different methods for their development involve varying levels of complexity and humanization. Humanized mice are widely used in the study of various diseases to provide a transitional stage for clinical research. However, several challenges persist, including improving the efficiency of reconstructing the human B cell immune response, extending lifespan, improving the survival rate of mice to extend the observation period, and improving the development of standardized commercialized models and as well as their use. Overall, there are many opportunities and challenges in the development of humanized immune system mouse models which can provide novel strategies for understanding the mechanisms and treatments of human disease.


Subject(s)
Leukocytes, Mononuclear , Mice , Humans , Animals , Mice, SCID , Mice, Inbred NOD , Mice, Nude , Disease Models, Animal , Mice, Knockout
11.
J Med Virol ; 94(12): 6000-6015, 2022 12.
Article in English | MEDLINE | ID: mdl-35965331

ABSTRACT

Oncolytic viruses (OVs), including oncolytic herpes simplex viruses (oHSVs), are promising therapeutics against cancer. Here, we report two ICP6-mutated HSVs (type I) generated by CRISPR/Cas9, rHSV1/∆RR (with ICP6 ribonucleotide reductase [RR] domain deleted) and rHSV1/∆ICP6 (with a complete deletion of ICP6), exhibiting potent antitumor efficacy against lung adenocarcinoma. Both the mutants showed strong cytotoxicity in vitro, comparable with the control viruses expressing intact ICP6, but in relatively lower titers. Moreover, these mutant viruses exhibited preferential killing ability against lung tumor cells rather than normal lung fibroblast cells. Further, unlike the control HSV-1 causing severe illness or death in the mouse model, the ICP6-mutated viruses did not induce significant pathogenicity but instead effectively reduced tumor burden in vivo and led to 100% survival of the animals, indicating notable antitumor activity and attenuated virulence. In addition, rHSV1/∆RR seemed to have even better antitumor efficacy than rHSV1/∆ICP6, albeit no statistical significance in inhibition of tumor volume. Histopathologically, rHSV1/∆RR induced massive neutrophil infiltration to the tumor microenvironment and consistently, triggered more antitumor immune and neutrophil chemotactic cytokines or higher expression levels of them (indicated by quantitative polymerase chain reaction and transcriptome analyses). These results demonstrate the anti-adenocarcinoma potential of the CRISPR/Cas9-engineered ICP6 mutant HSV1, especially the rHSV1/∆RR, which likely induces stronger innate antitumor immune response. Together, these findings may provide new valuable clues for further development of OV-based therapeutics against lung adenocarcinoma or other types of tumors.


Subject(s)
Adenocarcinoma of Lung , Herpesvirus 1, Human , Lung Neoplasms , Ribonucleotide Reductases , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/therapy , Animals , CRISPR-Cas Systems , Disease Models, Animal , Herpesvirus 1, Human/genetics , Heterografts , Humans , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Mice , Ribonucleotide Reductases/genetics , Tumor Microenvironment
12.
In Vivo ; 36(4): 1603-1607, 2022.
Article in English | MEDLINE | ID: mdl-35738609

ABSTRACT

BACKGROUND: Cancer of the Meckel's diverticulum (MD) is extremely rare. It is often advanced at the time of operation and the prognosis is poor. An effective treatment for this cancer has not yet been developed and there is no MD-carcinoma mouse model. MATERIALS AND METHODS: MD carcinoma was established as a patient-derived xenograft (PDX) in 5-week-old male nude mice by subcutaneous transplantation of surgical specimens together with surrounding normal tissue. Hematoxylin and eosin (H&E) staining was performed on paraffin-embedded tissue sections of the original tumor resected from patients and transplanted tumors grown in nude mice. RESULTS: Three of five mice implanted with MD tumor fragments grew. MD-carcinoma histopathology, observed with H&E-stained tissue sections of the tumors grown in the mice and tumor from the original patient, was concordant. Both showed the luminal structures characteristic of MD carcinoma, and the lumens were filled with serous fluid. CONCLUSION: The first PDX mouse model of MD carcinoma has been established. The PDX model maintained MD-carcinoma histology of the tumor in the patient. The MD carcinoma mouse model will enable basic research on MD carcinoma, as well as the testing of novel therapeutic agents.


Subject(s)
Carcinoma , Meckel Diverticulum , Animals , Disease Models, Animal , Humans , Male , Meckel Diverticulum/pathology , Meckel Diverticulum/surgery , Mice , Mice, Nude , Prognosis , Treatment Outcome
14.
Anticancer Res ; 42(2): 739-743, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35093872

ABSTRACT

BACKGROUND/AIM: Our laboratory pioneered the patient-derived orthotopic xenograft (PDOX) model. An important goal of PDOX-model development is facile visualization of metastasis in live mice. In the present report we evaluated tumor growth and metastasis in pancreatic cancer PDOX NOG [Non-obese diabetes (NOD)/Scid/IL2Rγnull]-and nude-mouse models using red fluorescent protein (RFP)-expressing tumor stroma to visualize the primary tumor and metastasis. MATERIALS AND METHODS: A patient-derived pancreatic cancer was initially implanted in transgenic RFP-expressing nude mice. Then, tumor fragments, which acquired RFP expressing stroma while growing in RFP-expressing nude mice were orthotopically implanted in nude and NOG mice. The primary pancreatic tumor and metastasis were observed 8 weeks after implantation. RESULTS: Lymph-node metastases expressing red fluorescence were detected only in NOG mice. Significantly faster growth of primary pancreatic tumors and a higher incidence of lymph-node metastasis occurred in NOG mice compared to nude mice. CONCLUSION: RFP-expressing tumor stroma, which traffics together with cancer cells to lymph nodes, is useful to observe tumor behavior, such as lymph-node metastasis in a PDOX NOG-mouse model which can be used for evaluation of novel anti-metastatic agents, as well as personalized therapy to identify effective drugs.


Subject(s)
Disease Models, Animal , Pancreatic Neoplasms/pathology , Animals , Humans , Intravital Microscopy , Luminescent Proteins/metabolism , Lymphatic Metastasis , Mice , Mice, Nude , Mice, SCID , Mice, Transgenic , Neoplasm Transplantation , Pancreatic Neoplasms/metabolism , Red Fluorescent Protein
15.
Acta Anatomica Sinica ; (6): 82-91, 2022.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1015373

ABSTRACT

Objective To explore the relationship between miR-26a and metadherin (MTDH), and to verify the relationship between miR-26a and MTDH in vivo in nude mice. Methods Immunohistochemical SP staining method was used to detect the expression of MTDH and in situ hybridization was used to detect the expression of miR-26a in 86 cases of esophageal cancer, and the correlation between the expressions was analyzed. The bioinformatics prediction Targetscan Human 7. 2 software could predicte the binding fragment of MTDH on the miR-26a sequence, and the luciferase report experiment was used to verify the targeted regulatory relationship between MTDH and miR-26a. Nude mice were injected esophageal cancer cell lines subcutaneously which were lentiviral interferenced and overexpressed miR-26a to observe the fonnation of tumors. The tumors from nude mice were made into paraffin, and each was detected. The expression of MTDH in miR-26a in the tumor groups was detected by immunohistochemical staining and in situ hybridization and the relationship was analyzed. Results The expression of miR-26a in esophageal cancer tissues was significantly lower than that in paired nonnal esophageal tissues, and the expression of MTDH in esophageal cancer tissues was significantly higher than that in paired normal esophageal tissues. The expression of miR-26a was related to the patient' s pathological grade (P<0. 05), N stage(P<0. 05), and tumor volume (P<0. 01). The expression of MTDH in esophageal cancer was related to the N stage (P<0. 05) and the degree of differentiation (P<0. 01). Targetscan Human7. 2 bioinformatics software predicted that the MTDH gene contained a target sequence of hsa-miR-26a.The luciferase reporter gene experiment also verified the targeted regulation relationship between miR-26a and MTDH. The expression of miR-26a was the highest in KYSE-450 cells and the lowest in Ecal09 cells. The mRNA expression of MTDH in the lv-miR-26a group was significantly lower than that in the lv-NC group, and the mRNA expression in the lv-miR-26a-inhibitor group was significantly higher than that in the lv-NC group. Alter tumor formation, miR-26a expression increased and MTDH expression decreased in miR-26a group. Alter tumor formation, the expression oi miR-26a decreased and the expression oi MTDH increased in miR-26a inhibitor group. Conclusion MiR-26a can inhibit the expression oi MTDH in esophageal cancer cells. Both in vitro and in vivo experiments can verily the targeted regulatory relationship between miR-26a and MTDH. MiR-26a ma)' play a role in the occurrence and development oi esophageal cancer through the MTDH.

16.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1014871

ABSTRACT

AIM: To optimize an orthopedic non-muscle invasive bladder cancer (NMIBC) model in nude mouse by comparing four different ways of cellular transplantation, and to evaluate the efficacy of drug by bladder instillation, so as to provide a stable and efficient animal model for the treatment of bladder cancer. METHODS: After disruption of bladder mucosa by dilute acid-alkali or silver nitrate, T24 cells were instilled into the nude mouse bladder. T24 cells were injected directly into the bladder with mechanical injury of bladder mucosa. T24 cells were injected into the bladder wall. On the 14th day after making models, the nude mice were sacrificed. And the bladder mass and histopathological changes of tumor (including bladder) was observe to confirm the formation of orthopedic bladder cancer. The dynamic changes of orthopedic bladder cancer were observed after injecting T24 cells into the bladder wall. Gemcitabine was used to verify the applicability of the model of injecting T24 cells into the bladder wall in vivo. RESULTS: No tumor was found in the bladder after intravesical instillation of T24 cells with dilute acid-alkali or silver nitrate treatment. With mechanical injury of bladder mucosa, all nude mice had tumors after injection T24 cells. But the number of tumors varied and often occurred at multiple sites. The tumor was found in the bladder of all nude mice by injecting T24 cells into bladder wall, and there was only one tumor. The tumor showed slow linear growth within 15 days and rapid linear growth from day 18 to 31. In vivo efficacy evaluation, gemcitabine 150 mg/kg intravesical perfusion could significantly inhibit the growth of NMIBC in nude mice replicated by direct injection of T24 cells into the bladder wall, and the tumor inhibition rate was 97.1%. CONCLUSION: The orthotopic NMIBC model can not be established with the bladder mucosa injuried by dilute acid-alkali or silver nitrate treatment. The number and size of orthotopic bladder cancer are different by mechanical injury of bladder mucosa. Injection of T24 cells into the bladder wall of nude mouse can successfully establish the orthotopic NMIBC model, which can be used for the evaluation of NMIBC therapeutic drugs.

17.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-932653

ABSTRACT

Objective:To evaluate the effect of compound UC2288 on the radiosensitivity of CNE-2R cell line and nude mouse transplanted tumor.Methods:The UC2288 concentration was referenced to previous experimental results (IC 50=12.20 μmol/L). The effect of UC2288 combined with 2, 4, 6, 8 Gy X-ray irradiation on the radiosensitivity of CNE-2R cell line was detected by clone formation experiment. The effect of UC2288 combined with 2, 4, 6, 8 Gy X-ray irradiation on the proliferation of CNE-2R cell line was determined by CCK8 assay. The nude mouse model of transplanted tumor was constructed with CNE-2R cell line. The radiosensitivity of transplanted tumor of UC2288 combined with 2 Gy/fraction X-ray irradiation for three consecutive days was evaluated. Results:The experimental concentration of UC2288 was 8 μmol/L. The clonality of CNE-2R cell line was reduced under UC2288 combined with X-ray 2, 4, 6, and 8 Gy irradiation, andthe radiosensitizationratio was 1.60. The proliferation of CNE-2R cell line was significantly decreased under UC2288 combined with X-ray 2, 4, 6, and 8 Gy irradiation. UC2288 inhibited the growth of transplanted tumor in nude mice, and the inhibitory effect was strengthened with the extension of observation time, and the most obvious effect was observed at 16 d. ( P<0.01). Theradiosensitizationratio was 4.33. The proliferation of CNE-2R cell line was decreased under UC2288 combined with X-ray irradiation. Conclusion:UC2288 can increase the radiosensitivity of nasopharyngeal carcinoma radioresistant cell line CNE-2R.

18.
Front Genet ; 12: 765131, 2021.
Article in English | MEDLINE | ID: mdl-34912374

ABSTRACT

Here, we report a model system using in vitro 7,12-dimethylbenz[a]anthracene (DMBA; 0.6 µM)-treated mammary tissue-derived organoids generated from heterozygous BALB/c-Trp53 knockout mice to induce tumors after injection into the nude mouse subcutis. In parallel, a single oral dose of DMBA (50 mg/kg bodyweight) to the same murine strain induced mammary adenocarcinomas, characterized by biphasic structures differentiated into luminal and myoepithelial lineages and frequent Hras mutations at codon 61. In the present study, the genetic and histological characteristics of DMBA-induced tumors in the organoid-based model were evaluated to validate its similarities to the in vivo study. The organoid-derived tumors were low-grade adenocarcinomas composed of luminal and basal/myoepithelial cells. When the organoid-derived carcinomas were passaged to other nude mice, they partly progressed to squamous cell carcinomas (SCCs). Whole exome sequencing revealed no mutations at Hras codon 61 in the organoid-derived tumors. However, various mutations were detected in other genes such as Tusc3 and Tgfbr2, which have been reported as cancer-associated or homeostatic squamous cell genes. The most common mutational pattern observed in these genes were the G:C to T:A transversions and G:C to A:T transitions, which are not typical of the mutations caused by DMBA treatment. In conclusion, DMBA exhibited carcinogenicity in the both the ex vivo and in vivo mammary carcinogenesis models, albeit with distinct histological and genetical alterations. Further studies are needed to clarify whether organoid-based carcinogenesis models generated following chemical treatment in vitro could be applied to the clarification of the novel mode of action of chemical carcinogenesis.

19.
Bull Exp Biol Med ; 172(1): 26-32, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34792718

ABSTRACT

The anti-metastasis effect of oridonin in combination with oxaliplatin on colorectal cancer liver metastasis was studied using a BALB/c nude mouse model. The liver condition, bloody ascites, cholestasis, and liver metastasis scores in the three groups receiving oxaliplatin combined with oridonin were significantly milder than in the control group and importantly the anti-migratory effect of oxaliplatin combined with oridonin was obviously the strongest (p<0.05). Oridonin possessed no hepatotoxicity; instead, it effectively alleviated liver injury caused by oxaliplatin. Oridonin alone or in combination with oxaliplatin significantly decreased serum levels of α-fetoprotein and carcinoembryonic antigen. Therefore, oridonin combined with oxaliplatin displays great potential to markedly increase the anti-metastasis effect of oxaliplatin in the treatment of liver metastases of colorectal cancer.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Diterpenes, Kaurane/pharmacology , Liver Neoplasms/prevention & control , Liver Neoplasms/secondary , Oxaliplatin/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Ascites/prevention & control , Carcinoembryonic Antigen/blood , Chemical and Drug Induced Liver Injury/drug therapy , Cholestasis/prevention & control , Drug Synergism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis/prevention & control , Oxaliplatin/adverse effects , alpha-Fetoproteins/analysis
20.
In Vivo ; 35(6): 3107-3110, 2021.
Article in English | MEDLINE | ID: mdl-34697141

ABSTRACT

BACKGROUND/AIM: Primary osteosarcoma of the mammary gland is a very rare disease, accounting for less than 1% of all mammary malignancies. There is no established first-line treatment and the prognosis is poor compared to normal breast cancer. We previously established the first patient tumor-derived animal model of this disease, grown subcutaneously in nude mice. In the present study, we established a patient derived orthotopic xenograft (PDOX) model of osteosarcoma of the breast and investigated the efficacy of cisplatinum (CDDP) and eribulin (ERB). MATERIALS AND METHODS: PDOX models of primary osteosarcoma of the breast were divided into 3 groups (5-6 mice per group): untreated control; CDDP treatment; ERB treatment. The tumor volume in the 3 groups was compared after 2 weeks. RESULTS: There were significant differences between control and CDDP, and control and ERB (p=0.036, 0.046, respectively). However, there was no significant difference between CDDP and ERB (p=0.964). CONCLUSION: CDDP and ERB are candidates for first-line clinical therapy of primary osteosarcoma of the breast.


Subject(s)
Bone Neoplasms , Osteosarcoma , Animals , Bone Neoplasms/drug therapy , Disease Models, Animal , Drug Resistance, Neoplasm , Furans , Heterografts , Humans , Ketones , Mice , Mice, Nude , Osteosarcoma/drug therapy , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...