Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Eur J Nucl Med Mol Imaging ; 50(11): 3452-3464, 2023 09.
Article in English | MEDLINE | ID: mdl-37278941

ABSTRACT

PURPOSE: Prostate-specific membrane antigen (PSMA)-positron emission tomography (PET) is a superior method to predict patients' risk of cancer progression and response to specific therapies. However, its performance is limited for neuroendocrine prostate cancer (NEPC) and PSMA-low prostate cancer cells, resulting in diagnostic blind spots. Hence, identifying novel specific targets is our aim for diagnosing those prostate cancers with low PSMA expression. METHODS: The Cancer Genome Atlas (TCGA) database and our cohorts from men with biopsy-proven high-risk metastatic prostate cancer were used to identify CDK19 and PSMA expression. PDX lines neP-09 and P-16 primary cells were used for cellular uptake and imaging mass cytometry in vitro. To evaluate in vivo CDK19-specific uptake of gallium(Ga)-68-IRM-015-DOTA, xenograft mice models and blocking assays were used. PET/CT imaging data were obtained to estimate the absorbed dose in organs. RESULTS: Our study group had reported the overexpression of a novel tissue-specific gene CDK19 in high-risk metastatic prostate cancer and CDK19 expression correlated with metastatic status and tumor staging, independently with PSMA and PSA levels. Following up on this new candidate for use in diagnostics, small molecules targeting CDK19 labeled with Ga-68 (68Ga-IRM-015-DOTA) were used for PET in this study. We found that the 68Ga-IRM-015-DOTA was specificity for prostate cancer cells, but the other cancer cells also took up little 68Ga-IRM-015-DOTA. Importantly, mouse imaging data showed that the NEPC and CRPC xenografts exhibited similar signal strength with 68Ga-IRM-015-DOTA, but 68Ga-PSMA-11 only stained the CRPC xenografts. Furthermore, target specificity was elucidated by a blocking experiment on a CDK19-bearing tumor xenograft. These data concluded that 68Ga-CDK19 PET/CT was an effective technology to detect lesions with or without PSMA in vitro, in vivo, and in the PDX model. CONCLUSION: Thus, we have generated a novel PET small molecule with predictive value for prostate cancer. The findings indicate that 68Ga-CDK19 may merit further evaluation as a predictive biomarker for PET scans in prospective cohorts and may facilitate the identification of molecular types of prostate cancer independent of PSMA.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Prostatic Neoplasms , Humans , Male , Animals , Mice , Gallium Radioisotopes , Positron Emission Tomography Computed Tomography/methods , Prospective Studies , Prostatic Neoplasms/pathology , Positron-Emission Tomography , Cyclin-Dependent Kinases
2.
J Cell Mol Med ; 26(5): 1621-1628, 2022 03.
Article in English | MEDLINE | ID: mdl-35088536

ABSTRACT

The Hippo signalling pathway has been considered as potential therapeutic target in self-renewal and differentiation of stem and progenitor cells. Thus, mammalian Ste20-like kinase 1/2 (MST1/2) as the core serine-threonine kinases in the Hippo signalling pathway has been investigated for its role in immunological disease. However, little information of MST1/2 function in bone marrow suppression induced by ionizing radiation was fully investigated. Here, we reported that MST1/2 inhibitor XMU-MP-1 could rescue the impaired haematopoietic stem cells (HSCs) and progenitor cells (HPCs) function under oxidative stress condition. Also, XMU-MP-1 pretreatment markedly alleviated the small intestinal system injury caused by the total body irradiation 9 Gy and extended the average survival days of the mice exposed to the lethal dose radiation. Therefore, irradiation exposure causes the serious pathological changes of haematopoietic and intestinal system, and XMU-MP-1 could prevent the ROS production, the haematopoietic cells impairment and the intestinal injury. These detrimental effects may be associated with regulating NOX/ROS/P38MARK pathway by MST1/2.


Subject(s)
Hematopoietic Stem Cells , Sulfonamides , Animals , Hematopoietic Stem Cells/metabolism , Mammals , Mice , Radiation, Ionizing , Reactive Oxygen Species/metabolism , Sulfonamides/pharmacology
3.
J Cell Mol Med ; 26(11): 3269-3280, 2022 06.
Article in English | MEDLINE | ID: mdl-35510614

ABSTRACT

Pulmonary fibrosis (PF) is a progressive interstitial lung disease with limited treatment options. The incidence and prevalence of PF is increasing with age, cell senescence has been proposed as a pathogenic driver, the clearance of senescent cells could improve lung function in PF. FOXO4-D-Retro-Inverso (FOXO4-DRI), a synthesis peptide, has been reported to selectively kill senescent cells in aged mice. However, it remains unknown if FOXO4-DRI could clear senescent cells in PF and reverse this disease. In this study, we explored the effect of FOXO4-DRI on bleomycin (BLM)-induced PF mouse model. We found that similar as the approved medication Pirfenidone, FOXO4-DRI decreased senescent cells, downregulated the expression of senescence-associated secretory phenotype (SASP) and attenuated BLM-induced morphological changes and collagen deposition. Furthermore, FOXO4-DRI could increase the percentage of type 2 alveolar epithelial cells (AEC2) and fibroblasts, and decrease the myofibroblasts in bleomycin (BLM)-induced PF mouse model. Compared with mouse and human lung fibroblast cell lines, FOXO4-DRI is inclined to kill TGF-ß-induced myofibroblast in vitro. The inhibited effect of FOXO4-DRI on myofibroblast lead to a downregulation of extracellular matrix (ECM) receptor interaction pathway in BLM-induced PF. Above all, FOXO4-DRI ameliorates BLM-induced PF in mouse and may be served as a viable therapeutic option for PF.


Subject(s)
Pulmonary Fibrosis , Animals , Bleomycin/adverse effects , Cell Cycle Proteins/metabolism , Disease Models, Animal , Fibroblasts/metabolism , Forkhead Transcription Factors/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Myofibroblasts/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism
4.
World J Surg Oncol ; 20(1): 167, 2022 May 27.
Article in English | MEDLINE | ID: mdl-35624511

ABSTRACT

OBJECTIVE: The present study comparatively analyzed short-term clinical effectiveness and long-term follow-up endpoints associated with robotic-assisted sphincter-preserving surgery (RAS) and laparoscopic-assisted sphincter-preserving surgery (LAS) when used to treat low rectal cancer. METHOD: Within such a single-center retrospective cohort analysis, low rectal cancer patients that underwent RAS (n=200) or LAS (n=486) between January 2015 and beginning of July 2018 were enrolled. RESULTS: The mean operative durations in the RAS and LAS cohorts were 249±64 min and 203±47 min, respectively (P<0.001). Temporary ileostomy rates in the RAS and LAS cohorts were 64.5% and 51.6% (P = 0.002). In addition, major variations across such cohorts regarding catheter removal timing, time to liquid intake, time to first leaving bed, and length of hospitalization (all P<0.001). This distal resection margin distance within the RAS cohort was diminished in comparison to LAS cohort (P=0.004). For patients within the LAS cohort, the time required to recover from reduced urinary/female sexual function was > 6 months post-surgery (P<0.0001), whereas within the RAS cohort this interval was 3 months (P<0.0001). At 6 months post-surgery, male sexual function within RAS cohort was improved in comparison to LAS cohort (P<0.001). At 6 months post-surgery, Wexner scores revealed similar results (P<0.001). No major variations within overall or disease-free survival were identified across these cohorts at 3 or 5 years post-surgery. CONCLUSION: Robotic sphincter-preserving surgery is a safe and effective surgical technique in low rectal patients in terms of postoperative oncological safety and long-term endpoints. And the RAS strategy provides certain additional benefits with respect to short-term urogenital/anorectal functional recovery in treated patients compared to LAS.


Subject(s)
Laparoscopy , Rectal Neoplasms , Robotic Surgical Procedures , Cohort Studies , Female , Follow-Up Studies , Humans , Laparoscopy/adverse effects , Laparoscopy/methods , Male , Rectal Neoplasms/surgery , Retrospective Studies , Robotic Surgical Procedures/adverse effects , Robotic Surgical Procedures/methods
5.
J Cell Mol Med ; 25(21): 10306-10312, 2021 11.
Article in English | MEDLINE | ID: mdl-34595829

ABSTRACT

Ionizing radiation (IR)-induced intestinal damage is the major and common injury of patients receiving radiotherapy. Urolithin A (UroA) is a metabolite of the intestinal flora of ellagitannin, a compound found in fruits and nuts such as pomegranates, strawberries and walnuts. UroA shows the immunomodulatory and anti-inflammatory capacity in various metabolic diseases. To evaluate the radioprotective effects, UroA(0.4, 2 and 10 mg/kg) were intraperitoneally injected to C57BL/6 male mice 48, 24, 1 h prior to and 24 h after 9.0Gy TBI. The results showed that UroA markedly upregulated the survival of irradiated mice, especially at concentration of 2 mg/kg. UroA improved the intestine morphology architecture and the regeneration ability of enterocytes in irradiated mice. Then, UroA significantly decreased the apoptosis of enterocytes induced by radiation. Additionally, 16S rRNA sequencing analysis showed the effect of UroA is associated with the recovery of the IR-induced intestinal microbacteria profile changes in mice. Therefore, our results determinated UroA could be developed as a potential candidate for radiomitigators in radiotherapy and accidental nuclear exposure. And the beneficial functions of UroA might be associated with the inhibition of p53-mediated apoptosis and remodelling of the gut microbes.


Subject(s)
Coumarins/pharmacology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/radiation effects , Radiation, Ionizing , Radiation-Protective Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Coumarins/metabolism , DNA Damage/drug effects , DNA Damage/radiation effects , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/radiation effects , Gastrointestinal Tract/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/radiation effects , Mice , Radiation Dosage
6.
Int J Colorectal Dis ; 35(4): 775-778, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32020267

ABSTRACT

INTRODUCTION: Perineal hernia is a protrusion of the pelvic floor which contains intra-abdominal viscera. The occurrence of perineal hernia after abdominoperineal resection (APR) is rare, but it has been reported in recent years that the incidence of perineal hernia after rectal cancer surgery has increased. This has been attributed to a shift towards extralevator abdominoperineal resection, together with more frequent and long-term use of neoadjuvant therapy. PRESENTATION OF CASE: Here, we report a patient with perineal hernia 5 years after APR surgery for rectal cancer. We decided to perform robot-assisted laparoscopic surgery on this patient using the da Vinci Surgical System. The perineal hernia was repaired by primary closure with the placement of a non-absorbable synthetic mesh as reinforcement for the pelvic floor. No complications occurred during the operation, and the patient was discharged on the third day after the operation. Clinical follow-up proceeded at the designated time intervals without difficulties. DISCUSSION: The recurrence rates of perineal hernia are still very high, and due to poor view, suturing, and mesh placement in the deep pelvis, surgeons face many challenges. Many methods have been described, but there is still no consensus as to the optimal repair technique for perineal hernia. CONCLUSION: Perineal hernias can be repaired with robot-assisted laparoscopy. Furthermore, compared with the open and laparoscopic methods, suturing and mesh placement are easier with the robot approach.


Subject(s)
Hernia, Abdominal/surgery , Laparoscopy , Perineum/surgery , Proctectomy/adverse effects , Robotic Surgical Procedures , Female , Follow-Up Studies , Hernia, Abdominal/diagnostic imaging , Humans , Magnetic Resonance Imaging , Perineum/diagnostic imaging , Surgical Mesh
7.
J Cell Mol Med ; 23(3): 2238-2247, 2019 03.
Article in English | MEDLINE | ID: mdl-30663222

ABSTRACT

Radiation-induced intestinal injury is one of the major side effects in patients receiving radiation therapy. There is no specific treatment for radiation enteritis in the clinic. We designed and synthesized a new compound named XH-105, which is expected to cleave into polyphenol and aminothiol in vivo to mitigate radiation injury. In the following study, we describe the beneficial effects of XH-105 against radiation-induced intestinal injury. C57BL/6J mice were treated by gavage with XH-105 1 hour before total body irradiation (TBI), and the survival rate was monitored. Histological changes were examined, and survival of Lgr5+ intestinal stem cells Ki67+ cells, villi+ enterocytes and lysozymes was determined by immunohistochemistry. DNA damage and cellular apoptosis in intestinal tissue were also evaluated. Compared to vehicle-treated mice after TBI, XH-105 treatment significantly enhanced the survival rate, attenuated structural damage of the small intestine, decreased the apoptotic rate, reduced DNA damage, maintained cell regeneration and promoted crypt proliferation and differentiation. XH-105 also reduced the expression of Bax and p53 in the small intestine. These data suggest that XH-105 is beneficial for the protection of radiation-induced intestinal injury by inhibiting the p53-dependent apoptosis signalling pathway.


Subject(s)
Benzopyrans/pharmacology , Enterocytes/drug effects , Intestine, Small/drug effects , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Benzopyrans/chemical synthesis , Benzopyrans/chemistry , Cell Line , DNA Damage/drug effects , DNA Damage/radiation effects , Enterocytes/metabolism , Intestine, Small/metabolism , Intestine, Small/radiation effects , Kaplan-Meier Estimate , Mice, Inbred C57BL , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/metabolism , Radiation-Protective Agents/chemical synthesis , Radiation-Protective Agents/chemistry , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/radiation effects , Tumor Suppressor Protein p53/metabolism , Whole-Body Irradiation/adverse effects , bcl-2-Associated X Protein/metabolism
8.
World J Surg Oncol ; 16(1): 238, 2018 Dec 21.
Article in English | MEDLINE | ID: mdl-30577805

ABSTRACT

OBJECTIVE: To evaluate the intraoperative and short-term postoperative outcomes of a novel robotic intracorporeal π-shaped esophagojejunostomy (EJS) after D2 total gastrectomy (TG) using the Da Vinci robotic surgical system for intracorporeal anastomosis after TG. BACKGROUND: Intracorporeal π-shaped EJS, using a linear stapler, was recently reported for laparoscopic total gastrectomy in patients with gastric cancer. However, robotic intracorporeal π-shaped EJS using a linear stapler has not been reported. This report aimed to describe the use of a novel technique for π-shaped EJS using the Da Vinci robotic system. METHODS: Robotic intracorporeal π-shaped esophagojejunostomy after total gastrectomy was performed in 11 consecutive patients diagnosed with early gastric cancer, and their perioperative outcomes were analyzed. RESULTS: All the operations were successful without conversion to open or laparoscopic surgery and postoperative complications. The total number of patients was 11 (7 males and 4 females). The mean age of the patients was 63.36 ± 10.56 years old. Seven patients were diagnosed with cardia cancer, 3 patients were diagnosed with gastric body cancer, and 1 patient was diagnosed with gastric antrum cancer. The patients' mean proximal resection margin was 3.18 ± 1.17 cm, the distal resection margin was 6.18 ± 1.40 cm, the mean length of the incision was 4.55 ± 0.69 cm, the mean operative time was 287.27 ± 30.69 min, the mean day of first flatus was 3.27 ± 0.79 days, the mean day of the start of diet was 2.91 ± 0.94 days, the mean postoperative hospital stay was 11.45 ± 5.13 days, and the mean operative blood loss was 47.27 ± 31.33 ml. No complications were observed during anastomosis, and the median anastomosis time was 19.5 min. The mean number of lymph node dissections was 17.91 ± 4.59, the mean number of positive lymph nodes was 0.45 ± 0.69, all patients were diagnosed with stage I-II gastric cancer, and the mean maximum diameter of the tumor was 2.67 ± 1.30 cm. All the patients had a smooth hospital discharge. CONCLUSION: A novel robotic gastrectomy with intracorporeal π-shaped EJS for esophagojejunal anastomosis described and shows acceptable resulted. This technique has the potential to offer better short-term surgical outcomes and overcomes the drawbacks of laparoscopy with a decreased risk of complications during and after surgery.


Subject(s)
Adenocarcinoma/surgery , Esophagus/surgery , Gastrectomy/methods , Jejunum/surgery , Robotic Surgical Procedures/methods , Stomach Neoplasms/surgery , Surgical Stapling/instrumentation , Adenocarcinoma/secondary , Female , Follow-Up Studies , Humans , Laparoscopy , Lymphatic Metastasis , Male , Middle Aged , Prognosis , Stomach Neoplasms/pathology , Surgical Stapling/methods
9.
Ann Hematol ; 96(7): 1085-1095, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28421266

ABSTRACT

The adverse effects of iron overload have raised more concerns as a growing number of studies reported its association with immune disorders. This study aimed to investigate alterations in the immune system by iron overload in patients with myelodysplastic syndrome (MDS) and an iron-overloaded mouse model. The peripheral blood from patients was harvested to test the effect of iron overload on the subsets of T lymphocytes, and the level of reactive oxygen species (ROS) was also evaluated. The data showed that iron-overloaded patients had a lower percentage of CD3+ T cells and disrupted T cell subsets, concomitant with higher ROS level in lymphocytes. In order to explore the mechanism, male C57Bl/6 mice were intraperitoneally injected with iron dextran at a dose of 250 mg/kg every 3 days for 4 weeks to establish an iron-overloaded mouse model and the blood of each mouse was collected for the analysis of the T lymphocyte subsets and T cell apoptosis. The results showed that iron overload could reduce the percentage of CD3+ T cells and the ratio of Th1/Th2 and Tc1/Tc2 but increase the percentage of regulatory T (Treg) cells and the ratio of CD4/CD8. We also found that iron overload induced the apoptosis of T lymphocytes and increased its ROS level. Furthermore, these effects could be partially recovered after treating with antioxidant N-acetyl-L-cysteine (NAC) or iron chelator deferasirox (DFX). Taken together, these observations indicated that iron overload could selectively affect peripheral T lymphocytes and induce an impaired cellular immunity by increasing ROS level.


Subject(s)
Iron Overload/metabolism , Myelodysplastic Syndromes/metabolism , Reactive Oxygen Species/metabolism , T-Lymphocyte Subsets/metabolism , Aged , Aged, 80 and over , Animals , CD3 Complex/blood , CD4-CD8 Ratio , Disease Models, Animal , Female , Flow Cytometry , Humans , Iron Overload/blood , Lymphocyte Count , Male , Mice, Inbred C57BL , Middle Aged , Myelodysplastic Syndromes/blood , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism
10.
Int J Mol Sci ; 17(6)2016 Jun 14.
Article in English | MEDLINE | ID: mdl-27314327

ABSTRACT

Antioxidants are prospective radioprotectors because of their ability to scavenge radiation-induced reactive oxygen species (ROS). The hematopoietic system is widely studied in radiation research because of its high radiosensitivity. In the present study, we describe the beneficial effects of 5-methoxytryptamine-α-lipoic acid (MLA), which was synthesized from melatonin and α-lipoic acid, against radiation-induced hematopoietic injury. MLA administration significantly enhanced the survival rate of mice after 7.2 Gy total body irradiation. The results showed that MLA not only markedly increased the numbers and clonogenic potential of hematopoietic cells but also decreased DNA damage, as determined by flow cytometric analysis of histone H2AX phosphorylation. In addition, MLA decreased the levels of ROS in hematopoietic cells by inhibiting NOX4 expression. These data demonstrate that MLA prevents radiation-induced hematopoietic syndrome by increasing the number and function of and by inhibiting DNA damage and ROS production in hematopoietic cells. These data suggest MLA is beneficial for the protection of radiation injuries.


Subject(s)
5-Methoxytryptamine/therapeutic use , Acute Radiation Syndrome/drug therapy , Hematopoiesis/drug effects , Radiation-Protective Agents/therapeutic use , Thioctic Acid/analysis , Thioctic Acid/therapeutic use , 5-Methoxytryptamine/chemical synthesis , 5-Methoxytryptamine/chemistry , 5-Methoxytryptamine/pharmacology , Acute Radiation Syndrome/metabolism , Acute Radiation Syndrome/prevention & control , Animals , DNA Damage/drug effects , Histones/metabolism , Male , Melatonin/chemistry , Mice , Mice, Inbred C57BL , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Radiation, Ionizing , Radiation-Protective Agents/administration & dosage , Radiation-Protective Agents/chemical synthesis , Radiation-Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Thioctic Acid/chemical synthesis , Thioctic Acid/chemistry , Thioctic Acid/pharmacology
11.
Int J Mol Sci ; 17(6)2016 Jun 08.
Article in English | MEDLINE | ID: mdl-27338355

ABSTRACT

Senescent hematopoietic stem cells (HSCs) accumulate with age and exposure to stress, such as total-body irradiation (TBI), which may cause long-term myelosuppression in the clinic. However, the methods available for long-term myelosuppression remain limited. Previous studies have demonstrated that sustained p38 mitogen-activated protein kinases (p38 MAPK) activation in HSCs following exposure to TBI in mice and the administration of its inhibitor twenty-four hours after TBI may partially prevent long-term myelosuppression. However, long-term myelosuppression is latent and identified long after the administration of radiation. In this study, we investigated the effects of SB203580 (a small molecule inhibitor of p38 MAPK) on long-term myelosuppression induced by TBI. Mice with hematopoietic injury were injected intraperitoneally with SB203580 every other day five times beginning 70 days after 6 Gy of (137)Cs γ ray TBI. Our results at 80 days demonstrated that SB203580 did not significantly improve the TBI-induced long-term reduction of peripheral blood cell and bone marrow nucleated cell (BMNC) counts, or defects in hematopoietic progenitor cells (HPCs) and HSC clonogenic function. SB203580 reduced reactive oxygen species (ROS) production and p-p38 expression; however, SB203580 had no effect on p16 expression in the HSCs of mice. In conclusion, these findings suggest that treatment with SB203580 70 days after TBI in mice inhibits the ROS-p38 oxidative stress pathway; however, it has no therapeutic effect on long-term myelosuppression induced by TBI.


Subject(s)
Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , Whole-Body Irradiation , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Blood Cells/drug effects , Blood Cells/metabolism , Blood Cells/radiation effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Colony-Forming Units Assay , Imidazoles/pharmacology , Male , Mice , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Pyridines/pharmacology , Reactive Oxygen Species/metabolism
12.
Molecules ; 21(12)2016 Dec 14.
Article in English | MEDLINE | ID: mdl-27983649

ABSTRACT

Sixteen novel epidermal growth factor receptor (EGFR)/vascular endothelial growth factor (VEGF)-2 inhibitors (nitroimidazole-substituted 4-anilinoquinazoline derivatives (16a-p)) were designed and prepared via the introduction of a nitroimidazole group in the piperidine side chain and modification on the aniline moiety of vandetanib. Preliminary biological tests showed that comparing with vandetanib, some target compounds exhibited excellent EGFR inhibitory activities and anti-proliferative over A549/H446 cells in hypoxia. Meanwhile, several of the above compounds demonstrated better bioactivity than vandetanib in VEGF gene expression inhibition. Owing to the excellent IC50 value (1.64 µmol/L), the inhibition ratios of 16f over A549 and H446 cells were 62.01% and 59.86% at the concentration of 0.5 µM in hypoxia, respectively. All of these results indicated that 16f was a potential cancer therapeutic agent in hypoxia and was worthy of further development.


Subject(s)
Cell Hypoxia , Nitroimidazoles/chemistry , Oxygen/metabolism , Piperidines/chemistry , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/chemistry , Cell Line, Tumor , ErbB Receptors/genetics , Gene Expression , Humans , Piperidines/chemical synthesis , Protein Kinase Inhibitors/chemistry , Quinazolines/chemical synthesis , Vascular Endothelial Growth Factor A/genetics
13.
Int J Mol Sci ; 15(6): 10541-53, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24927144

ABSTRACT

Hematopoietic injury is the most common side effect of radiotherapy. However, the methods available for the mitigating of radiation injury remain limited. Xuebijing injection (XBJ) is a traditional Chinese medicine used to treat sepsis in the clinic. In this study, we investigated the effects of XBJ on the survival rate in mice with hematopoietic injury induced by γ ray ionizing radiation (IR). Mice were intraperitoneally injected with XBJ daily for seven days after total body irradiation (TBI). Our results showed that XBJ (0.4 mL/kg) significantly increased 30-day survival rates in mice exposed to 7.5 Gy TBI. This effect may be attributable to improved preservation of white blood cells (WBCs) and hematopoietic cells, given that bone marrow (BM) cells from XBJ-treated mice produced more granulocyte-macrophage colony forming units (CFU-GM) than that in the 2 Gy/TBI group. XBJ also decreased the levels of reactive oxygen species (ROS) by increasing glutathione (GSH) and superoxide dismutase (SOD) levels in serum and attenuated the increased BM cell apoptosis caused by 2 Gy/TBI. In conclusion, these findings suggest that XBJ enhances the survival rate of irradiated mice and attenuates the effects of radiation on hematopoietic injury by decreasing ROS production in BM cells, indicating that XBJ may be a promising therapeutic candidate for reducing hematopoietic radiation injury.


Subject(s)
Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , Gamma Rays , Hematopoietic Stem Cells/drug effects , Animals , Apoptosis/radiation effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/radiation effects , Colony-Forming Units Assay , Glutathione/blood , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Male , Medicine, Chinese Traditional , Mice , Mice, Inbred ICR , Reactive Oxygen Species/metabolism , Superoxide Dismutase/blood , Whole-Body Irradiation
14.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 36(5): 538-41, 2014 Oct.
Article in Zh | MEDLINE | ID: mdl-25360654

ABSTRACT

Dipeptidyl peptidase-4 (DPP-4) is a protease that cleaves the peptides with alanine, praline, or other selective amino acids at the N-terminal penultimate position. The substrates of DPP-4 include many chemokines, colony-stimulating factors, and interleukins. Recent research has shown that DPP-4 can affect the hematopoietic stem and progenitor cells and transplantation by truncating the granulocyte colony stimulating factor. However, its regulatory effect on DPP-4 and most peptides truncation are still unknown. This review summarizes the recent advances in the DPP-4 research.


Subject(s)
Dipeptidyl Peptidase 4/physiology , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Humans
15.
Life Sci ; 341: 122486, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38331314

ABSTRACT

PURPOSE: Damage to the hematopoietic system and functional inhibition are severe consequences of radiation exposure. In this study, we have investigated the effect of empagliflozin on radiation-induced hematopoietic damage, with the aim of providing new preventive approach to such injuries. METHODS AND MATERIALS: Mice were given 4 Gy total body irradiation (TBI) 1 h after the oral administration of empagliflozin, followed by the continuous administration of the same dose of empagliflozin for 6d, and then sacrificed on the 10th day after irradiation. The reactive oxygen species (ROS) levels in hematopoietic cells and their regulatory mechanisms were also been investigated. Colony forming unit granulocyte macrophage assay and bone marrow transplantation assays were performed to detect the function of the bone marrow cells. KEY FINDINGS: Empagliflozin increased the cell viability, reduced ROS levels, and attenuated apoptosis in vitro after the bone marrow cells were exposed to 1 Gy radiation. Empagliflozin significantly attenuated ionizing radiation injuries to the hematopoietic system, increased the peripheral blood cell count, and enhanced the proportion and function of hematopoietic stem cells in mice exposed to 4 Gy TBI. These effects may be related to the NOX-4/ROS/p38 pathway-mediated suppression of MAPK in hematopoietic stem cells. Empagliflozin also influenced the expression of Nrf-2 and increased glutathione peroxidase activity, thereby promoting the clearance of reactive oxygen species. Furthermore, empagliflozin mitigated metabolic abnormalities by inhibiting the mammalian target of rapamycin. SIGNIFICANCE: Our study has demonstrated that empagliflozin can reduce radiation-induced injury in hematopoietic stem cells. This finding suggests that empagliflozin is a promising novel agent for preventing radiation-induced damage to the hematopoietic system.


Subject(s)
Glucosides , Hematopoietic Stem Cells , Radiation Injuries , Animals , Mice , Reactive Oxygen Species/metabolism , Colony-Forming Units Assay , Benzhydryl Compounds/pharmacology , Benzhydryl Compounds/metabolism , Radiation Injuries/metabolism , Whole-Body Irradiation , Mice, Inbred C57BL , Mammals/metabolism
16.
Int Immunopharmacol ; 130: 111695, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38401461

ABSTRACT

Ionizing radiation (IR)-induced damage to the hematopoietic system is a prominent symptom following exposure to total body irradiation (TBI). The exploration of strategies aimed at to mitigating radiation-induced hematopoietic damage assumes paramount importance. Time-restricted feeding (TRF) has garnered attention for its beneficial effects in various diseases. In this study, we evaluated the preventive effects of TRF on TBI-induced hematopoietic damage. The results suggested that TRF significantly enhanced the proportion and function of hematopoietic stem cells in mice exposed to 4 Gy TBI. These effects might be attributed to the inhibition of the NOX-4/ROS/p38 MAPK pathway in hematopoietic stem cells. TRF also influenced the expression of nuclear factor erythroid2-related factor 2 and increased glutathione peroxidase activity, thereby promoting the clearance of reactive oxygen species. Furthermore, TRF alleviated aberrations in plasma metabolism by inhibiting the mammalian target of rapamycin. These findings suggest that TRF may represent a novel approach to preventing hematopoietic radiation damage.


Subject(s)
Hematopoietic Stem Cells , p38 Mitogen-Activated Protein Kinases , Animals , Mice , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Radiation, Ionizing , Whole-Body Irradiation , Mice, Inbred C57BL , Mammals
17.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 35(5): 547-52, 2013 Oct.
Article in Zh | MEDLINE | ID: mdl-24183045

ABSTRACT

OBJECTIVE: To establish a mouse model of iron overload by intraperitoneal injection of iron dextran and investigate the impact of iron overload on bone marrow hematopoiesis. METHODS: A total of 40 C57BL/6 mice were divided into control group, low-dose iron group (12.5 mg/ml), middle-dose iron group (25 mg/ml), and high-dose iron group (50 mg/ml). The control group received normal saline (0.2 ml), and the rest were injected with intraperitoneal iron dextran every three days for six weeks. Iron overload was confirmed by observing the bone marrow, hepatic, and splenic iron deposits and the bone marrow labile iron pool. In addition, peripheral blood and bone marrow mononuclear cells were counted and the hematopoietic function was assessed. RESULTS: Iron deposits in bone marrow, liver, and spleen were markedly increased in the mouse models. Bone marrow iron was deposited mostly within the matrix with no significant difference in expression of labile iron pool.Compared with control group, the ability of hematopoietic colony-forming in three interventional groups were decreased significantly (P<0.05). Bone marrow mononuclear cells counts showed no significant difference. The amounts of peripheral blood cells (white blood cells, red blood cells, platelets, and hemoglobin) in different iron groups showed no significant difference among these groups;although the platelets were decreased slightly in low-dose iron group [(780.7±39.60)×10(9)/L], middle dose iron group [(676.2±21.43)×10(9)/L], and high-dose iron group [(587.3±19.67)×10(9)/L] when compared with the control group [(926.0±28.23)×10(9)/L], there was no significant difference(P>0.05). CONCLUSIONS: The iron-overloaded mouse model was successfully established by intraperitoneal administration of iron dextran. Iron overload can damage the hepatic, splenic, and bone marrow hematopoietic function, although no significant difference was observed in peripheral blood count.


Subject(s)
Bone Marrow/drug effects , Disease Models, Animal , Hematopoiesis/drug effects , Iron Overload/physiopathology , Iron-Dextran Complex/toxicity , Animals , Bone Marrow/physiopathology , Iron Overload/chemically induced , Iron-Dextran Complex/administration & dosage , Male , Mice , Mice, Inbred C57BL , Spleen/drug effects
18.
Stem Cells Dev ; 32(19-20): 581-591, 2023 10.
Article in English | MEDLINE | ID: mdl-37498768

ABSTRACT

As the body ages, it experiences a gradual decline in the functioning of cells, tissues, and systems, which eventually leads to dysfunction and increased susceptibility to disease. At the cellular level, a reduction in the activity or number of stem cells is an important feature of cell senescence, and such changes may also directly drive the aging of the organism. Thus, finding ways to prevent or even reverse stem cell senescence holds promise for the development of aging therapies in tissues and organisms. This review discusses the relationship between changes in stem cell senescence, tissues aging, and related diseases, focusing on four categories of tissue stem cells: hematopoietic stem cells, mesenchymal stromal/stem cells (MSCs), intestinal stem cells, and muscle stem cells.


Subject(s)
Cellular Senescence , Mesenchymal Stem Cells , Mesenchymal Stem Cells/physiology , Cells, Cultured
19.
Animal Model Exp Med ; 6(4): 329-336, 2023 08.
Article in English | MEDLINE | ID: mdl-37642199

ABSTRACT

The risk of internal and external exposure to ionizing radiation (IR) has increased alongside the development and implementation of nuclear technology. Therefore, serious security issues have emerged globally, and there has been an increase in the number of studies focusing on radiological prevention and medical countermeasures. Radioprotective drugs are particularly important components of emergency medical preparedness strategies for the clinical management of IR-induced injuries. However, a few drugs have been approved to date to treat such injuries, and the related mechanisms are not entirely understood. Thus, the aim of the present review was to provide a brief overview of the World Health Organization's updated list of essential medicines for 2023 for the proper management of national stockpiles and the treatment of radiological emergencies. This review also discusses the types of radiation-induced health injuries and the related mechanisms, as well as the development of various radioprotective agents, including Chinese herbal medicines, for which significant survival benefits have been demonstrated in animal models of acute radiation syndrome.


Subject(s)
Acute Radiation Syndrome , Civil Defense , Drugs, Essential , Medical Countermeasures , Radiation-Protective Agents , Animals , Acute Radiation Syndrome/drug therapy , Acute Radiation Syndrome/prevention & control , Radiation, Ionizing , Radiation-Protective Agents/pharmacology , Radiation-Protective Agents/therapeutic use
20.
Curr Pharm Biotechnol ; 24(14): 1827-1835, 2023.
Article in English | MEDLINE | ID: mdl-36941815

ABSTRACT

BACKGROUND: Gastric cancer (GC) ranks fifth among all common malignancies globally. Genetic research has revealed several genes that are frequently dis-regulated in GC, such as lysine-specific demethylase 6A (KDM6A) and cadherin-1 (CDH1). OBJECTIVE: This study aimed to examine the expression profile and role of KDM6A in GC, as well as the molecular pathway involved. METHODS: The expression profile and overall survival data of KDM6A were retrieved from the TCGA database. Expression levels of KDM6A were also measured in GC patient samples and compared with those of healthy controls. Furthermore, stable silencing of KDM6A was introduced into the GC cell line NCI-N87, followed by assessments of cell proliferation, migration and invasion, in the xenograft mouse model. The metastatic status of mice injected with NCI-N87 cells was also analyzed. RESULTS: In patients diagnosed with GC, KDM6A was upregulated. Silencing KDM6A reduced the proliferation, migration and invasion of cells, as well as the growth of xenograft tumors. KDM6A knockdown also inhibited metastatic behaviors of injected NCI-N87 cells, as well as elevated CDH1 expression, leading to reversed epithelial-mesenchymal transition. CONCLUSION: KDM6A serves as an oncogene in GC and exerts its pro-tumor functions by repressing the expression of CDH1.


Subject(s)
Stomach Neoplasms , Animals , Humans , Mice , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Histone Demethylases/genetics , Histone Demethylases/metabolism , Lysine/genetics , Lysine/metabolism , Stomach Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL