1.The in-vivo assessment of Turkish propolis and its nano form on testicular damage induced by cisplatin.
Pinar TATLI SEVEN ; Ismail SEVEN ; Selcan KARAKUS ; Seda IFLAZOGLU MUTLU ; Seyma OZER KAYA ; Gozde ARKALI ; Merve ILGAR ; Ezgi TAN ; Yesim Muge SAHIN ; Deniz ISMIK ; Ayben KILISLIOGLU
Journal of Integrative Medicine 2021;19(5):451-459
OBJECTIVE:
Chemotherapeutic drugs, such as cisplatin (CP), which are associated with oxidative stress and apoptosis, may adversely affect the reproductive system. This study tests whether administration of propolis and nano-propolis (NP) can alleviate oxidative stress and apoptosis in rats with testicular damage induced by CP.
METHODS:
In this study, polymeric nanoparticles including propolis were synthesized with a green sonication method and characterized using Fourier transform-infrared spectroscopy, Brunauer-Emmett-Teller, and wet scanning transmission electron microscopy techniques. In total, 56 rats were divided into the following seven groups: control, CP, propolis, NP-10, CP + propolis, CP + NP-10, and CP + NP-30. Propolis (100 mg/kg), NP-10 (10 mg/kg), and NP-30 (30 mg/kg) treatments were administered by gavage daily for 21 d, and CP (3 mg/kg) was administered intraperitoneally in a single dose. After the experiment, oxidative stress parameters, namely, malondialdehyde (MDA), glutathione (GSH), glutathione peroxidase (GPx), and catalase (CAT), and apoptotic pathways including B cell leukemia/lymphoma-2 protein (Bcl-2) and Bcl-2-associated X protein (Bax) were measured in testicular tissues. Furthermore, sperm quality and weights of the testis, epididymis, right cauda epididymis, seminal vesicles and prostate were evaluated.
RESULTS:
Propolis and NP (especially NP-30) were able to preserve oxidative balance (decreased MDA levels and increased GSH, CAT, and GPx activities) and activate apoptotic pathways (decreased Bax and increased Bcl-2) in the testes of CP-treated rats. Sperm motility in the control, CP, and CP + NP-30 groups were 60%, 48.75%, and 78%, respectively (P < 0.001). Especially, NP-30 application completely corrected the deterioration in sperm features induced by CP.
CONCLUSION
The results show that propolis and NP treatments mitigated the side effects of CP on spermatogenic activity, antioxidant situation, and apoptosis in rats.
Animals
;
Antioxidants/metabolism*
;
Cisplatin/toxicity*
;
Male
;
Oxidative Stress
;
Propolis
;
Rats
;
Rats, Sprague-Dawley
;
Sperm Motility
;
Testis
2.Biliverdin protects against cisplatin-induced apoptosis of renal tubular epithelial cells.
Qian LV ; Ying YAO ; Wei WANG ; Wei XIONG ; Wen-hui LIAO
Journal of Huazhong University of Science and Technology (Medical Sciences) 2016;36(1):48-52
Biliverdin (BV) has long been thought to be a cytotoxic metabolic waste product. It has also been demonstrated to have important cytoprotective functions during oxidative stress. The present study aimed to examine the cytoprotective effect of BV on NRK-52E cells, a proximal tubular cell line derived from rat kidney. Cells were treated with 50 µmol/L cisplatin for 24 h (cisplatin group) or pre-treated with BV for 30 min, then with 50 µmol/L cisplatin for 24 h (cisplatin+BV group). Those given no treatment served as a control. Cell apoptosis was evaluated by flow cytometry and cell viability by Cell Counting Kit-8 (CCK-8). The protein expressions of cleaved caspase3, Bax and Bcl-2 were assessed by Western blotting. Reactive oxygen species (ROS) levels were measured using carboxydichlorodihydrofluorescein diacetate (H2DCF). The results showed that cisplatin induced the apoptosis of NRK-52E cells, decreased cell viability, and increased the formation of ROS by upregulating the expression of cleaved caspase3 and Bax and decreasing Bcl-2 protein expression. These effects could be significantly reversed by pretreatment with BV. It was concluded that BV can protect against cisplatin-induced cell apoptosis through the anti-oxidative effects.
Animals
;
Antioxidants
;
pharmacology
;
Apoptosis
;
Biliverdine
;
pharmacology
;
Cell Line
;
Cisplatin
;
toxicity
;
Epithelial Cells
;
drug effects
;
metabolism
;
Kidney Tubules
;
cytology
;
Rats
;
Reactive Oxygen Species
;
metabolism
3.Bucillamine prevents cisplatin-induced ototoxicity through induction of glutathione and antioxidant genes.
Se Jin KIM ; Joon Ho HUR ; Channy PARK ; Hyung Jin KIM ; Gi Su OH ; Joon No LEE ; Su Jin YOO ; Seong Kyu CHOE ; Hong Seob SO ; David J LIM ; Sung K MOON ; Raekil PARK
Experimental & Molecular Medicine 2015;47(2):e142-
Bucillamine is used for the treatment of rheumatoid arthritis. This study investigated the protective effects of bucillamine against cisplatin-induced damage in auditory cells, the organ of Corti from postnatal rats (P2) and adult Balb/C mice. Cisplatin increases the catalytic activity of caspase-3 and caspase-8 proteases and the production of free radicals, which were significantly suppressed by pretreatment with bucillamine. Bucillamine induces the intranuclear translocation of Nrf2 and thereby increases the expression of gamma-glutamylcysteine synthetase (gamma-GCS) and glutathione synthetase (GSS), which further induces intracellular antioxidant glutathione (GSH), heme oxygenase 1 (HO-1) and superoxide dismutase 2 (SOD2). However, knockdown studies of HO-1 and SOD2 suggest that the protective effect of bucillamine against cisplatin is independent of the enzymatic activity of HO-1 and SOD. Furthermore, pretreatment with bucillamine protects sensory hair cells on organ of Corti explants from cisplatin-induced cytotoxicity concomitantly with inhibition of caspase-3 activation. The auditory-brainstem-evoked response of cisplatin-injected mice shows marked increases in hearing threshold shifts, which was markedly suppressed by pretreatment with bucillamine in vivo. Taken together, bucillamine protects sensory hair cells from cisplatin through a scavenging effect on itself, as well as the induction of intracellular GSH.
Animals
;
Antioxidants/*metabolism/*pharmacology
;
Apoptosis/drug effects
;
Caspase 3/metabolism
;
Caspase 8/metabolism
;
Cell Line
;
Cisplatin/*toxicity
;
Cysteine/*analogs & derivatives/pharmacology
;
Gene Expression Regulation/*drug effects
;
Gene Knockdown Techniques
;
Glutathione/*metabolism
;
Heme Oxygenase-1/genetics
;
Intracellular Space/metabolism
;
Male
;
Metabolic Detoxication, Phase II/genetics
;
Mice
;
NF-E2-Related Factor 2/genetics
;
Nitric Oxide/biosynthesis
;
Organ of Corti/*drug effects/*metabolism
;
RNA Interference
;
Rats
;
Reactive Oxygen Species/metabolism
;
Superoxide Dismutase/genetics
4.Granulocyte-colony stimulating factor decreases the extent of ovarian damage caused by cisplatin in an experimental rat model.
Ali AKDEMIR ; Burak ZEYBEK ; Levent AKMAN ; Ahment Mete ERGENOGLU ; Ahmet Ozgur YENIEL ; Oytun ERBAS ; Altug YAVASOGLU ; Mustafa Cosan TEREK ; Dilek TASKIRAN
Journal of Gynecologic Oncology 2014;25(4):328-333
OBJECTIVE: To investigate whether granulocyte-colony stimulating factor (G-CSF) can decrease the extent of ovarian follicle loss caused by cisplatin treatment. METHODS: Twenty-one adult female Sprague-Dawley rats were used. Fourteen rats were administered 2 mg/kg/day cisplatin by intraperitoneal injection twice per week for five weeks (total of 20 mg/kg). Half of the rats (n=7) were treated with 1 mL/kg/day physiological saline, and the other half (n=7) were treated with 100 microg/kg/day G-CSF. The remaining rats (n=7, control group) received no therapy. The animals were then euthanized, and both ovaries were obtained from all animals, fixed in 10% formalin, and stored at 4degrees C for paraffin sectioning. Blood samples were collected by cardiac puncture and stored at -30degrees C for hormone assays. RESULTS: All follicle counts (primordial, primary, secondary, and tertiary) and serum anti-Mullerian hormone levels were significantly increased in the cisplatin+G-CSF group compared to the cisplatin+physiological saline group. CONCLUSION: G-CSF was beneficial in decreasing the severity of follicle loss in an experimental rat model of cisplatin chemotherapy.
Animals
;
Anti-Mullerian Hormone/blood
;
Antineoplastic Agents/*toxicity
;
Biological Markers/blood
;
Cisplatin/*toxicity
;
Disease Models, Animal
;
Drug Evaluation, Preclinical/methods
;
Female
;
Fertility Preservation/methods
;
Granulocyte Colony-Stimulating Factor/*therapeutic use
;
Ovarian Follicle/drug effects/pathology
;
Primary Ovarian Insufficiency/blood/chemically induced/pathology/*prevention & control
;
Rats, Sprague-Dawley
5.Effects of Antioxidant on Reduction of Hindlimb Muscle Atrophy Induced by Cisplatin in Rats.
Journal of Korean Academy of Nursing 2014;44(4):371-380
PURPOSE: The purpose of this study was to examine the effects of Cu/Zn SOD on reduction of hindlimb muscular atrophy induced by cisplatin in rats. METHODS: Forty-two rats were assigned to three groups; control group, Cisplatin (CDDP) group and cisplatin with Cu/Zn SOD (CDDP-SOD) group. At day 35 hindlimb muscles were dissected. Food intake, activity, withdrawal threshold, muscle weight, and Type I, II fiber cross-sectional area (CSA) of dissected muscles were measured. Relative SOD activity and expression of MHC and phosphorylated Akt, ERK were measured after dissection. RESULTS: Muscle weight and Type I, II fiber CSA of hindlimb muscles in the CDDP group were significantly less than the control group. Muscle weight and Type I, II fiber CSA of hindlimb muscles, food intake, activity, and withdrawal thresholds of the CDDP-SOD group were significantly greater than the CDDP group. There were no significant differences in relative SOD activities of hindlimb muscles between the CDDP-SOD and CDDP groups. MHC expression and phosphorylated Akt, ERK of hindlimb muscles in the CDDP-SOD group were significantly greater than the CDDP group. CONCLUSION: Cu/Zn SOD attenuates hindlimb muscular atrophy induced by cisplatin through increased food intake and activity. Increment of phosphorylated Akt, ERK may relate to attenuation of hindlimb muscular atrophy.
Animals
;
Body Weight/drug effects
;
Cisplatin/*toxicity
;
Disease Models, Animal
;
Extracellular Signal-Regulated MAP Kinases/metabolism
;
Hindlimb
;
Male
;
Muscle, Skeletal/*drug effects/enzymology/metabolism
;
Muscular Atrophy/*chemically induced/metabolism/pathology
;
Phosphorylation
;
Proto-Oncogene Proteins c-akt/metabolism
;
Rats
;
Rats, Sprague-Dawley
;
Recombinant Proteins/biosynthesis/genetics/pharmacology
;
Superoxide Dismutase/genetics/metabolism/pharmacology
;
Superoxides/metabolism
6.Ligustrazine fought against cisplatin induced ototoxicity in guinea pigs.
Xian-Chang SUN ; Li-Xia SUN ; Xian-Jun SHI ; Song-Jian KANG
Chinese Journal of Integrated Traditional and Western Medicine 2014;34(8):982-986
OBJECTIVETo study the protective effect of Ligustrazine Injection (LI) against cisplatin-induced ototoxicity and to explore its mechanism.
METHODSThirty healthy adult guinea pigs were randomly divided into three groups, 10 in each group, i.e., the normal control group, the cisplatin group, and the LI group. Guinea pigs in the normal control group were intraperitoneally injected with normal saline at 3 mL/kg for 7 consecutive days. Those in the cisplatin group were intraperitoneally injected with cisplatin at 3 mg/kg for 7 consecutive days. Those in the LI group were intraperitoneally injected with LI at 140 mg/kg for 7 days, but cisplatin (3 mg/kg) was intraperitoneally injected from the opposite side starting from the 4th day. Brainstem auditory evoked potential (BAEP) was performed in all animals before and after injection. All animals were sacrificed after the final testing under anesthesia and their cochleas collected. Half the cochleas of each group were collected for silver nitrate staining of cochlear basilar membrane stretched. The other half the cochleas of each group made into paraffin sections to observe the apoptosis of cochlea cells by TUNEL method and the expression levels of c-Jun detected by immunohistochemistry.
RESULTSThere was no statistical difference in the difference of BAEP threshold among the 3 groups before injection (P > 0.05), but the BAEP threshold increased in the cisplatin group and the LI group (P < 0.05). Besides, it was higher in the cisplatin group (P < 0.05). In the cisplatin group, most hair cells were missing, spiral ganglion cells obviously decreased, more TUNEL positive cells occurred, and the expression of c-Jun was stronger. But the aforesaid impairment in the LI group was obviously lessened (P < 0.05).
CONCLUSIONSLI showed certain antagonist effect on cisplatin-induced ototoxicity. Its mechanism might be associated with scavenging oxygen radicals of the cochlea tissue, improving the microcirculation, and fighting against apoptosis.
Animals ; Apoptosis ; drug effects ; Cisplatin ; toxicity ; Cochlea ; drug effects ; metabolism ; pathology ; Evoked Potentials, Auditory, Brain Stem ; Guinea Pigs ; Pyrazines ; pharmacology ; Reactive Oxygen Species ; metabolism
7.Low doses of ethanolic extract of Boldo (Peumus boldus) can ameliorate toxicity generated by cisplatin in normal liver cells of mice in vivo and in WRL-68 cells in vitro, but not in cancer cells in vivo or in vitro.
Jesmin MONDAL ; Kausik BISHAYEE ; Ashis Kumar PANIGRAHI ; Anisur Rahman KHUDA-BUKHSH ; E-mail: prof_arkb@yahoo.co.in, KHUDABUKHSH_48@REDIFFMAIL.COM.
Journal of Integrative Medicine 2014;12(5):425-438
OBJECTIVEUse of cisplatin, a conventional anticancer drug, is restricted because it generates strong hepatotoxicity by accumulating in liver. Therefore its anticancer potential can only be fully exploited if its own toxicity is considerably reduced. Towards this goal, ethanolic extract of the plant, Boldo (Peumus boldus), known for its antihepatotoxic effects, was used simultaneously with cisplatin, to test its ability to reduce cisplatin's cytotoxicity without affecting its anticancer potential.
METHODSThe cytotoxicity of Boldo extract (BE) and cisplatin, administered alone and in combination, was determined in three cancer cell lines (A549, HeLa, and HepG2) and in normal liver cells (WRL-68). Drug-DNA interaction, DNA damage, cell cycle, apoptosis, reactive oxygen species (ROS) and mitochondrial membrane potential (MMP, ΔΨ) were also studied. Hepatotoxicity and antioxidant activity levels were determined by alanine aminotransferase, aspartate aminotransferase, lactate dehydrogenase and glutathione assays in mice. The cytotoxicity of related proteins was tested by Western blotting.
RESULTSCo-administration of BE and cisplatin increased viability of normal cells, but had no effect on the viability of cancer cells. Boldo protected liver from damage and normalized different antioxidant enzyme levels in vivo and also reduced ROS and re-polarized MMP in vitro. Bax and cytochrome c translocation was reduced with caspase 3 down-regulation. Further, a drug-DNA interaction study revealed that BE reduced cisplatin's DNA-binding capacity, resulting in a reduction in DNA damage.
CONCLUSIONResults indicated that a low dose of BE could be used beneficially in combination with cisplatin to reduce its toxicity without hampering cisplatin's anticancer effect. These findings signify a potential future use of BE in cancer therapy.
Animals ; Antineoplastic Agents ; toxicity ; Cells, Cultured ; Cisplatin ; toxicity ; DNA Damage ; Female ; Glutathione ; metabolism ; Hepatocytes ; drug effects ; metabolism ; pathology ; Humans ; Male ; Mice ; Neoplasms ; drug therapy ; pathology ; Peumus ; Plant Extracts ; pharmacology
8.Effect of low dosage of ciplatin on the shape and otoferlin in cochlea inner hair cells.
Ying SUN ; Xiaojun JIANG ; Xiaohong CHEN ; Wei YUAN
Journal of Clinical Otorhinolaryngology Head and Neck Surgery 2014;28(11):793-796
OBJECTIVE:
To establish the stable and efficient hearing damage model by using low dosage of cispla tin, and investigate the mechanism.
METHOD:
C57 mice were divided into 7 groups (every group, n = 8), the first group was the control group, the others were separately intraperitoneally injected with different dosages of cispla tin for different time. We measured the auditory brainstem response (ABR) of the mice, and obtained the basal coil of organ Corti. We observed the shape of inner hair cells (IHC) by staining AgNO3 and marked otoferlin in the IHC by immunofluorescence,successively sliced by laser confocal microscopy. The RNA fragments were amplified by RT-PCR.
RESULT:
After cisplatin administration for four days, the thresholds of the ABR improved in 1.5 mg/kg and 3.0 mg/kg group, and compared with the control group, the ABR thresholds improved in each group with ciplatin administration for seven days. With the same dosage, the ABR threshold of the 0.75 mg/kg x 7 d group was higher than 0.75 mg/kg x 4 d group, and there was no time-effect relationship existing in other groups with different dosage. The otoferlin was less expressed 3.0 mg/kg groups than the control group. However, the oto ferlin expressed in the 1.5 mg/kg groups were almost the same as the control group. The alteration of the IHC was observed most remarkablely in 3.0 mg/kg x 7 d group.
CONCLUSION
Low dosage of cisplatin can impair the hearing, and the expression of otoferlin may involve in the underlying mechanism.
Animals
;
Cisplatin
;
toxicity
;
Cochlea
;
drug effects
;
metabolism
;
pathology
;
Hair Cells, Auditory, Inner
;
drug effects
;
pathology
;
Membrane Proteins
;
metabolism
;
Mice
;
Mice, Inbred C57BL
9.The effect of forsythiaside on the expression of c-jun induced by cisplatin in the cochlea of guinea pig.
Journal of Clinical Otorhinolaryngology Head and Neck Surgery 2014;28(10):731-734
OBJECTIVE:
To study the effect of forsythiaside on the expression of c-jun induced by cisplatin in the cochlea of guinea pig.
METHOD:
Thirty guinea pigs were randomly divided into control group (10), cisplatin group (10) and forsythiaside group (10). The ototoxicity model was done with intraperitoneal injection of cisplatin solution (8 mg/kg per day) for 7 days. Forsythiaside (25 mg/kg per day) was injected 30 min before cisplatin solution treated in guinea pigs of forsythiaside group for 7 consecutive days. The saline instead of cisplatin was injected in normal control group. The distortion product otoacoustic emission (DPOAE) was detected before animals were killed. The expression of c-jun in cochlea of guinea pigs was detected by western blotting. The expression of c-jun mRNA in cochlea of guinea pigs was detected by reverse transcriptase polymerase chain reaction (RT-PCR).
RESULT:
DPOAE amplitudes in cisplatin group was significantly lower than in control group (P < 0.01). Compared with cisplatin group, DPOAE amplitudes in forsythiaside group was increased significantly (P < 0.05). The expression of c-jun protein and mRNA were significantly increased in cisplatin group than in control group (P < 0.01). Compared with cisplatin group, the expression of c-jun protein and mRNA were significantly decreased in forsythiaside group.
CONCLUSION
Forsythiaside can significantly reduce the side effects induced by cisplatin through down-regulating the expression of c-jun.
Animals
;
Cisplatin
;
toxicity
;
Cochlea
;
drug effects
;
metabolism
;
Female
;
Glycosides
;
pharmacology
;
Guinea Pigs
;
Male
;
Proto-Oncogene Proteins c-jun
;
metabolism
10.Effect of Anorexia and Neuropathic Pain Induced by Cisplatin on Hindlimb Muscles of Rat.
Journal of Korean Academy of Nursing 2013;43(3):361-369
PURPOSE: The purpose of this study was to examine the effect of anorexia and neuropathic pain induced by cisplatin on hindlimb muscles of rats. METHODS: Adult male Sprague-Dawley rats were divided into two groups, a cisplatin-treated group (n=10) and a control group (n=10). In the cisplatin-treated group, cisplatin at a dose of 2 mg/kg was injected intraperitoneally two times a week up to a cumulative dose of 20 mg/kg over 5 weeks, and in the control group saline (0.9% NaCl) was injected intraperitoneally at the same dose and duration as the cisplatin-treated group. At 34 days all rats were anesthetized, after which the soleus and plantaris muscles were dissected. Withdrawal threshold, body weight, food intake, activity, muscle weight, Type I and II fiber cross-sectional areas and myofibrillar protein content of the dissected muscles were determined. RESULTS: Compared with the control group, the cisplatin-treated group showed significant decreases (p<.05) in withdrawal threshold, activity, food intake, body weight, Type I and II fiber cross-sectional areas, myofibrillar protein content and weight of the soleus and plantaris muscles. CONCLUSION: Muscular atrophy in hindlimb occurs due to anorexia and neuropathic pain induced by the cisplatin treatment.
Animals
;
*Anorexia
;
Body Weight
;
Cisplatin/*toxicity
;
Eating
;
Hindlimb
;
Injections, Intraperitoneal
;
Male
;
Motor Activity
;
Muscle Fibers, Skeletal/metabolism/pathology
;
Muscle Proteins/metabolism
;
Muscle, Skeletal/*drug effects/physiology
;
Neuralgia/*chemically induced/pathology
;
Rats
;
Rats, Sprague-Dawley

Result Analysis
Print
Save
E-mail