Search results

Filters

  • Journals
  • Authors
  • Keywords
  • Date
  • Type

Search results

Number of results: 21
items per page: 25 50 75
Sort by:
Download PDF Download RIS Download Bibtex

Abstract

Calretinin (CR), a calcium-binding protein from EF-hand family, is localised in non-pyramidal GABA-ergic interneurons of the hippocampus. CR takes part in maintaining calcium binding homeostasis, which suggests its neuroprotective role. Hippocampal neurons contain membrane transient receptor potential vanilloid 1 (TRPV1) which binds to capsaicin (CAP) contained in habanero pepper fruits. Few in vivo studies have revealed the effect of CAP on interneurons containing CR. The aim of the present study was to investigate the CR immunoreac- tivity in interneurons of the hippocampal CA1 field and dentate gyrus (DG) in adult rats after intragastric admin- istration of the habanero pepper fruits. Wistar rats received a peanut oil – control group (C), and oil suspension of habanero pepper fruits at doses of 0.025 g dm/kg b.w. – group I and 0.08 g dm/kg b.w. – group II for 28 days. After euthanasia, the brains were collected and embedded in paraffin blocks using a routine histological tech- nique. Frontal hippocampal sections were immunohistochemically stained for CR by using a peroxidase-antiper- oxidase method. CR immunoreactive (CR-IR) interneurons were morphologically and morphometrically ana- lyzed under a light microscope. The results showed similar shapes and distribution of cells in both areas of the brain in group C and I of animals. However, CR-IR interneurons in the hippocampal CA1 field and in DG were occasionally observed in the group II of rats.

The results of morphometric studies did not reveal statistically significant differences in the surface area and shape index of cells between examined brain regions from groups I and II compared to group C.

Only in group II of rats, an increase in the digital immunostaining intensity of CR-IR interneurons was found in DG. Low number of CR-IR interneurons in the hippocampal CA1 field and in the DG, under the influence of a large dose of habanero pepper fruits containing CAP, may be caused by the activation of TRPV1 receptors and the increase in Ca2+ ions in these cells. This phenomenon may ultimately lead to neuronal death and may disturb neuronal conduction.

Go to article

Authors and Affiliations

J. Jaworska-Adamu
A. Krawczyk
K. Rycerz
M. Gołyński
A. Wawrzyniak
K. Lutnicki
I. Balicki
Download PDF Download RIS Download Bibtex

Abstract

The present study was aimed to investigate oxidative stress, DNA damage, and histopatholog- ical alterations in hepatic tissues of splenectomized Wistar rats experimentally infected with Ba- besia bigemina. Rats were challenged with 5x106 infected erythrocytes. Babesia infection was con- firmed both with Giemsa’s staining blood smears and nested-PCR amplified region of apical membrane antigen-1 (AMA-1) gene. Parasitemia reached approximately 10 % at day 5 post-in- fection. Livers of infected rats were enlarged and darker in color, became extremely brittle with marked congestion. Microscopic evaluation showed cytoplasmic clearing of hepatocytes and se- vere hydropic changes with significantly dilated sinusoids containing macrophages and also intra- sinosoidal parasitized erythrocytes. Severe infiltration of lymphoplasma cells was also present throughout the liver parenchyma. Furthermore, Kupffer cells were enlarged and, occasionally, containing Babesia-parasitized erythrocytes. The activity of Glutathione (GSH) and catalase (CAT), and total antioxidant capacity (TAC) were also significantly decreased (p < 0.05) after infection of rats with B. bigemina. B. bigemina infection also induced a significant increase (p < 0.05) in hepatic malondialdehyde (MDA) and nitric oxide-derived products (NOx) concentra- tions as well as amount of endogenous hepatocytes DNA damage. Hepatic damage was also re- flected through the measurement of lactic acid dehydrogenase (LDH) and protein carbonyl con- tent (PCO) in liver cells. These two indices of liver injury were also significantly elevated (p < 0.5) during B. bigemina infection. Evaluation of correlation between assayed variables in infected rats revealed that MDA levels were positively correlated with PCO, NOx, LDH and DNA damage in the infected group and negatively correlated with GSH, CAT and TAC. There was also an inverse relationship between the antioxidant enzymes activities of GSH, CAT and TAC with PCO, NOx and DNA damage in infected rats. However, NOx showed positive correlation with PCO and DNA damage in infected rats. On the basis of the above results it can be concluded that the Ba- besia infection increases oxidative stress markers, protein carbonyl content and DNA damage and decreases antioxidant enzymes activities in the liver. These results suggest that B. bigemina infec- tion could alter the liver histopathology and causes DNA damage following oxidative stress in hepatic tissue. Further studies are needed to precisely define how hepatic tissue damage takes place in B. bigemina infection.

Go to article

Authors and Affiliations

B. Esmaeilnejad
M. Tavassoli
A. Samiei
A. Abbasi
A. Shafipour
N. Esmaeilnejad
Download PDF Download RIS Download Bibtex

Abstract

Introduction: Effective and safe anesthesia for rodents has long been a leading concern among biomedical researchers. Intraperitoneal injection constitutes an alternative to inhalant anesthesia.

Purpose: The aim of this study was to identify a safe, reliable, and effective anesthesia and postoperative analgesia protocol for laboratory rats exposed to painful procedures.

Material and methods: Twenty-seven female Wistar rats in an ongoing study that required surgery were randomized into groups for three different intraperitoneal anesthesia protocols and three different analgesia regimens. The anesthesia groups were (1) medetomidine + ketamine (MK), (2) ketamine + xylacine (KX), and (3) fentanyl + medetomidine (FM). Three analgesia groups were equally distributed among the anesthesia groups: (1) local mepivacaine + oral ibuprofen (MI), (2) oral tramadol + oral ibuprofen (TI), and (3) local tramadol + oral tramadol + oral ibuprofen (TTI). A core was assigned to measure anesthesia (0-3) and analgesia (0-2) effectiveness; the lower the score, the more effective the treatment.

Results: The mean MK score was 0.44 versus 2.00 for FM and 2.33 for KX. Mean score for analgesia on the first postoperative day was TTI (4.66) TI (9.13), and MI (10.14). Mean score 48 hours after surgery was TTI (3.4), TI (6.71), and MI (9.5). These differences were statistically significant.

Conclusion: MK was shown to be a reliable, safe, and effective method of anesthesia. The TTI analgesia regimen is strongly recommended in light of these results.

Go to article

Authors and Affiliations

F. Moreno-Mateo
B. García-Medrano
A. Álvarez-Barcia
M.J. Gayoso
M.A. Martín-Ferrero
Download PDF Download RIS Download Bibtex

Abstract

The study was undertaken to determine the effect of continuation or changes of the diet on the morphometry and histomorphometry of bone in male and female Wistar rats with experimen- tally induced obesity by high energetic diet. Sixty-four 90-day-old Wistar rats obtained from obese parents (16 male, 16 female) and control parents (16 male, 16 female) were used in this study. After 21 days of the baby period, rats were divided into four groups: obese rats fed with high energy feed (F/F), control rats fed with a standard diet (C/C), obese rats with changed diet from high energy diet to control diet (F/C) and control rats with changed diet from control diet to high energy diet (C/F). After 90 days of experimental feeding, the rats were sacrificed. Thereafter, body weight and the isolated humerus were measured and next, the histological stainings and counts were done. Our results revealed that change in the parent’s diet from F to C in the female leads to increased bone growth length and reduction of body weight in female and male. Reverse diet changes (from C to F) lead to decreased bone length only in the female. Moreover, the con- tinuation by offspring of both sexes with a high-energy diet contributes to a reduction in osteo- cytes, reduction in bone marrow cavity and cortical expansion, but a change in nutrition from parents’ standard diet to high-energy diet leads to increase in osteocytes dimensions. The contin- uation of feeding with F diet promotes the accumulation of adipocytes in the bone marrow in female and male, and correction of nutrition from F to standard diet leads to a reduction in their number in the bone marrow compared to groups continuing feeding with high-energy diet.

Go to article

Authors and Affiliations

D. Wolski
A. Wawrzyniak
M. Bieńko
J. Michalik
R.P. Radzki
M.B. Arciszewski
Download PDF Download RIS Download Bibtex

Abstract

Rebaudioside A (RebA) is a steviol glycoside used for production of sweeteners. It was shown that the glycosides affect memory and learning processes. The aim of the study was to investigate neurons immunoreactive for acetylcholinesterase (AChE) and M1 muscarinic receptors (mAChRs-M1) of the hippocampal CA1 and CA3 fields and striatal caudateputamen (CP) and globus pallidus (GP) in rats receiving RebA. RebA was administrated to adult rats for 45 days in dilutions of 1 mg and 2 mg RebA/ml water. Indirect peroxidase-antiperoxidase immunohistochemical reaction was conducted on frontal sections containing the hippocampus and striatum with use of antibodies against AChE and mAChRs-M1. Immunoreactive for the studied proteins neurons were morphologically and morphometrically assessed in hippocampal CA1 and CA3 fields and in CP and GP. Microscopic observations did not reveal significant changes in morphology of immunoreactive neurons, which suggests no neurotoxic effect of the studied glycoside on these cells. Morphometric analyses revealed an increase in the density of AChE and mAChRs-M1 immunoreactive neurons. A decrease in reaction intensity of AChE-positive neurons was also demonstrated in the hippocampal CA1 field and in GP. In contrast, an increase in reaction intensity of mAChRs-M1-positive neurons was found in CA1, CA3 fields and in CP and GP. The results of our preliminary studies indicate that RebA administrated to rats has an impact on cholinergic neurons in the studied area. The results suggest a possible increase in the activity of the cholinergic system, responsible for memory and learning processes, after administration of RebA.
Go to article

Bibliography


Abdel-Aal RA, Abdel-Rahman MS, Al Bayoumi S, Ali LA (2021) Effect of stevia aqueous extract on the antidiabetic activity of saxagliptin in diabetic rats. J Ethnopharmacol 265: 113188.
Abudula R, Jeppesen PB, Rolfsen SE, Xiao J, Hermansen K (2004) Rebaudioside A potently stimulates insulin secretion from isolated mouse islets: studies on the dose-, glucose-, and calcium-dependency. Metabolism 53: 1378-1381.
Bazzari AH, Parri HR (2019) Neuromodulators and Long-Term Synaptic Plasticity in Learning and Memory: A Steered-Glutamatergic Per-spective. Brain Sci 9:300.
Betterton RT, Broad LM, Tsaneva-Atanasova K, Mellor JR (2017) Acetylcholine modulates gamma frequency oscillations in the hippocampus by activation of muscarinic M1 receptors. Eur J Neurosci 45: 1570-1585.
Casas-Grajales S, Ramos-Tovar E, Chávez-Estrada E, Alvarez-Suarez D, Hernández-Aquino E, Reyes -Gordillo K, Cerda-García-Rojas CM, Camacho J, Tsutsumi V, Lakshman MR, Muriel P (2019) Antioxidant and immunomodulatory activity induced by stevioside in liver damage: In vivo, in vitro and in silico assays. Life Sci 224: 187-196.
Chavushyan VA, Simonyan KV, Simonyan RM, Isoyan AS, Simonyan GM, Babakhanyan MA, Hovhannisyian LE, Nahapetyan KH, Avetisyan LG, Simonyan MA (2017) Effects of stevia on synaptic plasticity and NADPH oxidase level of CNS in conditions of metabolic disorders caused by fructose. BMC Complement Altern Med 17: 540.
Dyduch-Siemińska M, Najda A, Gawroński J, Balant S, Świca K, Żabaet A (2020) Stevia Rebaudiana Bertoni, a Source of High-Potency Natural Sweetener-Biochemical and Genetic Characterization. Molecules 25: 767.
El Falougy H, Kubikova E, Benuska J (2008) The microsco pical structure of the hippocampus in the rat. Bratisl Lek Listy 109: 106-110.
Hampel H, Mesulam MM, Cuello AC, Farlow MR, Giacobini E, Grossberg GT, Khachaturian AS, Vergallo A, Cavedo E, Snyder PJ, Kha-chaturian ZS (2018) The cholinergic system in the pathophysiology and treatment of Alzheimer’s disease. Brain 141: 1917-1933.
Hazali N, Mohamed A, Ibrahim M, Masri M, Isa KA, Nor NM, Ayob MK, Fadzlan FN (2014) Effect of acute Stevia consumption on blood glucose response in healthy Malay young adults. Sains Malays 43: 649-654.
Ihara R, Vincent BD, Baxter MR, Franklin EE, Hassenstab JJ, Xiong C, Morris JC, Cairns NJ (2018) Relative neuron loss in hippocampal sclerosis of aging and Alzheimer’s disease. Ann Neurol 84: 741-753.
Jahanshahi M, Nickmahzar EG, Seif-hoseini S, Babakordi F, Moharreri A (2013) Scopolamine reduces the density of M1 muscarinic neurons in rats’ hippocampus. Int J Morphol 31: 1227-1232.
Janeczek M, Gefen T, Samimi M, Kim G, Weintraub S, Bigio E, Rogalski E, Mesulam M, Geula C (2018) Variations in Acetylcholinesterase Activity within Human Cortical Pyramidal Neurons Across Age and Cognitive Trajectories. Cereb Cortex 28: 1329-1337.
Juyal DS, Arun K, Ganga B (2010a) Effect of Stevia rebau diana (Bert.) extract on memory and acetylcholinesterase activity in young and aged rats. J Glob Pharma Technol 2: 62-68.
Juyal DS, Bisht G, Kumar A (2010b) Memory enhancing effect of ethanolic extract of Stevia rebaudiana (Bert.). Int J Phytomed 2: 166-171.
Konar A, Gupta R, Shukla RK, Maloney B, Khanna VK, Wadhwa R, Lahiri DK, Thakur MK (2019) M1 musca rinic receptor is a key target of neuroprotection, neuroregeneration and memory recovery by i-Extract from Withania somnifera. Sci Rep 9: 13990.
Krawczyk MC, Millan J, Blake MG, Boccia MM (2021) Critical role of hippocampal muscarinic acetylcholine receptors on memory recon-solidation in mice. Neurobiol Learn Mem 177: 107360.
Mann T, Zilles K, Klawitter F, Cremer M, Hawlitschka A, Palomero-Gallagher N, Schmidt O, Wree A (2018) Acetylcholine Neurotransmitter Receptor Densities in the Striatum of Hemiparkinsonian Rats Following Botulinum Neurotoxin-A Injection. Front Neuroanat 12: 65.
Momtazi-Borojeni AA, Esmaeili SA, Abdollahi E, Sahebkar A (2017) A Review on the Pharmacology and Toxicology of Steviol Glycosides Extracted from Stevia rebaudiana. Curr Pharm Des 23: 1616-1622.
Nguyen DH, Zhou T, Shu J, Mao J (2013) Quantifying chromogen intensity in immunohistochemistry via reciprocal intensity. Cancer In-Cytes 2: 1-4.
Ni R, Huang Z, Shu Y, Wang Y, Li T, Zhou J (2018) Atlas of the Striatum and Globus Pallidus in the Tree Shrew: Comparison with Rat and Mouse. Neurosci Bull 34: 405-418.
Nunes AP, Ferreira-Machado SC, Nunes RM, Dantas FJ, De Mattos JC, Caldeira-de-Araujo A (2007) Analysis of genotoxic potentiality of stevioside by comet assay. Food Chem Toxicol 45: 662-666.
Pasqualli T, Chaves PE, Pereira CL, Serpa EA, Oliviera LF, Machado MM (2020) Steviol, the active principle of the stevia sweetener, causes a reduction of the cells of the immunological system even consumed in low concentrations. Immunopharmacol Immunotoxicol 42: 504-508.
Picciotto MR, Higley MJ, Mineur YS (2012) Acetylcholine as a neuromodulator: cholinergic signaling shapes nervous system function and behavior. Neuron 76: 116-129.
Radu BM, Osculati AM, Suku E, Banciu A, Tsenov G, Merigo F, Di Chio M, Banciu DD, Tognoli C, Kacer P, Giorgetti A, Radu M, Bertini G, Fabene PF (2017) All muscarinic acetylcholine receptors (M1-M5) are expressed in murine brain microvascular endothelium. Sci Rep 7: 5083.
Scarpa M, Hesse S, Bradley SJ (2020) M1 muscarinic acetylcholine receptors: A therapeutic strategy for symptomatic and disease-modifying effects in Alzheimer’s disease? Adv Pharmacol 88: 277-310.
Sharma D, Puri M, Tiwary AK, Singh N, Jaggi AS (2010) Antiamnesic effect of stevioside in scopolamine-treated rats. Indian J Pharmacol 42: 164-167.
Tao R, Cho S (2020) Consumer-based sensory characterization of steviol glycosides (Rebaudioside A, D, and M). Foods 9: 1026.
Veurink G, Perry G, Singh SK (2021) Role of antioxidants and a nutrient rich diet in Alzheimer’s disease. Open Biol 10: 200084.
Yi JH, Whitcomb DJ, Park SJ, Martinez-Perez C, Barbati SA, Mitchell SJ, Cho K (2020) M1 muscarinic acetylcholine receptor dysfunction in moderate Alzheimer’s disease pathology. Brain Commun 2: fcaa058.
Go to article

Authors and Affiliations

K. Rycerz
1
J. Jaworska-Adamu
1
A. Krawczyk
1
M.B. Arciszewski
1

  1. Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences, Akademicka 12, 20-033, Lublin, Poland
Download PDF Download RIS Download Bibtex

Abstract

Twenty eight male Sprague Dawley rats (aged 3 months) were used in the study. The animals were given feed and water as ad libitum. Sprague dawley rats were randomly divided into 4 groups as 7 rats in each group. Except for the control one, aflatoxin B1 (7.5 μg / 200 g), resvera- trol (60 mg / kg) was administered to rats of 3 other groups. At the end of the 16th day, blood, semen and tissue specimens were taken by decapitation under ether anesthesia. When we evaluate the spermatological parameters, it is understood that resveratrol has a statistically significant difference in terms of sperm motility and viability (membrane integrity) compared to the control group and aflatoxin B1 administration groups, indicating a protective effect on spermatological parameters. In terms of pathological parameters - histopathological examination - in the control and resveratrol groups, seminiferous tubules were observed to be in normal structure. In the group treated with aflatoxin, the regular structure of the spermatogenic cells deteriorated and the seminiferous tubules became necrotic and degenerative. In the group treated with Afb1 + res, the decreasing of necrotic and degenerative changes were determined compared with in the group treated with aflatoxin. As immunohistochemical examination, cleaved caspase 3 expression was found to be very low in the control and resveratrol groups. Cleaved caspase 3 expression was severely exacerbated in seminiferous tubules in aflatoxin group but cleaved caspase 3 expression level decreased in Afb1 + res. In the biochemical direction, resveratrol has been shown to inhibit the adverse effects of aflatoxin on antioxidant levels and to show a protective effect. For this purpose, the use of resveratrol with antioxidant activity was investi- gated in preventing or ameliorating damage to aflatoxin B1. It has been concluded that resveratrol effectively prevent the aflatoxin-induced testicular damage and lipid peroxidation. It has also been shown that resveratrol has protective effects on sperm motility and viability.

Go to article

Authors and Affiliations

A.D. Omur
B. Yildirim
Y.S. Saglam
S. Comakli
M. Ozkaraca
Download PDF Download RIS Download Bibtex

Abstract

Nickel damages the male reproductive system. We investigated the beneficial effects of silibinin which has metal-chelating and antioxidant properties over nickel toxicity. Both antioxidative effects in testes and overall effects related to sperm motility, membrane and acrosome integrity of orally administered Silibinin were evaluated against the harmful effects of 30 day of intraperitoneal nickel sulfate (5 mg/kg/day) administration in rats. Male rats were randomized into control (Group1; n=6) and three experimental groups (n=6, each): Group2 Nickel sulfate (5 mg/kg/day), Group3 Silibinin (150 mg/kg/day), and Group 4 Nickel sulfate (5 mg/kg/day) + Silibinin (150 mg/kg/day). We found higher sperm motility, viable sperm and total sperm count in Groups 3 and 4 than the Group 2 treatment groups and the percentage of abnormal spermatozoa was similar in both groups (Groups 2 and 4). Increased apoptosis, activation of caspase3, 8, 9 and TUNEL were detected in Group 2. However, activation of caspase3, 8, 9 and TUNEL was reduced in Group 4. The protective effects of silibinin were demonstrated on histopathologic findings and some sperm parameters (sperm motility percentage, viable spermatozoa, sperm count, and abnormal spermatozoa percentage) in rats exposed to nickel.
Go to article

Authors and Affiliations

F. Temamogullari
1
A. Atessahin
2
C. Cebi Sen
3
N. Yumusak
4
M.S. Dogru
5

  1. Department of Pharmacology and Toxicology, University of Harran, Faculty of Veterinary Medicine, 63200 Şanlıurfa, Turkey
  2. Department of Pharmacology and Toxicology, University of Fırat, Faculty of Veterinary Medicine, 23119 Elazığ, Turkey
  3. Department of Reproduction and Artificial Insemination, University of Harran, Faculty of Veterinary Medicine, 63200 Şanlıurfa, Turkey
  4. Department of Pathology, University of Harran, Faculty of Veterinary Medicine, 63200 Şanlıurfa, Turkey
  5. Department of Pharmacology and Toxicology, University of Aksaray, Faculty of Veterinary Medicine, 6800 Aksaray, Turkey
Download PDF Download RIS Download Bibtex

Abstract

The purpose of this study is to determine the effects of proanthocyanidin (PA) on spermatological parameters and testicular toxicity in male rats exposed to glyphosate (GLP). In our study, four groups were formed out of 24 male rats, each group would include 6 rats. The rats in the PA group were given a dose of 400 mg/kg/day dissolved in DMSO via gastric gavage. The rats in the GLP+PA groups were first given GLP at the LD50/10 dose of 787.85 mg/kg/day, followed by administering PA at a dose of 400 mg/kg/day dissolved in DMSO via gastric gavage. The rats in the GLP group were given GLP at the LD50/10 dose of 787.85 mg/kg/day dissolved in DMSO via gastric gavage. It was determined that in terms of motility, in comparison to the control group, the decreases in the GLP group and the increases in the PA and GLP+PA groups were statistically significant (p<0.001). The administration of GLP increased DNA damage compared to the control group, but the GLP+PA and PA applications reduced DNA damage (p<0.001). The analysis of testosterone levels indicated a statistically significant reduction in the GLP group compared to the other groups. Consequently, it was determined that PA effectively prevented the decreases in the spermatological parameters lowered as a result of GLP exposure and the oxidative stress and toxicity in testicular tissue.
Go to article

Bibliography

1. Abarikwu SO, Akiri OF, Durojaiye MA, Adenike A (2015) Combined effects of repeated administration of Bretmont Wipeout (glypho-sate) and Ultrazin (atrazine) on testosterone, oxidative stress and sperm quality of Wistar rats. Toxicol Mech Methods 25: 70-80.
2. Abdel-Kawi SH, Hashem KS, Abd-Allah S (2016) Mechanism of diethylhexylphthalate (DEHP) induced testicular damage and of grape seed extract-induced protection in the rat. Food Chem Toxicol 90: 64-75.
3. Acquavella JF, Alexander BH, Mandel JS, Gustin C, Baker B, Chapman P, Bleeke M (2004) Glyphosate biomonitoring for farmers and their families: results from the Farm Family Exposure Study. Environ Health Perspect 112: 321-326.
4. Al-Daraji HJ (2012) The use of certain grape constituents for improve semen quality and storage ability of diluted Roosters’ semen. Am J Pharm Tech Res 2: 308-322.
5. Attia SM, Al-Bakheet SA, Al-Rasheed NM (2010) Proanthocyanidins produce significant attenuation of doxorubicininduced mutagenic-ity via suppression of oxidative stress. Oxid Med Cell Longev 3: 404-413.
6. Avdatek F, Birdane YO, Türkmen R, Demirel HH (2018) Ameliorative effect of resveratrol on testicular oxidative stress, spermatological parameters and DNA damage in glyphosate-based herbicide-exposed rats. Andrologia 50: e13036.
7. Bashir N, Shagirtha K, Manoharan V, Miltonprabu S (2019) The molecular and biochemical insight view of grape seed proanthocya-nidins in ameliorating cadmium-induced testes-toxicity in rat model: implication of PI3K/Akt/Nrf-2 signaling. Biosci Rep 39: BSR20180515.
8. Bayatli F, Akkuş D, Kilic E, Saraymen R, Sönmez MF (2013) The protective effects of grape seed extract on MDA, AOPP, apoptosis and eNOS expression in testicular torsion: an experimental study. World J Urol 31: 615-622.
9. Beutler E, Kelly BM (1963) The effect of sodium nitrite on red cell GSH. Experientia 19: 96-97.
10. Bolognesi C, Carrasquilla G, Volpi S, Solomon KR, Marshall EJ (2009) Biomonitoring of genotoxic risk in agricultural workers from five colombian regions: association to occupational exposure to glyphosate. J Toxicol Environ Health A 72: 986-997.
11. Cai W, Ji Y, Song X, Guo H, Han L, Zhang F, Liu X, Zhang H, Zhu B, Xu M (2017) Effects of glyphosate exposure on sperm concen-tration in rodents: A systematic review and meta-analysis. Environ Toxicol Pharmacol 55: 148-155.
12. Cavas T, Könen S (2007) Detection of cytogenetic and DNA damage in peripheral erythrocytes of goldfish (Carassius auratus) exposed to a glyphosate formulation using the micronucleus test and the comet assay. Mutagenesis 22: 263-268.
13. Choy YY, Waterhouse AL (2014) Proanthocyanidin metabolism, a mini review. Nutr Aging 2: 111-116.
14. Dai P, Hu P, Tang J, Li Y, Li C (2016) Effect of glyphosate on reproductive organs in male rat. Acta Histochem 118: 519-526.
15. de La Iglesia R, Milagro FI, Campión J, Boqué N, Martínez JA (2010) Healthy properties of proanthocyanidins. Biofactors 36: 159-168.
16. El-Beltagi EM, Elwan WM, El-Bakry NA, Salah EF (2017) Histological and immunohistochemical study on the effect of gibberellic acid on the seminiferous tubules of testis of adult albino rat and the possible protective role of grape seeds proanthocyanidin extract. Tanta Med J 45: 79.
17. Fine AM (2000) Oligomeric proanthocyanidin complexes: history, structure, and phytopharmaceutical applications. Altern Med Rev: 5: 144-151.
18. Hajizadeh Z, Mehranjani MS, Najafi G, Shariatzadeh SM, Jalali AS (2016) Black grape seed extract modulates fluoxetine-induced oxida-tive stress and cytotoxicity in the mouse testis. Jundishapur J Nat Pharm Prod 11: e27512.
19. He L, Li P, Yu LH, Li L, Zhang Y, Guo Y, Long M, He JB, Yang SH (2018) Protective effects of proanthocyanidins against cadmi-um-induced testicular injury through the modification of Nrf2-Keap1 signal path in rats. Environ Toxicol Pharmacol 57: 1-8.
20. Ikpeme EV, Udensi O, Ekaluo UB, Solomon TO (2012) Efficacy of ascorbic acid in reducing glyphosate-ınduced toxicity in rats. Br Bi-otechnol J 2: 157-168.
21. Joshi SS, Kuszynski CA, Bagchi D (2001) The cellular and molecular basis of health benefits of grape seed proanthocyanidin extract. Curr Pharm Biotechnol 2: 187-200.
22. Kumaravel TS, Jha AN (2006) Reliable Comet assay measurements for detecting DNA damage induced by ionising radiation and chem-icals. Mutat Res 605: 7-16.
23. Long M, Yang S, Zhang Y, Li P, Han J, Dong S, Chen X, He J (2017) Proanthocyanidin protects against acute zearalenone-induced tes-ticular oxidative damage in male mice. Environ Sci Pollut Res Int 24: 938-946.
24. Lopes FM, Varela Junior AS, Corcini CD, da Silva AC, Guazzelli VG, Tavares G, da Rosa CE (2014) Effect of glyphosate on the sperm quality of zebrafish Danio rerio. Aquat Toxicol 155: 322-326.
25. Nithya R, Elango V (2015) Pesticide effect in male hormones and antioxidant status in male albino rats. J Acad Indus Res 4: 140-143.
26. Ohkawa H, Ohishi N, Yagi K (1979) Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 95: 351-358.
27. Ou K, Gu L (2014) Absorption and metabolism of proanthocyanidins. J Funct Food 7: 43-53.
28. Owagboriaye FO, Dedeke GA, Ademolu KO, Olujimi OO, Ashidi JS, Adeyinka AA (2017) Reproductive toxicity of Roundup herbi-cide exposure in male albino rat. Exp Toxicol Pathol 69: 461-468.
29. Poletta GL, Larriera A, Kleinsorge E, Mudry MD (2009) Genotoxicity of the herbicide formulation Roundup (glyphosate) in broad-snouted caiman (Caiman latirostris) evidenced by the Comet assay and the Micronucleus test. Mutat Res 672: 95-102.
30. Richmond ME (2018) Glyphosate: a review of its global use, environmental impact, and potential health effects on humans and other species. J Environ Stud Sci 8: 416-434.
31. Rodríguez-Pérez C, García-Villanova B, Guerra-Hernández E, Verardo V (2019) Grape seeds proanthocyanidins: an overview of ın vivo bioactivity in animal models. Nutrients 11: 2435
32. Romano RM, Romano MA, Bernardi MM, Furtado PV, Oliveira CA (2010) Prepubertal exposure to commercial formulation of the herbicide glyphosate alters testosterone levels and testicular morphology. Arch Toxicol 84: 309-317.
33. Soleimani M, Masoumi N (2017) The effect of grape seed extract on semen oxidative stress markers in men with idiopathic infertility: a cross-sectional before-after study. Nephro-Urol Mont 9: e13837.
34. Sönmez MF, Tascioglu S (2016) Protective effects of grape seed extract on cadmium-induced testicular damage, apoptosis, and endothe-lial nitric oxide synthases expression in rats. Toxicol Ind Health 32: 1486-1494.
35. Su L, Deng Y, Zhang Y, Li C, Zhang R, Sun Y, Zhang K, Li J, Yao S (2011) Protective effects of grape seed procyanidin extract against nickel sulfate-induced apoptosis and oxidative stress in rat testes. Toxicol Mech Methods 21: 487-494.
36. Wang EH, Yu ZL, Bu YJ, Xu PW, Xi JY, Liang HY (2019) Grape seed proanthocyanidin extract alleviates high-fat diet induced testicu-lar toxicity in rats. RSC Adv 9: 11842-11850.
37. Watson PF (1975) Use of a Giemsa stain to detect changes in acrosomes of frozen ram spermatozoa. Vet Rec 97: 12-15.
38. Yousef MI, Salem MH, Ibrahim HZ, Helmi S, Seehy MA, Bertheussen K (1995) Toxic effects of carbofuran and glyphosate on semen characteristics in rabbits. J Environ Sci Health B 30: 513-534.
39. Zhao YM, Gao LP, Zhang HL, Guo JX, Guo PP (2014) Grape seed proanthocyanidin extract prevents DDP-induced testicular toxicity in rats. Food Funct 5: 605-611.
Go to article

Authors and Affiliations

F. Avdatek
1
M. Kirikkulak
1
D. Yeni
1

  1. Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, University of Afyon Kocatepe, 03030, Afyonkarahisar, Turkey
Download PDF Download RIS Download Bibtex

Abstract

Therefore, the aim of the present study was to evaluate the possible effect of bilberry fruit (Vaccinium myrtillus L.) supplement in a daily diet on the cognitive behaviour of the rats and the expression of paravalbumin (PV) in populations of hippocampal neurons. It has been postulated that the antioxidants present in bilberry fruit may act as neuroprotective factors playing also a significant role as memory enhancements. Forty Wistar rats with a similar average body weight (460 ± 0.4 g) were divided into four groups (n=10 per group). The control group received standard feed (210 g/week), whereas animals of experimental groups received standard feed supplemented with bilberry (per os) at consumed doses of 2 g (group I), 5 g (group II), and 10 g/kg b.w./ /day (group III). After three months of feeding with bilberry, the modified elevated plus-maze test (mEPM) was performed. After 32 weeks of feeding, brains were collected and PV-immunoreactive (ir) neurons were immunohistochemically visualized. In the modified elevated plus-maze test, transfer latency examined 2 h and 24 h after the acquisition session was significantly shorter (p<0.05) in the group II in comparison with the control group. In CA1 and CA2/CA3 hippocampal fields as well as dentate gyrus of all experimental groups, a significant (p<0.05) decrease in number of PV-ir neurons were found. In relation to the control group, the mean subpopulation of PV-ir neurons found in groups II and III were significantly reduced. The subpopulations of PV-ir neurons found in DG of all experimental groups were significantly reduced in comparison to the control. In conclusion the in the present paper we demonstrated a relationship between the diet rich in a bilberry fruit and process of memory as well as numbers of calcium- binding protein-expressing hippocampal neurons. Our results may be source of basic knowledge for further research aiming at neuroprotective role of the bilberry fruit.

Go to article

Authors and Affiliations

K. Borowiec
M. Matysek
D. Szwajgier
G. Biała
M. Kruk-Słomka
R. Szalak
J. Ziętek
M.B. Arciszewski
Z. Targoński
Download PDF Download RIS Download Bibtex

Abstract

Arsenic is an important metalloid that can cause poisoning in humans and domestic animals. Exposure to arsenic causes cell damage, increasing the production of reactive oxygen species. Chitosan is a biopolymer obtained by deacetylation of chitin with antioxidant and metal ion chelating properties. In this study, the protective effect of chitosan on arsenic-induced nephrotoxicity and oxidative damage was investigated. 32 male Wistar-albino rats were divided into 4 groups of 8 rats each as control group (C), chitosan group (CS group), arsenic group (AS group), and arsenic+chitosan group (AS+CS group). The C group was given distilled water by oral gavage, the AS group was given 100 ppm/day Na-arsenite ad libitum with drinking water, the CS group was given 200 mg/kg/day chitosan dissolved in saline by oral gavage, the AS+CS group was given 100 ppm/day Na-arsenite ad libitum with drinking water and 200 mg/kg/day chitosan dissolved in saline by oral gavage for 30 days. At the end of the 30-day experimental period, 90 mg/kg ketamine was administered intraperitoneally to all rats, and blood samples and kidney tissues were collected. Urea, uric acid, creatinine, P, Mg, K, Ca, Na, Cystatin C (CYS-C), Neutrophil Gelatinase Associated Lipocalin (NGAL) and Kidney Injury Molecule 1 (KIM-1) levels were measured in serum samples. Malondialdehyde (MDA), Glutathione (GSH), Catalase (CAT) and Superoxide dismutase (SOD) levels in the supernatant obtained from kidney tissue were analyzed by ELISA method. Compared with AS group, uric acid and creatinine levels of the AS+CS group were significantly decreased (p<0.001), urea, KIM-1, CYS-C, NGAL, and MDA levels were numerically decreased and CAT, GSH, and SOD levels were numerically increased (p>0.05). In conclusion, based on both biochemical and histopathological-immunohistochemical- immunofluorescence findings, it can be concluded that chitosan attenuates kidney injury and protects the kidney.
Go to article

Bibliography

1. Aboulthana WM, Ibrahim NE (2018) A renoprotective role of chitosan against lithium-induced renal toxicity in rats. Bull Natl Res Cent 42: 1-11.
2. Akao Y, Yamada H, Nakagawa Y (2000) Arsenic-induced apoptosis in malignant cells in vitro. Leuk Lymphoma 37: 53-63.
3. Aleksunes LM, Augustine LM, Scheffer GL, Cherrington NJ, Manautou JE (2008) Renal xenobiotic transporters are differentially ex-pressed in mice following cisplatin treatment. Toxicology 250: 82-88.
4. Aras S, Gerin F, Aydin B, Ustunsoy S, Sener U, Turan BC, Armutcu F (2015) Effects of sodium arsenite on the some laboratory signs and therapeutic role of thymoquinone in the rats. Eur Rev Med Pharmacol Sci 19: 658-663.
5. Bagga A, Bajpai A, Menon S (2005) Approach to renal tubular disorders. Indian J Pediatr 72: 771-776.
6. Bakan M (2019) Arsenic Metabolism and Arsenolipid Biosynthesis. MedFar 2: 83-88.
7. Büget Mİ, Özkilitçi E, Küçükgergin C, Seçkin Ş, Küçükay S, Yenigün Y, Orhun G, Akıncı İÖ, Özcan PE (2014) Early Diagnosis in Acute Kidney Failure: Neutrophil Gelatinase Associated Lipocain (NGAL), Kidney Injury Molecule-1 (KIM-1), Interleukine-18 (IL-18), Cystatin C. J Turk Soc Intens Care 12: 94-100.
8. Cai X, Yu Y, Huang Y, Zhang L, Jia PM, Zhao Q, Chen Z, Tong JH, Dai W, Chen GQ (2003) Arsenic trioxide-induced mitotic arrest and apoptosis in acute promyelocytic leukemia cells. Leukemia 17: 1333-1337.
9. Chou CK, Li YC, Chen SM, Shih YM, Lee JA (2015) Chitosan prevents gentamicin-induced nephrotoxicity via a carbonyl stress-dependent pathway. Biomed Res Int 2015: 675714.
10. Danışman B, Çiçek B, Yıldırım S, Yüce N, Bolat I (2023) Gastroprotective effects of bromelain on indomethacin-induced gastric ulcer in rats. GSC Biological and Pharmaceutical Sciences 23: 277-286.
11. Demir A, Seventekin N (2009) Chitin, Chitosan And General Application Areas. TTED 3: 92-103.
12. Devarajan P (2010) Review: neutrophil gelatinase-associated lipocalin: a troponin-like biomarker for human acute kidney injury. Neph-rology (Carlton) 15: 419-428.
13. Dhondup T, Qian Q (2017) Electrolyte and acid-base disorders in chronic kidney disease and end-stage kidney failure. Blood Purif 43: 179-188.
14. Dieterle F, Perentes E, Cordier A, Roth DR, Verdes P, Grenet O, Pantano S, Moulin P, Wahl D, Mahl A, End P, Staedtler F, Legay F, Carl K, Laurie D, Chibout SD, Vonderscher J, Maurer G (2010) Urinary clusterin, cystatin C, beta2-microglobulin and total protein as markers to detect drug-induced kidney injury. Nat Biotechnol 28: 463-U114.
15. Duker AA, Carranza EJ, Hale M (2005) Arsenic geochemistry and health. Environ Int 31: 631-641.
16. Elieh-Ali-Komi D, Hamblin MR (2016) Chitin and chitosan: production and application of versatile biomedical nanomaterials. Int J Adv Res (Indore) 4: 411–427.
17. Ewere EG, Okolie NP, Eze GI, Jegede DA (2019) Irvingia gabonensis leaves mitigate arsenic-induced renal toxicity in Wistar rats. Asian J Biomed Pharmaceut Sci 9: 17-25.
18. Flora SJ (2011) Arsenic-induced oxidative stress and its reversibility. Free Radic Biol Med 51: 257-281.
19. Florea AM, Yamoah EN, Dopp E (2005) Intracellular calcium disturbances induced by arsenic and its methylated derivatives in relation to genomic damage and apoptosis induction. Environ Health Perspect 113: 659-664.
20. Galanis A, Karapetsas A, Sandaltzopoulos R (2009) Metal-induced carcinogenesis, oxidative stress and hypoxia signalling. Mutat Res 674: 31-35.
21. Ghys L, Paepe D, Smets P, Lefebvre H, Delanghe J, Daminet S (2014) Cystatin C: a new renal marker and its potential use in small ani-mal medicine. J Vet Intern Med 28: 1152-1164.
22. Hughes MF (2002) Arsenic toxicity and potential mechanisms of action. Toxicol Lett 133: 1-16.
23. Ibaokurgil F, Aydin H, Yildirim S, Sengul E (2023) Melatonin alleviates oxidative stress, inflammation, apoptosis, and DNA damage in acrylamide–induced nephrotoxicity in rats. Asian Pac J Trop Biomed 13: 121-130.
24. Ishaq A, Gulzar H, Hassan A, Kamran M, Riaz M, Parveen A, Chattha MS, Walayat N, Fatima S, Afzal S, Fahad S (2021) Ameliorative mechanisms of turmeric-extracted curcumin on arsenic (As)-induced biochemical alterations, oxidative damage, and impaired organ func-tions in rats. Environ Sci Pollut Res Int 28: 66313-66326.
25. Jeon TI, Hwang SG, Park NG, Jung YR, Shin SI, Choi SD, Park DK (2003) Antioxidative effect of chitosan on chronic carbon tetra-chloride induced hepatic injury in rats. Toxicology 187: 67-73.
26. Jia G, Sone H, Nishimura N, Satoh M, Tohyama C (2004) Metallothionein (I/II) suppresses genotoxicity caused by dimethylarsinic acid. Int J Oncol 25: 325-333.
27. Jomova K, Valko M (2011) Advances in metal-induced oxidative stress and human disease. Toxicology 283: 65-87.
28. Kamran M, Malik Z, Parveen A, Huang L, Riaz M, Bashir S, Mustafa A, Abbasi GH, Xue B, Ali U (2020) Ameliorative effects of bio-char on rapeseed (Brassica napus L.) growth and heavy metal immobilization in soil irrigated with untreated wastewater. J Plant Growth Regul 39: 266-281.
29. Kaya Z, Eraslan G (2013) The effects of evening primrose oil on arsenic-induced oxidative stress in rats. Toxicol Environ Chem 95: 1416-1423.
30. Kitchin KT, Conolly R (2010) Arsenic-induced carcinogenesis oxidative stress as a possible mode of action and future research needs for more biologically based risk assessment. Chem Res Toxicol 23: 327-335.
31. Liu J, Waalkes MP (2008) Liver is a target of arsenic carcinogenesis. Toxicol Sci 105: 24-32.
32. Liu YC, Huang H (1997) Involvement of calcium-dependent protein kinase C in arsenite-induced genotoxicity in chinese hamster ovary cells. J Cell Biochem 64: 423-433.
33. Mehrzadi S, Fatemi I, Malayeri AR, Khodadadi A, Mohammadi F, Mansouri E, Rashno M, Goudarzi M (2018) Ellagic acid mitigates sodium arsenite-induced renal and hepatic toxicity in male Wistar rats. Pharmacol Rep 70: 712-719.
34. Mussap M, Plebani M (2004) Biochemistry and clinical role of human cystatin C. Crit Rev Clin Lab Sci 41: 467-550.
35. Muzaffar S, Khan J, Srivastava R, Gorbatyuk MS, Athar M (2023) Mechanistic understanding of the toxic effects of arsenic and warfare arsenicals on human health and environment. Cell Biol Toxicol 39: 85-110.
36. Namgung U, Xia Z (2001) Arsenic induces apoptosis in rat cerebellar neurons via activation of JNK3 and p38 MAP kinases. Toxicol Appl Pharmacol 174: 130-138.
37. Narayana KR, Reddy MS, Chaluvadi MR, Krishna DR (2001) Bioflavonoids classification, pharmacological, biochemical effects and therapeutic potential. Indian J Pharmacol 33: 2-16.
38. Noman AS, Dilruba S, Mohanto NC, Rahman L, Khatun Z, Riad W, Al Mamun A, Alam S, Aktar S, Chowdhury S, Saud ZA, Rahman Z, Hossain K, Haque A (2015) Arsenic-induced histological alterations in various organs of mice. J Cytol Histol 6: 323.
39. Nomier YA, Alshahrani S, Elsabahy M, Asaad GF, Hassan A, El-Dakroury WA (2022) Ameliorative effect of chitosan nanoparticles against carbon tetrachloride-induced nephrotoxicity in Wistar rats. Pharm Biol 60: 2134-2144.
40. Nozohour Y, Jalilzadeh-Amin G (2019) Histopathological changes and antioxidant enzymes status in oxidative stress induction using Sodium arsenite in rats. J Appl Biotechnol Rep 6: 40-44.
41. Özdek U, Toz H, Kömüroğlu AU, Mis L, Huyut Z, Değer Y (2019) Protective Effect of Chitosan Against Lead-Induced Oxidative Stress in Rat Kidney. Van Vet J 30: 187-191.
42. Patel HV, Kalia K (2010) Sub-chronic arsenic exposure aggravates nephrotoxicity in experimental diabetic rats. Indian J Exp Biol 48: 762-768.
43. Peters BA, Hall MN, Liu X, Neugut YD, Pilsner JR, Levy D, Ilievski V, Slavkovich V, Islam T, Factor-Litvak P, Graziano JH, Gamble MV (2014) Creatinine, arsenic metabolism, and renal function in an arsenic-exposed population in Bangladesh. PLoS One 9: e113760.
44. Poręba R, Gać P, Poręba M, Antonowicz-Juchniewicz J, Andrzejak R (2011) Relation between occupational exposure to lead, cadmium, arsenic and concentration of cystatin C. Toxicology 283: 88-95.
45. Robles-Osorio ML, Sabath-Silva E, Sabath E (2015) Arsenic-mediated nephrotoxicity. Ren Fail 37: 542-547.
46. Roy M, Roy S (2011) Ameliorative potential of Psidium guajava in induced arsenic toxicity in Wistar rats. Vet World 4: 82-83.
47. Selby LA, Case AA, Osweiler GD, Hayes HM Jr (1977) Epidemiology and toxicology of arsenic poisoning in domestic ani-mals. Environ Health Perspect 19: 183-189.
48. Sharma S, Kaur T, Sharma AK, Singh B, Pathak D, Yadav HN, Singh AP (2022) Betaine attenuates sodium arsenite-induced renal dys-function in rats. Drug Chem Toxicol 45: 2488-2495.
49. Shen ZY, Shen WY, Chen MH, Shen J, Cai WJ, Yi Z (2002) Nitric oxide and calcium ions in apoptotic esophageal carcinoma cells in-duced by arsenite. World J Gastroenterol 8: 40-43.
50. Shen ZY, Shen J, Cai WJ, Hong CQ, Zheng MH (2000) The alteration of mitochondria is an early event of arsenic trioxide induced apoptosis in esophageal carcinoma cells. Int J Mol Med 5: 155-158.
51. Singh MK, Yadav SS, Yadav RS, Singh US, Shukla Y, Pant KK, Khattri S (2014) Efficacy of crude extract of Emblica officinalis (amla) in arsenic-induced oxidative damage and apoptosis in splenocytes of mice. Toxicol Int 21: 8-17.
52. Sinha M, Manna P, Sil PC (2008) Arjunolic acid attenuates arsenic-induced nephrotoxicity. Pathophysiology 15: 147-156.
53. Smith DL, Harris AD, Johnson JA, Silbergeld EK, Morris JG Jr (2002) Animal antibiotic use has an early but important impact on the emergence of antibiotic resistance in human commensal bacteria. Proc Natl Acad Sci USA 99: 6434-6439.
54. Tilako BH, Ogbodo SO, Okonkwo IN, Nubila IN, Shuneba IL, Ogbonna E, Odoma S, Gali RM, Bassey BE, Shu EN (2020) Distribu-tion and interactions of priority heavy metals with some antioxidant micronutrients in inhabitants of a lead-zinc mining community of eb-onyi state, Nigeria. Adv Toxicol Toxic Effects 4: 011-017.
55. Toz H, Değer Y (2018) The Effect of Chitosan on the Erythrocyte Antioxidant Potential of Lead Toxicity-Induced Rats. Biol Trace Elem Res 184:114-118
56. Wang Z, Yan Y, Yu X, Li W, Li B, Qin C (2016) Protective effects of chitosan and its water-soluble derivatives against lead-induced oxidative stress in mice. Int J Biol Macromol 83: 442-449.
57. Wan Muhamad Salahudin WS, Norlelawati AT, Nor ZA, Aung S, Asmah HH, Zunariah B (2021) Histopathological changes in chronic low dose organic arsenic exposure in rats kidney. IIUM Med J Malays 20: 91-98.
58. Wu KY, Wu M, Fu ML, Li H, Yang Y, Zhang H, Cheng C, Wang ZZ, Wang XY, Lu XB, Liu DG, Li H, Gao R (2006) A novel chi-tosan CpG nanoparticle regulates cellular and humoral immunity of mice. Biomed Environ Sci 19: 87-95.
59. Yuan WP, Liu B, Liu CH, Wang XJ, Zhang MS, Meng XM, Xia XK (2009) Antioxidant activity of chito-oligosaccharides on pancreatic islet cells in streptozotocin-induced diabetes in rats. World J Gastroenterol 15: 1339.
60. Zalups RK (1997) Reductions in renal mass and the nephropathy induced by mercury. Toxicol Appl Pharmacol 143: 366-379.
61. Zeng L, Qin C, Wang W, Chi W, Li W (2008) Absorption and distribution of chitosan in mice after oral administra-tion. Carbohydr Polym 71: 435-440.
62. Zhang Z, Lu B, Sheng X, Jin N (2011) Cystatin C in prediction of acute kidney injury: a systemic review and meta-analysis. Am J Kid-ney Dis 58: 356-365.
63. Zheng L, Kuo CC, Fadrowski J, Agnew J, Weaver VM, Navas-Acien A (2014) Arsenic and chronic kidney disease: a systematic re-view. Curr Environ Health Rep 1: 192-207.

Go to article

Authors and Affiliations

K. İrak
1
Ö.Y. Çelik
2
M. Bolacalı
3
T. Tufan
4
S. Özcan
4
S. Yıldırım
5
İ. Bolat
5

  1. Department of Biochemistry, Faculty of Veterinary Medicine, Siirt University, Siirt, Turkey
  2. Department of Internal Medicine, Faculty of Veterinary Medicine, Siirt University, Siirt, Turkey
  3. Kırsehir Ahi Evran University, Faculty of Medicine, Department of Biostatistics and Medical Informatics, Kirsehir, Turkey
  4. Department of Animal Nutrition and Nutritional Disease, Faculty of Veterinary Medicine, Siirt University, Siirt, Turkey
  5. Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, Turkey
Download PDF Download RIS Download Bibtex

Abstract

Background: The histology of the lung includes a variety of cell types. Fibrosis is a universal process, occurring in the skin, intestine, heart, muscles, kidney, blood vessels, liver, and also the lungs. Telocytes are a type of cells with a wide range of properties, which were previously described in healthy and disease-affected organs of human and animal organisms.

Aim: This study aimed to identify telocytes in the lungs of rats and discuss their possible role in the development of pulmonary fibrosis.

Methods: Tissue samples were taken from a group of ten male Wistar rats. Further histological and immunohistochemical analysis was performed. Double immunolabeling for c-kit, vimentin, CD34, and PDGFRα has revealed telocytes in the lungs.

Results: In all tissue samples, telocytes have been identified (in the area of interalveolar septa, close to blood vessels, and between the airway epithelium).

Conclusion: Telocytes might be directly and indirectly (through contact with stem cells, secretomes, and reduction in number) involved in the development of pulmonary fibrosis. The heterogeneity of the telocyte population in different pathologies and their subtypes, as well as their tendency to be common stress their important role in pathological physiology.
Go to article

Bibliography

1. Schraufnagel D.E.: Electron microscopy of the lung. New York: Marcel Dekker, CRC Press; 1990.
2. Hogan B., Tata P.R.: Cellular organization and biology of the respiratory system. Nat Cell Biol. 2019. https://doi.org/10.1038/s41556-019-0357-7.
3. Evren E., Ringqvist E., Willinger T.: Origin and ontogeny of lung macrophages: from mice to humans. Immunology. 2020; 160 (2): 126–138.
4. Awad M., Gaber W., Ibrahim D.: Onset of Appearance and Potential Significance of Telocytes in the Developing Fetal Lung. Microsc Microanal. 2019; 25 (5): 1246–1256.
5. Popescu L.M., Gherghiceanu M., Suciu L.C., Manole C.G., Hinescu M.E.: Telocytes and putative stem cells in the lungs: electron microscopy, electron tomography and laser scanning microscopy. Cell Tissue Res. 2011; 345 (3): 391–403.
6. Aleksandrovych V., Pasternak A., Basta P., Sajewicz M., Walocha J.A., Gil K.: Telocytes: facts, speculations and myths (Review article). Folia Med Cracov. 2017; 57 (1): 5–22.
7. Zheng Y., Li H., Manole C.G., Sun A., Ge J., Wang X.: Telocytes in trachea and lungs. J Cell Mol Med. 2011; 15: 2262–2268.
8. Aleksandrovych V., Walocha J.A., Gil K.: Telocytes in female reproductive system (human and animal). J Cell Mol Med. 2016; 20 (6): 994–1000.
9. Díaz-Flores L., Gutiérrez R., Díaz-Flores L.J.R., Goméz M.G., Sáez F.J., Madrid J.F.: Behaviour of telocytes during physiopathological activation. Semin Cell Dev Biol. 2016; 55: 50–61.
10. Hussein M.M., Mokhtar D.M.: The roles of telocytes in lung development and angiogenesis: An immunohistochemical, ultrastructural, scanning electron microscopy and morphometrical study. Dev Biol. 2018; 443 (2): 137–152.
11. Popescu L.M., Faussone-Pellegrini M.S.: TELOCYTES — a case of serendipity: the winding way from interstitial cells of Cajal (ICC), via interstitial Cajal-like cells (ICLC) to TELOCYTES. J Cell Mol Med. 2010; 14: 729–740.
12. Ibba-Manneschi L., Rosa I., Manetti M.: Telocyte implications in human pathology: An overview. Biol. 2016; 55: 62–69.
13. Liao Z., Chen Y., Duan C., Zhu K., Huang R., Zhao H., et al.: Cardiac telocytes inhibit cardiac microvascular endothelial cell apoptosis through exosomal miRNA-21-5p-targeted cdip1 silencing to improve angiogenesis following myocardial infarction. Theranostics. 2021; 11 (1): 268–291.
14. Zhaofu L., Dongqing C.: Cardiac Telocytes in Regeneration of Myocardium After Myocardial Infarction. Adv Exp Med Biol. 2016; 913: 229–239.
15. Milia A.F., Ruffo M., Manetti M., Rosa I., Conte D., Fazi M., et al.: Telocytes in Crohn’s disease. Cell Mol Med. 2013; 17 (12): 1525–1536.
16. Ibba-Manneschi L., Rosa I., Manetti M.: Telocytes in Chronic Inflammatory and Fibrotic Diseases. Adv Exp Med Biol. 2016; 913: 51–76.
17. Wolnicki M., Aleksandrovych V., Gil A., Pasternak A., Gil K.: Relation between ureteral telocytes and the hydronephrosis development in children. Folia Med Cracov. 2019; 59 (3): 31–44.
18. Aleksandrovych V., Pasternak A., Gil K.: Telocytes in the architecture of uterine fibroids. Folia Med Cracov. 2019; 59 (4): 33–44.
19. Aleksandrovych V., Białas M., Pasternak A., Bereza T., Sajewicz M., Walocha J., et al.: Identification of uterine telocytes and their architecture in leiomyoma. Folia Med Cracov. 2018; 58 (3): 89–102.
20. Manole C.G., Gherghiceanu M., Simionescu O.J.: Telocyte dynamics in psoriasis. Cell Mol Med. 2015; 19 (7): 1504–1519.
21. Jin L., Wang Z., Qi X.: Diffuse idiopathic pulmonary neuroendocrine cell hyperplasia: Case series and a review of the literature. Medicine (Baltimore). 2018; 97 (52): e13806.
22. Leuenberger P.: [Clinical importance of non-respiratory functions of the lung]. Schweiz Med Wochenschr. 1983; 113 (29): 1006–1010.
23. Wittmann J.: [Respiratory and non-respiratory functions of the lung]. Tierarztl Prax Suppl. 1987; 2: 33–36.
24. Aleksandrovych V., Kurnik-Łucka M., Bereza T., Białas M., Pasternak A., Cretoiu D., Walocha J.A., Gil K.: The Autonomic Innervation and Uterine Telocyte Interplay in Leiomyoma Formation. Cell Transplant. 2019; 28: 619–629.
25. Song D., Cretoiu D., Cretoiu S.M., Wang X.: Telocytes and lung disease. Histol Histopathol. 2016; 31 (12): 1303–1314.
26. Shi L., Dong N., Chen C., Wang X.: Potential roles of telocytes in lung diseases. Semin Cell Dev Biol. 2016; 55: 31–39.
27. Ciechanowicz A.: Stem Cells in Lungs. Adv Exp Med Biol. 2019; 1201: 261–274.
28. Rokicki W., Rokicki M., Wojtacha J., Dżeljijli A.: The role and importance of club cells (Clara cells) in the pathogenesis of some respiratory diseases. Kardiochir Torakochirurgia Pol. 2016; 13 (1): 26–30.
29. Ibba-Manneschi L., Rosa I., Manetti M.: Telocytes in Chronic Inflammatory and Fibrotic Diseases. Adv Exp Med Biol. 2016; 913: 51–76.
Go to article

Authors and Affiliations

Anna Gil
1
Veronika Aleksandrovych
1

  1. Department of Pathophysiology, Jagiellonian University Medical College, Kraków, Poland
Download PDF Download RIS Download Bibtex

Abstract

Rat robots have great potential in rescue and search tasks because of their excellent motion ability. However, most of the current rat-robot systems relay on human guidance due to variable voluntary motor behaviour of rats, which limits their application. In this study, we developed a real-time system to detect a rat robot’s transient motion states, as the prerequisite for further study of automatic navigation. We built the detection model by using a wearable inertial sensor to capture acceleration and angular velocity data during the control of a rat robot. Various machine learning algorithms, including Decision Trees, Random Forests, Logistic Regression, and SupportVector Machines,were employed to performthe classification of motion states. This detection system was tested in manual navigation experiments, with detection accuracy achieving 96.70%. The sequence of transient motion states could be further used as a promising reference for offline behaviour analysis.
Go to article

Authors and Affiliations

Yuxin Chen
1
Haoze Xu
2 3
Wei Yang
1 4
Canjun Yang
1 4
Kedi Xu
2 5

  1. Zhejiang University, State Key Laboratory of Fluid Power and Mechatronic Systems, Hangzhou, China
  2. Zhejiang University, Qiushi Academy for Advanced Studies (QAAS), Hangzhou, China
  3. Zhejiang University, Key Laboratory of Biomedical Engineering of Education Ministry, Hangzhou, China
  4. Zhejiang University, Ningbo Research Institute, Ningbo, China
  5. Zhejiang Lab, Hangzhou, China
Download PDF Download RIS Download Bibtex

Abstract

Applications of cold atmospheric plasma/nitric oxide (CAP/NO) gas have recently garnered popularity when treating impaired wound healing in patients with diabetes. In this study, we aimed to investigate the effects of NO gas application for 60 and 120 s on wound healing in diabetic rats. A dorsal excision 3 cm in diameter was performed in 15 diabetic rats; these rats were categorized into the following 3 groups: DC (untreated diabetic control); DNO/60 (exposure to 200 ppm NO gas for 60 s/day); and DNO/120 (exposure to 200 ppm NO gas for 120 s/day). Wound contraction on days 0, 3, 7, 11, and 14 and wound contraction rate between days 0 and 14 were evaluated. On day 14, tissue samples were collected for histopathologic assessment of inflammation, epithelial regeneration, angiogenesis congestion, and collagen fiber organization. Normality of distribution was assessed using the Shapiro-Wilk test, and intergroup comparisons were performed using the Mann-Whitney U test (NPar Test) and the Kruskal-Wallis test (non-parametric ANOVA). Wound contraction during treatment days 7-14 was significantly greater in the NO-treatment groups than in the DC group (p<0.05). The NO60 s and NO120 s groups showed a significantly higher wound contraction rate than the DC group (p=0.033, p=0.049, respectively). Significant differences were noted between the control and NO groups in terms of inflammation (p<0.05) and between the control group and DNO/60 and DNO/120 groups in terms of collagen organization (p<0.05, p<0.01, respectively). Evaluation of epithelialization revealed significant intergroup differences between the control and NO treatment groups (p<0.01). In this study, the application of NO once a day for 60 seconds and 120 seconds in diabetic wounds contributed equally to wound healing.
Go to article

Bibliography

  1. Ahmed R, Augustine R, Chaudhry M, Akhtar UA, Zahid AA, Tariq M, Falahati M, Ahmad IS, Hasan A (2022) Nitric oxide-releasing biomateri-als for promoting wound healing in impaired diabetic wounds: State of the art and recent trends. Biomed Pharmacother 149: 112707.
  2. Anuk T, Öztürk S, Özaydın İ, Kahramanca Ş, Yayla S, Aksoy, Demirkan I (2016) Comparison of Three Fixation Methods for the prevention of wound contractions in diabetic and non-diabetic mice with full-thickness skin excision. Kafkas Univ Vet Fak Derg 22: 647-651.
  3. Bae SH, Bae YC, Nam SB, Choi SJ (2012) A skin fixation method for decreasing the influence of wound contraction on wound healing in a rat model. Arch Plast Surg 39: 457-462.
  4. Bryan NS (2015) Nitric oxide enhancement strategies. Future Sci OA 1: FSO48.
  5. Burgess JL, Wyant WA, Abujamra B, Kirsner RS, Jozic I (2021) Diabetic wound-healing science. Medicina (Kaunas) 57: 1072.
  6. Caskey RC, Liechty KW (2013) Novel animal models for tracking the fate and contributions of bone marrow derived cells in diabetic healing. Methods: 99-115.
  7. Förstermann U, Sessa WC (2012) Nitric oxide synthases: Regulation and function. Eur Heart J 33: 829-837
  8. Fridman G, Friedman G, Gutsol A, Shekhter AB, Vasilets VN, Fridman A (2008) Applied Plasma Medicine. Plasma Process Polym 5: 503-533.
  9. Ghaffari A, Jalili R, Ghaffari M, Miller C, Ghahary A (2007) Efficacy of gaseous nitric oxide in the treatment of skin and soft tissue infec-tions. Wound Repair Regen 15: 368-377.
  10. Ghaffari A, Miller CC, McMullin B, Ghahary A (2006) Potential application of gaseous nitric oxide as a topical antimicrobial agent. Nitric Ox-ide 14: 21-29.
  11. Ghaffari A, Neil DH, Ardakani A, Road J, Ghahary A, Miller CC (2005) A direct nitric oxide gas delivery system for bacterial and mammalian cell cultures. Nitric Oxide 12: 129-140.
  12. Gronbach M, Mitrach F, Lidzba V, Müller B, Möller S, Rother S, Schulz-Siegmund M (2020) Scavenging of Dickkopf-1 by macromer-based biomaterials covalently decorated with sulfated hyaluronan displays pro-osteogenic effects. Acta Biomater 114: 76-89.
  13. Güngör GÇ, Gültekin Ç, Kükner A, Etikan İ, Temizel M, Özgencil FE (2022) Effect of topical insulin and ozonized cream for the treatment of full-thickness dermal burn injuries: A clinical and histopathological study in diabetic rats. Pak Vet J 42: 229-235.
  14. Krausz A, Friedman AJ (2015) Nitric oxide as a surgical adjuvant. Future Sci OA 1: FSO56.
  15. Luo JD, Chen AF (2005) Nitric oxide: A newly discovered function on wound healing. Acta Pharmacol Sin 26: 259-264.
  16. Malone-Povolny MJ, Maloney SE, Schoenfisch MH (2019) Nitric oxide therapy for diabetic wound healing. Adv Healthc Mater 8: e1801210.
  17. Grada A, Mervis J, Falanga V (2018) Research techniques made simple: Animal models of wound healing. J Invest Dermatol 138: 2095-2105..
  18. Mieczkowski M, Mrozikiewicz-Rakowska B, Kowara M, Kleibert M, Czupryniak L (2022) The problem of wound healing in diabetes-from mo-lecular pathways to the design of an animal model. Int J Mol Sci 23: 7930.
  19. Miersch S, Espey MG, Chaube R, Akarca A, Tweten R, Ananvoranich S, Mutus B (2008) Plasma membrane cholesterol content affects nitric oxide diffusion dynamics and signaling. J Biol Chem 283: 18513-18521.
  20. Miller CC, Miller MK, Ghaffari A, Kunimoto B (2004) Treatment of chronic nonhealing leg ulceration with gaseous nitric oxide: A case study. J Cutan Med Surg 8: 233-238.
  21. Pekshev AV, Shekhter AB, Vagapov AB, Sharapov NA, Vanin AF (2018) Study of plasma-chemical NO-containing gas flow for treatment of wounds and inflammatory processes. Nitric Oxide 73: 74-80.
  22. Powers JG, Higham C, Broussard K, Phillips TJ (2016) Wound healing and treating wounds: Chronic wound care and management. J Am Acad Dermatol 74: 607-625.
  23. Moeen Rezakhanlou A, Miller C, McMullin B, Ghaffari A, Garcia R, Ghahary A (2011) Gaseous nitric oxide exhibits minimal effect on skin fibroblast extracellular matrix gene expression and immune cell viability. Cell Biol Int 35: 407-415.
  24. Shekhter AB, Pekshev AV, Vagapov AB, Telpukhov VI, Panyushkin PV, Rudenko TG, Fayzullin AL, Sharapov NA, Vanin AF (2019) Physi-cochemical parameters of NO-containing gas flow affect wound healing therapy. An experimental study. Eur J Pharm Sci 128: 193-201.
  25. Shekhter AB, Serezhenkov VA, Rudenko TG, Pekshev AV, Vanin AF (2005) Beneficial effect of gaseous nitric oxide on the healing of skin wounds. Nitric Oxide 12: 210-219.
  26. Wu M, Lu Z, Wu K, Nam C, Zhang L, Guo J (2021) Recent advances in the development of nitric oxide-releasing biomaterials and their application potentials in chronic wound healing. J Mater Chem B 9: 7063-7075.
  27. Yang Y, Qi PK, Yang ZL, Huang N (2015) Nitric oxide based strategies for applications of biomedical devices. Biosurf Biotribol 1: 177-201.
  28. Young LH, Ikeda Y, Lefer AM (2001) Caveolin-1 peptide exerts cardioprotective effects in myocardial ischemia-reperfusion via nitric oxide mechanism. Am J Physiol Heart Circ Physiol 280: H2489-H2495.
Go to article

Authors and Affiliations

G. Tatlıcıoğlu
1
A. Çürükoğlu
1
G. Akan
2
G. Yeşilovalı
1
G. Öğütçü
3
R. Aktaş
1
A. Kükner
3
M. Temizel
4
Z.K. Sarıtaş
5
F.E. Özgencil
1

  1. Surgery Department, Near East University, Faculty of Veterinary Medicine, Yakın Doğu St, VIC: 99138 Nicosia / TRNC Mersin 10 – Turkey
  2. DESAM Institute, Near East University, Yakın Doğu St, VIC: 99138 Nicosia / TRNC Mersin 10 – Turkey
  3. Histology Department, Near East University, Faculty of Medicine, Yakın Doğu St, VIC: 99138 Nicosia / TRNC Mersin 10 – Turkey
  4. Experimental Animal Research Center, Near East University Faculty of Veterinary Medicine, Yakın Doğu St, VIC: 99138 Nicosia / TRNC Mersin 10- Turkey
  5. Surgery Department, Afyon Kocatepe University, Faculty of Veterinary Medicine, ANS Campus, Erenler, Afyonkarahisar/Turkey
Download PDF Download RIS Download Bibtex

Abstract

Postoperative adhesion (POA) is a common and well-known complication with an estimated risk of 50-100%. The antioxidant effect of n-acetyl-cysteine (NAC) can increase intracellular glutathione levels, thereby reducing adhesion. This study was conducted to compare the outcomes of NAC nanoparticles (Nano-NAC) on intra-abdominal adhesion (IAA) after laparotomy in rat. A total of 25 male Wistar rats were randomized into five groups: 50 mg/kg Nano-NAC, 75 mg/kg Nano-NAC, 150 mg/kg Nano-NAC, NAC and control. During the surgical procedure, some sections (2×2cm) were collected through abdominal midline incision to ensure the infliction of peritoneal damage by a standard adhesion. Macroscopic evaluation was performed on the 14th and 28th day and blood samples were collected to evaluate the inflammatory factor (C-reactive protein) on days 0, 14 and 28. According to the serologic results (CRP test), C-reactive protein was at highest level in 150 mg/kg Nano-NAC and control groups and at lowest level in 50 mg/kg Nano-NAC and 75 mg/kg Nano-NAC groups (p<0.001). The macroscopic evaluation results showed that frequency of adhesion bands was significantly lower in 50 mg/kg Nano-NAC group than the control at the intervals. Results showed that the intraperitoneal administration of lower Nano-NAC dosages (50 and 75 mg/kg) had a major role in the management of postoperative inflammation. Nano-NAC administration was proved feasible, safe and effective in reduction of the C-reactive protein level.

Go to article

Authors and Affiliations

Sh. Shahzamani
AR. Jahandideh
GhR. Abedi
A. Akbarzadeh
S. Hesaraki
Download PDF Download RIS Download Bibtex

Abstract

5-fluorouracil (5-FU), which is a commonly used chemotherapy agent exerts undesired cardiac toxicity. Mitochondrial dysfunction is thought to be one of potentially important mechanisms of 5-FU- induced cardiotoxicity. α-ketoglutarate dehydrogenase (α-KGDHC) is the key regulatory enzyme of TCA cycle. The complex consists of multiple copies of three catalytic subunits: α-ketoglutarate dehydrogenase (E1), dihydrolipoamide succinyltransferase (E2) and dihydrolipoamide dehydrogenase (E3). α-KGDHC together with branched chain α-ketoacid dehydrogenase (BCKDHC) and pyruvate dehydrogenase (PDHC), are the members of 2-oxoacid dehydrogenases family that share some structural and functional similarities. Recently, it has been found that 5-FU stimulates BCKDHC in rat’s cardiac muscle. Therefore, we hypothesize that 5-FU modifies α-KGDHC activity and affects cardiac muscle metabolism. The aim of this study was to determine the effect of 5-FU on α-KGDHC activity and protein levels of E1 and E2 subunits of the complex in rat’s cardiac muscle. Wistar male rats were administered with 4 doses of 5-FU, 150 mg/kg b.wt. each (study group) or 0.3% methylcellulose (control group). α-KGDHC activity was assayed spectrophotometrically. The E1 and E2 proteins levels were quantified by Western blot. 5-FU administration resulted in stimulation of myocardial α-KGDHC activity in rats. In addition, E2 protein level increased in response to 5-FU treatment, while the E1 protein level remained unchanged. Up- regulation of α-KGDHC appears to result from change in E2 subunit protein level. However, the effect of 5-FU on factors modifying α-KGDHC activity at post-translational level cannot be excluded.
Go to article

Authors and Affiliations

Małgorzata Knapik-Czajka
1
Anna Gawędzka
1
Michał Jurczyk
2
Jagoda Drąg
1
Małgorzata Belczyk
1
Veronika Aleksandrovych
2
Anna Gil
3
Krzysztof Gil
1

  1. Department of Biochemical Analytics, Jagiellonian University Medical College, Kraków, Poland
  2. Department of Pathophysiology, Jagiellonian University Medical College, Kraków, Poland
  3. Department of Anatomy, Jagiellonian University Medical College, Kraków, Poland
Download PDF Download RIS Download Bibtex

Abstract

This study aimed to determine the effects of dexamethasone and minocycline alone and combined treatment with N-acetylcysteine (NAC) and vitamin E on serum coenzyme Q10 (CoQ10) and matrix metalloproteinase-9 (MMP-9) levels in rats administered aflatoxin B1 (AFB1). The study was carried out on 66 male Wistar rats. Following the intraperitoneal (IP) administration of AFB1 at dose of 2 mg/kg, minocycline (45 and 90 mg/kg, IP) and dexamethasone (5 and 20 mg/kg, IP) were administered alone and combined with NAC (200 mg/kg, IP) and vitamin E (600 mg/kg, IP). CoQ10 and MMP-9 levels were analyzed using the HPLC-UV method and a commercial kit by ELISA, respectively. AFB1 increased MMP-9 level and decreased CoQ10 level compared to the control group. After dexamethasone and minocycline administration, there is no increase in CoQ10 level, which is caused by AFB1. However, dexamethasone and minocycline combined with NAC+vitamin E caused significant increases in CoQ10 levels. Dexamethasone and minocycline alone and combined with NAC+vitamin E decreased MMP-9 levels compared to the single AFB1 treated group. The use of MMPs inhibitors and oxidative stress-reducing agents is anticipated to be beneficial in the poisoning with AFB1.
Go to article

Authors and Affiliations

B. Tras
1
H. Eser Faki
1
Z. Ozdemir Kutahya
2
E. Bahcivan
3
B. Dik
1
K. Uney
1

  1. Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Selcuk, Alaeddin Keykubat Campus, New Istanbul Road. No:371, Konya, 42130, Turkey
  2. Department of Pharmacology and Toxicology, Faculty of Ceyhan Veterinary Medicine, University of Cukurova, Fatih Sultan Mehmet District, Inonu Boulevard, No: 242, Adana, 01330, Turkey
  3. Department of Medical Pharmacology, Faculty of Medicine, University of Amasya, Akbilek District. Dominion Street, National Sovereignty Campus, No:4/3, 05100, Turkey
Download PDF Download RIS Download Bibtex

Abstract

In this investigation, the effects of genistein (GEN) on the expression of steroidogenic genes such as steroidogenic acute regulatory protein (StAR), side-chain cleavage enzymes (P450scc) and cytochrome P450 aromatase (CYP19) were assessed. For this study, forty young female Sprague Dawley (SD) rats at aged 2-3 months (200±20 g) and forty aged female SD rats aged 10-12 months (490±20 g) were selected. Also, based on weight they were divided into a negative control group (NC), three different GEN dose groups, which received GEN of 15, 30, 60 mg/kg, and a positive control group (PC). The experiment lasted 30 days. Concentrations of serum hormones were determined by Enzyme-linked immunosorbent assay (ELISA). Gene and protein expressions of StAR, P450scc and CYP19 were determined by Real-Time PCR and western blot techniques. It was observed that 30-60 mg/kg GEN could increase the expression of androgen generating key enzymes in the young rat ovary. GEN also significantly increased progesterone and E2 levels in the serum of aged rats and reduced the levels of LH and FSH in the serum of both young and aged rats. Compared with young rats, the effect of GEN on the ovary of aged rats was stronger and a lower dose of GEN (15 mg/kg) showed an obvious effect on these indicators. GEN influenced both estrogen level and indicators associated with estrogen and androgen transformation processes, which indicates that GEN can impair the growth and maturation of the ovary.

Go to article

Authors and Affiliations

X.X. Chi
X.L. Chu
T. Zhang
L.K. Cao
Download PDF Download RIS Download Bibtex

Abstract

The purpose of the study was to study the activity of the phytoestrogen genistein (GEN) act- ing on FSHR and LHR in rat ovaries with polycystic ovary syndrome (PCOS). Sixty rats were di- vided into six groups. Rats in the dose group received genistein at a concentration of either 5 (low genistein dose group, L-gen), 10 (middle genistein dose group, M-Gen) or 20 (high genistein dose group, H-Gen) mg per kg of body weight per day. Estrogen group (EG, received 0.5 mg/kg Dieth- ylstilbestrol). Concentration of sex hormones in serum was quantified by enzyme-linked immuno- sorbent assay (ELISA). Expressions of follicle-stimulating hormone receptor (FSHR) and lutein- izing hormone receptor (LHR) protein were determined by immunohistochemistry. Treatment with genistein resulted in a strong stimulation of the concentration of sex hormone in serum. The concentration of progesterone and FSH was significantly higher in H-Gen when compared to the PCOS model control group (MG) (P < 0.01). In contrast, the concentration of testosterone, LH and the ratio of LH/FSH decreased in GEN treatment groups compared to MG, the effect was statistically significant, tested by the ANOVA test (p<0.01). For hormone receptor activity, treat- ment with genistein resulted in an improvement of ovarian function with LHR protein expression being enhanced and FSHR protein expression being suppressed. Our results demonstrate that Genistein played a significant role in regulating FSH and LH receptor expression to improve perimenopausal ovarian and uterine function.

Go to article

Authors and Affiliations

T. Zhang
X.X. Chi
Download PDF Download RIS Download Bibtex

Abstract

White adipose tissue plays an important role in the catabolism of branched chain amino acids (BCAAs). Two initial regulatory steps in BCAAs catabolism are catalyzed by branched chain aminotrans-ferase (BCAT) and branched chain α-keto acid dehydrogenase complex (BCKDH complex), respectively. It has been demonstrated that synthetic ligands for PPARγ receptors increased mRNA levels for enzymes involved in BCAAs catabolism. We hypothesized that feeding rats with diet rich in linoleic acid (LA), a natural PPARγ agonist modifies mRNA levels for enzymes catalyzing BCAAs degradation in adipose tissue. The current pilot study was aimed at the investigation of the effect of diet rich in LA on mRNA levels for BCATm, branched chain α-keto acid dehydrogenase (E1 component of the BCKDH), and mRNA levels for the regulatory enzymes of BCKDH complex, a specific kinase (BDK) and a specific phosphatase (PPM1K) in epididymal white adipose tissue (eWAT). Wistar male rats were fed with high unsaturated fat diet containing mainly linoleic acid (study group) or with the high saturated fat diet (control group). The relative mRNA levels were quantified by reverse transcription PCR. We have found that in rats fed diet rich in LA mRNA level for BCATm decreased, while mRNA amount for BDK increased. There was no difference between mRNA levels for BCKDH E1 and PPM1K. It is con-ceivable that changes in mRNA levels for enzymes involved in BCAAs metabolism in eWAT may lead to modification of BCAAs catabolic rate. Further studies are required to fully elucidate this issue.
Go to article

Bibliography

1. Nie C., He T., Zhang W., Zhang G., Ma X.: Branched chain amino acids: Beyond nutrition metabolism. Int J Mol Sci. 2018; 19 (4): 954. doi: 10.3390/ijms19040954
2. Brosnan J.T., Brosnan M.E.: Branched-chain amino acids: enzyme and substrate regulation. J Nutr. 2006; 136: 207S-11S. doi: 10.1093/jn/136.1.207S
3. Conway M.E., Hutson S.M.: The cytosolic and mitochondrial branched chain aminotransferase. in Branched chain amino acids in clinical nutrition. Ed. R. Rajendram, V.R. Preedy, V.B. Patel. Nutrition and Health. Humana Press, New York, NY. 2015; 25–40. doi: 10.1007/978-1-4939-1923-9_3
4. Cole J.T.: Metabolism of BCAAs. in Branched chain amino acids in clinical nutrition. Ed. R. Rajendram, V.R. Preedy, V.B. Patel. Humana Press, New York, NY. 2015; 13–24. doi: 10.1007/978-1-4939-1923-9_2
5. Yeaman S.J.: The mammalian 2-oxoacid dehydrogenases: a complex family. Trends in Biochemical Sciences. 1986; 11 (7): 293–296.
6. Wynn R.M., Li J., Brautigam C.A., Chuang J.L., Chuang D.T.: Structural and biochemical characterization of human mitochondrial branched-chain α-ketoacid dehydrogenase phosphatase. J Biol Chem. 2012; 287 (12): 9178–9192. doi: 10.1074/jbc.M111.314963
7. Herman M.A., She P., Peroni O.D., Lynch C.J., Kahn B.B.: Adipose tissue branched chain amino acid (BCAA) metabolism modulates circulating BCAA levels. J Biol Chem. 2010; 285 (15): 11348–11356. doi: 10.1074/jbc.M109.075184
8. Sun C., Mao S., Chen S., Zhang W., Liu C.: PPARs-orchestrated metabolic homeostasis in the adipose tissue. Int J Mol Sci. 2021; 22 (16): 8974. doi: 10.3390/ijms22168974
9. Kliewer S.A., Sundseth S.S., Jones S.A., et al.: Fatty acids and eicosanoids regulate gene expression through direct interactions with peroxisome proliferator-activated receptors alpha and gamma. Proc Natl Acad Sci U S A. 1997; 94 (9): 4318–4323. doi: 10.1073/pnas.94.9.4318
10. Rasouli N., Kern P.A., Elbein S.C., Sharma N.K., Das S.K.: Improved insulin sensitivity after treatment with PPARγ and PPARα ligands is mediated by genetically modulated transcripts. Pharmacogenet Genomics. 2012; 22 (7): 484–497. doi: 10.1097/FPC.0b013e328352a72e
11. Popov K.M., Zhao Y., Shimomura Y., Jaskiewicz J., Kedishvilli N.Y., Inwin J., Goodwin G.W., Harris R.A.: Dietary control and tissue expression of branched-chain alpha-ketoacid dehydrogenase kinase. Arch Biochem Biophys. 1995; 316 (1): 148–154. doi: 10.1006/abbi.1995.1022
12. Gillim S.E., Paxton R., Cook G.A., Harris R.A.: Activity state of the branched-chain alpha-ketoacid dehydrogenase complex in heart, liver, and kidney of normal, fasted, diabetic, and protein-starved rats. Bioch Biophys Res Comm. 1983; 111 (1): 74–81. doi: 10.1016/s0006-291x(83)80119-3
13. Blanchard P.G., Moreira R.J., Castro É., et al.: PPARγ is a major regulator of branched-chain amino acid blood levels and catabolism in white and brown adipose tissues. Metabolism. 2018; 89: 27–38. doi: 10.1016/j.metabol.2018.09.007
14. Palou M., Priego T., Sánchez J., Rodríguez A.M., Palou A., Picó C.: Gene expression patterns in visceral and subcutaneous adipose depots in rats are linked to their morphologic features. Cell Physiol Biochem. 2009; 24 (5–6): 547–556. doi: 10.1159/000257511
15. Nellis M.M., Doering C.B., Kasinski A., Danner D.J.: Insulin increases branched-chain alpha-ketoacid dehydrogenase kinase expression in Clone 9 rat cells. Am J Physiol Endocrinol Metab. 2002; 283 (4): E853-E860. doi: 10.1152/ajpendo.00133.2002
16. Lai M.C., Teng T.H., Yang C.: The natural PPAR agonist linoleic acid stimulated insulin release in the rat pancreas. J Vet Med Sci. 2013; 75 (11): 1449–1454. doi: 10.1292/jvms.13-0189
17. Lian K., Du C., Liu Y., et al.: Impaired adiponectin signaling contributes to disturbed catabolism of branched-chain amino acids in diabetic mice. Diabetes. 2015; 64 (1): 49–59. doi: 10.2337/db14-0312
18. Pérez-Matute P., Martínez J.A., Marti A., Moreno-Aliaga M.J.: Linoleic acid decreases leptin and adiponectin secretion from primary rat adipocytes in the presence of insulin. Lipids. 2007; 42 (10): 913–920. doi: 10.1007/s11745-007-3092-y
19. Harris R.A., Joshi M., Jeoung N.H.: Mechanisms responsible for regulation of branched-chain amino acid catabolism. Biochem Biophys Res Commun. 2004; 313 (2): 391–396. doi: 10.1016/j.bbrc.2003.11.007
20. Zhou M., Lu G., Gao C., Wang Y., Sun H.: Tissue-specific and nutrient regulation of the branched- chain α-keto acid dehydrogenase phosphatase, protein phosphatase 2Cm (PP2Cm). J Biol Chem. 2012; 287 (28): 23397–23406. doi: 10.1074/jbc.M112.351031
21. Shimomura Y., Obayashi M., Murakami T., Harris R.A.: Regulation of branched-chain amino acid catabolism: nutritional and hormonal regulation of activity and expression of the branched-chain alpha-keto acid dehydrogenase kinase. Curr Opin Clin Nutr Metab Care. 2001; 4 (5): 419–423. doi: 10.1097/00075197-200109000-00013
22. Harris R.A., Kobayashi R., Murakami T., Shimomura Y.: Regulation of branched-chain alpha-keto acid dehydrogenase kinase expression in rat liver. J Nutr. 2001; 131 (3): 841S–845S. doi: 10.1093/jn/131.3.841S
23. Green C.R., Wallace M., Divakaruni A.S., et al.: Branched-chain amino acid catabolism fuels adipocyte differentiation and lipogenesis. Nat Chem Biol. 2016; 12 (1): 15–21. doi: 10.1038/nchembio.1961
24. Koh P.L., Ho J.P., Pang C., Tan H.C., Kovalik J.P.: PH-10 — The role of leucine in stimulation of adipocyte lipolysis. Diabetes Res Clin Pract. 2016; 120: 181–182. doi: 10.1016/S0168-8227(16)31406-1
25. Liang H., Mokrani A., Chisomo-Kasiya H., et al.: Dietary leucine affects glucose metabolism and lipogenesis involved in TOR/PI3K/Akt signaling pathway for juvenile blunt snout bream Megalobrama amblycephala. Fish Physiol Biochem. 2019; 45 (2): 719–732. doi: 10.1007/s10695-018-0594-x
26. Nilsen M.S., Jersin R.Å., Ulvik A., et al.: 3-Hydroxyisobutyrate, a strong marker of insulin resistance in type 2 diabetes and obesity that modulates white and brown adipocyte metabolism. Diabetes. 2020; 69 (9): 1903–1916. doi: 10.2337/db19-1174
Go to article

Authors and Affiliations

Małgorzata Knapik-Czajka
1
Justyna Bieleń
1
Monika Zajonz
1
Anna Gawędzka
1
Jagoda Drąg
1
Małgorzata Belczyk
1

  1. Department of Biochemical Analytics, Jagiellonian University Medical College, Kraków, Poland
Download PDF Download RIS Download Bibtex

Abstract

Undisturbed branched-chain amino acids (BCAA) catabolism is necessary for normal heart function. The key enzyme in BCAA catabolism is a multienzyme branched-chain α-keto acid dehydro-genase complex (BCKDH). BCKDH activity is regulated mainly by reversible dephosphorylation (activa-tion)/phosphorylation (inactivation) cycle catalyzed by regulatory enzymes, a specific phosphatase (PPM1K) and kinase (BDK). 5-fluorouracil (5-FU) is widely used in the treatment of different types of cancer. 5-FU has the potential to cause a wide spectrum of cardiotoxicity, ranging from asymptomatic electrocardiographic changes to cardiomyopathy and subsequent cardiac failure. We hypothesize that 5-FU modifies BCKDH activity and affects cardiac muscle metabolism. The current study was aimed at the investigation of the in vivo effect of 5-FU on BCKDH activity and mRNA levels for E1, PPM1K and BDK. Wistar male rats were administered with 4 doses of 5-FU, 150 mg/kg b.wt. each (study group) or 0.3% methylcellulose (control group). BCKDH activity was assayed spectrophotometrically. The mRNA levels were quantified by real-time PCR. 5-FU treatment caused an increase in BCKDH activity that appears to result mainly from increased dephosphorylation of the complex and is associated with an increase of PPM1K mRNA level and reduction of BDK and E1 mRNA levels. It is conceivable that 5-FU stimulation of BCKDH is an adaptive reaction with the purpose of enhancing the BCAA catabolism and protecting from toxic effect caused by excessive accumulation of these amino acids in heart.
Go to article

Authors and Affiliations

Małgorzata Knapik-Czajka
1
Michał Jurczyk
2
Justyna Bieleń
1
Veronika Aleksandrovych
2
Anna Gawędzka
1
Paulina Stach
2
Jagoda Drąg
1
Krzysztof Gil
3

  1. Department of Biochemical Analytics, Jagiellonian University Medical College, Kraków, Poland
  2. Department of Pathophysiology, Jagiellonian University Medical College, Kraków, Poland
  3. 2Department of Pathophysiology, Jagiellonian University Medical College, Kraków, Poland

This page uses 'cookies'. Learn more