Search In this Thesis
   Search In this Thesis  
العنوان
Investigating the effect of azilsartan and adipose tissue-derived mesenchymal stem cells against cisplatin-induced cytotoxicity /
المؤلف
Baskharon, Olivia Nady Beshay.
هيئة الاعداد
باحث / اوليفيا نادى بشاى بسخرون
مشرف / مصطفى فتحى رمضان
مشرف / مايكل عاطف فوزي
مشرف / أماني عبدالرحيم بخيت
الموضوع
Biochemistry.
تاريخ النشر
2023.
عدد الصفحات
162 p. :
اللغة
الإنجليزية
الدرجة
الدكتوراه
التخصص
الصيدلة ، علم السموم والصيدلانيات
تاريخ الإجازة
11/6/2023
مكان الإجازة
جامعة المنيا - كلية الصيدلة - الكيمياء الحيوية
الفهرس
Only 14 pages are availabe for public view

from 185

from 185

Abstract

CIS is a platinum derivative chemotherapeutic drug that is largely utilized for managing of a wide range of cancers. However, its therapeutic value and efficacy are hindered owing to its adverse effects, which include ototoxicity, kidney toxicity, and hepatotoxicity. Traditionally, CIS administration triggers ROS which could damage either tumor cells or normal cells, without distinction. Overproduction of ROS, which cause the development of CIS-induced cytotoxicity, is thought to be triggered by several processes, including oxidative stress, inflammation as well as apoptosis. Furthermore, the members of MAPKs class have attributed to triggering a variety of injuries, notably CIS-evoked cytotoxicity. TGF-β signaling pathway and its fibrogenic effects are reported to be elicited by ROS. EMT stimulation has a number of cytokines involved, but TGFβ-1 has been shown to be the key regulatory actor among them. EMT, a complex biological phenomenon, causes cells to lose their epithelial phenotype and is linked by a decrease in epithelial markers such E-cad. These cells develop a mesenchymal character and generate more mesenchymal markers such as fibronectin, vimentin, and α-SMA.
Cell-based therapeutics are becoming as a viable substitute to traditional pharmaceutical treatments. MSCs provide several advantages in regenerative medicine, like multi- differentiation potential, minimal immunogenicity and their higher proliferation rate. Interestingly, numerous studies have shown that MSCs therapy alleviated CIS-evoked nephrotoxicity. Additionally, it has been demonstrated that MSCs has the ability to protect the liver against CIS-evoked hepatotoxicity. However, maintaining their characters and selecting the optimum conditions for their therapeutic efficacy are somewhat challenging.
AZL, a novel ARBs, has the strongest antihypertensive activity when compared to other drugs in the same family. Previous researches demonstrated other pharmacological actions and pleiotropic health benefits of AZL. Repurposing drugs and looking for new pharmacological actions for synthetic or natural candidates have got a great attention. So, the aims of the present study were to:
1- Investigate, for the first time, the potential protective impact of AZL or AT-MSCs against kidney CIS–evoked EMT. In addition to examine the impact of AZL on the potential renal-protective effect of AT-MSCs against CIS–evoked EMT on molecular bases by inspecting the contribution of TGF-β/Smad signaling cascade mediated in this effect.
2- Assess the potential protective impact of AZL or AT-MSCs against CIS–evoked hepatotoxicity as well as to demonstrate the effect of AZL on the potential hepato-protective activity of AT-MSCs on molecular bases by investigating MAPK and apoptotic signaling pathways which may be implicated in this effect.
The animals utilized in this investigation were male Wister strain albino rats weighing 170-200. These animals were allocated to 9 groups as follows:
1. NC group: Rats received 0.5% w/v carboxy methylcellulose solution, orally for two weeks and on the 8th day received isotonic saline single i.p. injection.
2. AZL.4 group: Rats orally received AZL (4 mg kg−1) for two weeks.
3. CIS group: Rats received vehicle (0.5% carboxy methylcellulose solution) orally for two weeks and on the 8th day received CIS (6 mg kg−1) (single i.p. injection).
4. CIS + AZL.2 group: Rats orally received AZL (2 mg kg−1) for two weeks and on the 8th day received CIS (6 mg kg−1) (single i.p. injection).
5. CIS + AZL.4 group: Rats orally received AZL (4 mg kg−1) for two weeks and on the 8th day received CIS (6 mg kg−1) (single i.p. injection).
6. CIS + AT-MSCs group: Rats received CIS (6 mg kg−1) (single i.p. injection) and were administered AT-MSCs (1×106 cells) by tail vein injection 24 h after the CIS injection.
7. CIS + AZL.2 + AT-MSCs group: Rats orally received AZL (2 mg kg−1) for two weeks and on the 8th day received CIS (6 mg kg−1) (single i.p. injection) and were administered AT-MSCs (1×106 cells) by tail vein injection 24 h after the CIS injection.
8. CIS + AZL.4 + AT-MSCs group: Rats orally received AZL (4 mg kg−1) for two weeks and on the 8th day received CIS (6 mg kg−1) (single i.p. injection) and were administered AT-MSCs (1×106 cells) by tail vein injection 24 h after the CIS injection.
9. CIS + VIT C group: Rats orally received VIT C (20 mg kg−1) for two weeks and on the 8th day received CIS (6 mg kg−1) (single i.p. injection). CIS + VIT C group utilized as a positive control group.
The rats were sacrificed seven days after CIS injection. Blood samples and the tissues were collected. Weights of liver and kidney were registered for estimation of liver and kidney indices. A portion of tissue samples was placed in ten percent neutral buffered formalin for histological studies before paraffin embedding. Other tissue samples were homogenized in cold potassium phosphate buffer (0.05 M, pH 7.4) and were centrifuged for collecting the supernatant in order to estimate oxidative stress markers. The remainder was utilized for western blotting analysis and qRT-PCR.
This study revealed that CIS considerably raised SCr, BUN, uric acid as well as kidney MDA levels, with concurrent reduction in serum total protein, kidney GSH level and SOD activity. Moreover, it reduced the expression of renal E-cad gene, elevated the renal α-SMA, N-cadherin as well as vimentin genes expression, lowered the expression of E-cad protein and raised the α-SMA, TGF-β1, p/t-Smad2/3 and Snail proteins expression. In addition, kidney tissues exhibited significant histopathological changes as well as substantial collagen buildup. Contrarily, the administration of either AZL or AT-MSCs dramatically reduced these CIS-evoked changes. Remarkably, the combination of AZL with AT-MSCs provides a greater alleviation impact than AT-MSCs alone.
Furthermore, our findings demonstrated that CIS elevated the serum ALT and AST levels, reduced albumin level and remarkably changed the liver architecture. It increased the hepatic TNF-α and IL-6 genes expression as well as up-regulated the p/t-ERK1/2, p/t-JNK1/2, p/t-p38, Bax and cleaved caspase-3 proteins and down-regulated Bcl-2 protein. This study found that the combination therapy (AZL + AT-MSCs) had a great impact on both liver’s architecture and biomarkers via restoring ALT, AST and albumin levels, reducing the MDA level and increasing the antioxidants GSH and SOD levels over the single treatment groups. Furthermore, the combination therapy considerably reduced the level of hepatic TNF-α and IL-6 genes expression, comparatively to AZL or AT-MSCs mono-therapy, as well as significantly up-regulated Bcl-2 protein expression, and down-regulated the hepatic expression of p/t-ERK1/2, p/t-JNK1/2, p/t-p38, Bax and cleaved caspase-3 proteins.
In conclusion, for the first time, this work indicated that AZL and/ or AT-MSCs efficiently alleviated CIS-evoked EMT through modulating oxidative stress as well as TGF-β/Smad signaling cascade. Furthermore, AZL enhanced the effect of AT-MSCs which may afford a significant protective action against CIS-induced EMT over the use of AT-MSCs alone. Consequently, AZL and AT-MSCs are valuable therapies for protecting the kidney from CIS-evoked EMT which can lead to kidney fibrosis and failure.
Furthermore, this study indicated that dual administration of AZL plus AT-MSCs may provide markedly greater hepatoprotection against CIS-induced hepatotoxicity than AZL or AT-MSCs usage alone. They were effective in reducing CIS-evoked hepatotoxicity by suppressing oxidative stress, inflammation, MAPKs and apoptotic pathways. AZL considerably improved the hepatoprotective capability of AT-MSCs against CIS-evoked hepatotoxicity, recommending this strategy to increase the therapeutic potential of AT-MSCs.
Further pre-clinical studies are recommended to reinforce these findings and explore more possible molecular mechanisms explaining the impact of AZL plus AT-MSCs on CIS-induced nephrotoxicity and hepatotoxicity.