Pub Date : 2025-01-17DOI: 10.1016/j.tice.2025.102751
Ting Wang, Meng-Yan Li, Zhong Pei, Qiu-Xia Chen, Qiu-Sheng Cheng, Ze Li
Background: Parkinson's Disease (PD) often presents with a compromised blood-brain barrier (BBB), which hyperglycemia may exacerbate. Pericytes, a key cell for BBB integrity, are potential therapeutic targets for neurodegenerative disorders. Few studies have developed 3D PD cell models incorporating neurovascular units (NVU) through the co-culture of human endothelial, pericytes, astrocytes, and SH-SY5Y cells to evaluate BBB impairment and the role of pericytes under hyperglycemic condition.
Method: A 3D PD like cell model was developed using 6-OHDA-affected SH-SY5Y cells, combined with endothelial cells, pericytes, and astrocytes through the Real Architecture for Tissue (RAFT) 3D co-culture system. PD incorporating reduced (30 % and 89 %) PDGFRβ NVU (RPN) with or without hyperglycemic model (HM) were also established. BBB permeability to sodium fluorescein was assessed, and BBB impairment was evaluated using BBB-associated proteins (ZO-1, CD54, CD144), cell-specific proteins (CD31, GFAP, PDGFRβ, CD13), tyrosine hydroxylase (TH), α-synuclein, oligomeric α-synuclein, and α-synuclein (ser9).
Results: PD 3D cell models incorporating RPN with or without hyperglycemia were successfully established in vitro. Graduately increased BBB impairment was observed in PD, PD with RPN, and PD with RPN combined with HM, indicated by decreased BBB-associated and cell-specific proteins, reduced TH, and increased α-synuclein, oligomeric α-synuclein, and α-synuclein (ser9) compared to the NVU model.
Conclusion: Reduced pericyte PDGFRβ could increase BBB permeability, accelerate PD progression, and exacerbate under hyperglycemic condition.
{"title":"Down-regulation of platelet-derived growth factor receptor β in pericytes increases blood-brain barrier permeability and significantly enhances α-synuclein in a Parkinson's Disease 3D cell model in vitro under hyperglycemic condition.","authors":"Ting Wang, Meng-Yan Li, Zhong Pei, Qiu-Xia Chen, Qiu-Sheng Cheng, Ze Li","doi":"10.1016/j.tice.2025.102751","DOIUrl":"https://doi.org/10.1016/j.tice.2025.102751","url":null,"abstract":"<p><strong>Background: </strong>Parkinson's Disease (PD) often presents with a compromised blood-brain barrier (BBB), which hyperglycemia may exacerbate. Pericytes, a key cell for BBB integrity, are potential therapeutic targets for neurodegenerative disorders. Few studies have developed 3D PD cell models incorporating neurovascular units (NVU) through the co-culture of human endothelial, pericytes, astrocytes, and SH-SY5Y cells to evaluate BBB impairment and the role of pericytes under hyperglycemic condition.</p><p><strong>Method: </strong>A 3D PD like cell model was developed using 6-OHDA-affected SH-SY5Y cells, combined with endothelial cells, pericytes, and astrocytes through the Real Architecture for Tissue (RAFT) 3D co-culture system. PD incorporating reduced (30 % and 89 %) PDGFRβ NVU (RPN) with or without hyperglycemic model (HM) were also established. BBB permeability to sodium fluorescein was assessed, and BBB impairment was evaluated using BBB-associated proteins (ZO-1, CD54, CD144), cell-specific proteins (CD31, GFAP, PDGFRβ, CD13), tyrosine hydroxylase (TH), α-synuclein, oligomeric α-synuclein, and α-synuclein (ser9).</p><p><strong>Results: </strong>PD 3D cell models incorporating RPN with or without hyperglycemia were successfully established in vitro. Graduately increased BBB impairment was observed in PD, PD with RPN, and PD with RPN combined with HM, indicated by decreased BBB-associated and cell-specific proteins, reduced TH, and increased α-synuclein, oligomeric α-synuclein, and α-synuclein (ser9) compared to the NVU model.</p><p><strong>Conclusion: </strong>Reduced pericyte PDGFRβ could increase BBB permeability, accelerate PD progression, and exacerbate under hyperglycemic condition.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102751"},"PeriodicalIF":2.7,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143029638","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-16DOI: 10.1016/j.tice.2025.102745
Ziyang Ma, Yirong Wang, Xiaoyu Zhang, Shi Ding, Jian Fan, Tian Li, Xin Xiao, Jing Li
Objective: Patients with osteosarcoma (OS) exhibit metastasis upon diagnosis, and the condition frequently acquires resistance to traditional chemotherapy treatments, failing the therapy. The objective of this research was to examine the impact of curculigoside (Cur), a key phenolic compound discovered in the rhizome of C. orchioides Gaertn, on OS cells and the surrounding tumor environment.
Methods: We assessed the impact of curculigoside on tumor inhibition in four osteosarcoma cell lines and mice tumor xenograft models using various techniques including cell viability assay, wound healing assay, cell apoptosis analysis, immunofluorescent staining, and IHC. Moreover, we created a mini-PDX model by utilizing freshly obtained primary OS cells from surgically removed OS tissues to evaluate the possible clinical use of Cur.
Result: The results of our study show that Cur triggers cell death in OS cells and enhances the maturation of RAW264.7 cells. By effectively inhibiting the growth of OS cells, these actions mechanistically trigger the catastrophic buildup of unbound iron and uncontrolled lipid peroxidation, ultimately resulting in ferroptosis. Moreover, additional validation of Cur's substantial antineoplastic impact is obtained through in vivo experiments employing xenograft and mini-PDX models.
Conclusions: To sum up, this research is the initial one to exhibit the anti-tumor effects of Cur on OS using various methods, indicating that Cur shows potential as a viable approach for treating OS.
{"title":"Curculigoside exhibits multiple therapeutic efficacy to induce apoptosis and ferroptosis in osteosarcoma via modulation of ROS and tumor microenvironment.","authors":"Ziyang Ma, Yirong Wang, Xiaoyu Zhang, Shi Ding, Jian Fan, Tian Li, Xin Xiao, Jing Li","doi":"10.1016/j.tice.2025.102745","DOIUrl":"https://doi.org/10.1016/j.tice.2025.102745","url":null,"abstract":"<p><strong>Objective: </strong>Patients with osteosarcoma (OS) exhibit metastasis upon diagnosis, and the condition frequently acquires resistance to traditional chemotherapy treatments, failing the therapy. The objective of this research was to examine the impact of curculigoside (Cur), a key phenolic compound discovered in the rhizome of C. orchioides Gaertn, on OS cells and the surrounding tumor environment.</p><p><strong>Methods: </strong>We assessed the impact of curculigoside on tumor inhibition in four osteosarcoma cell lines and mice tumor xenograft models using various techniques including cell viability assay, wound healing assay, cell apoptosis analysis, immunofluorescent staining, and IHC. Moreover, we created a mini-PDX model by utilizing freshly obtained primary OS cells from surgically removed OS tissues to evaluate the possible clinical use of Cur.</p><p><strong>Result: </strong>The results of our study show that Cur triggers cell death in OS cells and enhances the maturation of RAW264.7 cells. By effectively inhibiting the growth of OS cells, these actions mechanistically trigger the catastrophic buildup of unbound iron and uncontrolled lipid peroxidation, ultimately resulting in ferroptosis. Moreover, additional validation of Cur's substantial antineoplastic impact is obtained through in vivo experiments employing xenograft and mini-PDX models.</p><p><strong>Conclusions: </strong>To sum up, this research is the initial one to exhibit the anti-tumor effects of Cur on OS using various methods, indicating that Cur shows potential as a viable approach for treating OS.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102745"},"PeriodicalIF":2.7,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143047998","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
In clinical practice, there is a demand for innovative wound healing methods to tackle full thickness skin injuries, especially in those with diabetes. In this study, we examined if collagen-based hydrogel from amniotic membrane (CHAM) loaded with quercetin could enhance healing in diabetic rats. Sixty diabetic rats were randomly divided into the control group, CHAM group, quercetin group, and CHAM+Quercetin group. Sampling took place on days 4 and 8 for additional evaluations. Our findings showed that the rates of wound contraction, volumes of new epidermis and dermis, fibroblast and blood vessel counts, collagen deposition, and concentrations of TGF-β1 and VEGF cytokines were significantly higher in the treatment groups compared to the control group, with these changes being more pronounced in the CHAM+Quercetin group. This is while the counts of neutrophils and macrophages, along with the concentration levels of IL-6, IL-1β, and TNF-α cytokines dropped more noticeably in the CHAM+Quercetin group in comparison to the other groups. In summary, it was determined that the combination of CHAM and quercetin significantly enhances diabetic wound healing.
{"title":"Collagen-based hydrogel derived from amniotic membrane loaded with quercetin accelerates wound healing by improving stereological parameters and reducing inflammation in a diabetic rat model.","authors":"Xin Xiang, Weijun Peng, Qi Lu, Shiqi Ma, Jinfeng Wang, Jinling Ma, Xiaokai Wei, Mengmeng Li, Hongfeng Wang","doi":"10.1016/j.tice.2025.102743","DOIUrl":"https://doi.org/10.1016/j.tice.2025.102743","url":null,"abstract":"<p><p>In clinical practice, there is a demand for innovative wound healing methods to tackle full thickness skin injuries, especially in those with diabetes. In this study, we examined if collagen-based hydrogel from amniotic membrane (CHAM) loaded with quercetin could enhance healing in diabetic rats. Sixty diabetic rats were randomly divided into the control group, CHAM group, quercetin group, and CHAM+Quercetin group. Sampling took place on days 4 and 8 for additional evaluations. Our findings showed that the rates of wound contraction, volumes of new epidermis and dermis, fibroblast and blood vessel counts, collagen deposition, and concentrations of TGF-β1 and VEGF cytokines were significantly higher in the treatment groups compared to the control group, with these changes being more pronounced in the CHAM+Quercetin group. This is while the counts of neutrophils and macrophages, along with the concentration levels of IL-6, IL-1β, and TNF-α cytokines dropped more noticeably in the CHAM+Quercetin group in comparison to the other groups. In summary, it was determined that the combination of CHAM and quercetin significantly enhances diabetic wound healing.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102743"},"PeriodicalIF":2.7,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012220","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-15DOI: 10.1016/j.tice.2025.102744
Marwa M Khalaf, Emad H M Hassanein, Hamada S Qebesy, Abdullatif A Ahmed, Heba M Mahmoud
Doxorubicin (DOX) is an anthracycline chemotherapy employed in treating malignancies. Unfortunately, the clinical application of DOX is limited due to its nephrotoxicity. Granisetron (GRAN) is a 5-HT3 receptor blocker used widely to manage post-chemotherapy nausea and vomiting with anti-inflammatory, anti-oxidant, and anti-apoptotic bioactivities. We plan to examine the renoprotective effect of GRAN against DOX-associated renal toxicity. In this investigation, twenty-four adult male Wistar rats were allocated to control, DOX (30 mg/kg, i.p), and GRAN (2.5 mg/kg, p.o) + DOX groups. GRAN attenuated renal impairment induced by DOX in rats by decreasing the BUN, creatinine, KIM-1, and Cys-C levels, and such finding is supported by attenuating histological alterations caused by DOX. GRAN combated oxidative stress proved by decreasing MDA content and elevating GSH and CAT levels mediated by Nrf2 activation. GRAN suppressed inflammation evidenced by decreasing IL-6 and TNF-α levels mediated by downregulation of inflammatory sensitive controllers TLR-4, NLRP3, and p38 MAPK. GRAN prevented apoptosis by controlling renal expression of BAX, caspase-3 and Bcl2. Therefore, GRAN holds promise agent against DOX-induced renal toxicity by upregulating Nrf2 and suppressing apoptosis and inflammatory cascadeTLR4/p38 MAPK/ NLRP3.
{"title":"Granisetron ameliorates doxorubicin-evoked nephrotoxicity via modulation of Nrf2 and TLR4/p38 MAPK/NLRP3 signals in rats.","authors":"Marwa M Khalaf, Emad H M Hassanein, Hamada S Qebesy, Abdullatif A Ahmed, Heba M Mahmoud","doi":"10.1016/j.tice.2025.102744","DOIUrl":"https://doi.org/10.1016/j.tice.2025.102744","url":null,"abstract":"<p><p>Doxorubicin (DOX) is an anthracycline chemotherapy employed in treating malignancies. Unfortunately, the clinical application of DOX is limited due to its nephrotoxicity. Granisetron (GRAN) is a 5-HT3 receptor blocker used widely to manage post-chemotherapy nausea and vomiting with anti-inflammatory, anti-oxidant, and anti-apoptotic bioactivities. We plan to examine the renoprotective effect of GRAN against DOX-associated renal toxicity. In this investigation, twenty-four adult male Wistar rats were allocated to control, DOX (30 mg/kg, i.p), and GRAN (2.5 mg/kg, p.o) + DOX groups. GRAN attenuated renal impairment induced by DOX in rats by decreasing the BUN, creatinine, KIM-1, and Cys-C levels, and such finding is supported by attenuating histological alterations caused by DOX. GRAN combated oxidative stress proved by decreasing MDA content and elevating GSH and CAT levels mediated by Nrf2 activation. GRAN suppressed inflammation evidenced by decreasing IL-6 and TNF-α levels mediated by downregulation of inflammatory sensitive controllers TLR-4, NLRP3, and p38 MAPK. GRAN prevented apoptosis by controlling renal expression of BAX, caspase-3 and Bcl2. Therefore, GRAN holds promise agent against DOX-induced renal toxicity by upregulating Nrf2 and suppressing apoptosis and inflammatory cascadeTLR4/p38 MAPK/ NLRP3.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102744"},"PeriodicalIF":2.7,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143041958","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-14DOI: 10.1016/j.tice.2025.102739
Tao Su, Yuting Si
Background: Postoperative cognitive dysfunction (POCD) is a postoperative complication that can be induced by anaesthesia. PCSK9 has been shown to have a role in neuronal development and apoptosis. However, PCSK9 has not been studied in sevoflurane-induced POCD-related disorders.
Objective: To explore whether PCSK9 can exacerbate sevoflurane-induced neuroinflammatory response and apoptosis by up-regulating cGAS-STING signalling.
Methods: A POCD model was constructed by stimulating BV2 microglia with Sevoflurane. CCK8 was used to detect the cell viability, and immunofluorescence was used to observe the expression of microglial activation markers (Iba-1, CD11b) and BDNF to determine the activation of BV2 microglia. Cell proliferation was measured by EDU staining, and apoptosis was analyzed by flow cytometry and western blot. The levels of inflammatory cytokines, ROS, MDA, SOD and CAT were respectively detected by ELISA, DCFH-DA staining, and kits to determine the neuroinflammation and oxidative stress of cells. Mitochondrial ROS, mitochondrial membrane potential, mtDNA and ATP levels were measured to evaluate cellular mitochondrial function.
Results: Transfection of si-PCSK9 inhibited Sevoflurane-induced microglial activation and restored cellular viability, promoted cell proliferation, inhibited apoptosis and neuroinflammation, decreased ROS and MDA levels in the cells while up-regulating the levels of SOD and CAT, thus inhibiting oxidative stress, restored the mitochondrial membrane potential to normal and decreased mitochondrial ROS and mtDNA levels and increased ATP production, thereby alleviating mitochondrial dysfunction. Moreover, PCSK9 depletion also down-regulated the expression of cGAS and STING to inactivate cGAS-STING signaling. However, cGAS overexpression partially reversed the effects of si-PCSK9.
Conclusion: PCSK9 exacerbates sevoflurane-induced neuroinflammatory response and apoptosis by upregulating cGAS-STING signaling.
{"title":"PCSK9 exacerbates sevoflurane-induced neuroinflammatory response and apoptosis by up-regulating cGAS-STING signal.","authors":"Tao Su, Yuting Si","doi":"10.1016/j.tice.2025.102739","DOIUrl":"https://doi.org/10.1016/j.tice.2025.102739","url":null,"abstract":"<p><strong>Background: </strong>Postoperative cognitive dysfunction (POCD) is a postoperative complication that can be induced by anaesthesia. PCSK9 has been shown to have a role in neuronal development and apoptosis. However, PCSK9 has not been studied in sevoflurane-induced POCD-related disorders.</p><p><strong>Objective: </strong>To explore whether PCSK9 can exacerbate sevoflurane-induced neuroinflammatory response and apoptosis by up-regulating cGAS-STING signalling.</p><p><strong>Methods: </strong>A POCD model was constructed by stimulating BV2 microglia with Sevoflurane. CCK8 was used to detect the cell viability, and immunofluorescence was used to observe the expression of microglial activation markers (Iba-1, CD11b) and BDNF to determine the activation of BV2 microglia. Cell proliferation was measured by EDU staining, and apoptosis was analyzed by flow cytometry and western blot. The levels of inflammatory cytokines, ROS, MDA, SOD and CAT were respectively detected by ELISA, DCFH-DA staining, and kits to determine the neuroinflammation and oxidative stress of cells. Mitochondrial ROS, mitochondrial membrane potential, mtDNA and ATP levels were measured to evaluate cellular mitochondrial function.</p><p><strong>Results: </strong>Transfection of si-PCSK9 inhibited Sevoflurane-induced microglial activation and restored cellular viability, promoted cell proliferation, inhibited apoptosis and neuroinflammation, decreased ROS and MDA levels in the cells while up-regulating the levels of SOD and CAT, thus inhibiting oxidative stress, restored the mitochondrial membrane potential to normal and decreased mitochondrial ROS and mtDNA levels and increased ATP production, thereby alleviating mitochondrial dysfunction. Moreover, PCSK9 depletion also down-regulated the expression of cGAS and STING to inactivate cGAS-STING signaling. However, cGAS overexpression partially reversed the effects of si-PCSK9.</p><p><strong>Conclusion: </strong>PCSK9 exacerbates sevoflurane-induced neuroinflammatory response and apoptosis by upregulating cGAS-STING signaling.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102739"},"PeriodicalIF":2.7,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012284","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Psychotropic stimulants like methamphetamine (METH) have an impact on the physiology, behavior, and psychology of human beings and can damage the reproductive and neuroendocrine systems in them. These deleterious impacts include a temporary drop in the relative weight of the testis along with adverse effects on spermatogenesis. Sambucus nigra, also known as elderberry (EB) or sweet elder, is a source of bioactive compounds that has drawn growing attention for its potential beneficial impact in preventing and treating several diseases. In this experimental research, 36 adult male rats were classified into three groups: (1) control, (2) METH, and (3) METH receiving the EB diet. The rats were injected with METH at a dose of 20 mg/kg for 28 days during treatment with the EB diet. Then, the rats were euthanized, and their sperm samples were collected for sperm parameters analysis. Afterward, the testis samples were taken for histopathological experimentations, immunohistochemistry against TNF-α and caspase-3, and serum testosterone levels. Our findings indicated largely improved sperm and stereological parameters, like spermatogonia, primary spermatocyte, round spermatid, and Leydig cells, and an increased serum testosterone level in the METH group receiving the EB diet compared to the other METH group. The results also revealed a significantly decreased TNF-α and caspase-3 expression in the METH+EB group compared to the METH group. In conclusion, the EB diet is regarded as an alternative treatment for improving the spermatogenesis process in reproductive toxicity induced by METH exposure.
{"title":"Elderberry diet improves sperm quality and histological parameters of testicular tissue in adult male rats exposed to methamphetamine.","authors":"Haleh Hemmatparast, Zahra Shams Mofarahe, Mohammad-Amin Abdollahifar, Samira Ezi, Ghazal Khanjari, Nahal Babaeian Amini, Zahra Niakan, Pourya Raee, Meysam Hassani Moghaddam, Mobina Fathi, Kimia Vakili, Amirreza Beirami, Maral Hasanzadeh, Ramtin Hajibeygi, Abbas Aliaghaei, Mojtaba Sani, Mohsen Norouzian","doi":"10.1016/j.tice.2025.102732","DOIUrl":"https://doi.org/10.1016/j.tice.2025.102732","url":null,"abstract":"<p><p>Psychotropic stimulants like methamphetamine (METH) have an impact on the physiology, behavior, and psychology of human beings and can damage the reproductive and neuroendocrine systems in them. These deleterious impacts include a temporary drop in the relative weight of the testis along with adverse effects on spermatogenesis. Sambucus nigra, also known as elderberry (EB) or sweet elder, is a source of bioactive compounds that has drawn growing attention for its potential beneficial impact in preventing and treating several diseases. In this experimental research, 36 adult male rats were classified into three groups: (1) control, (2) METH, and (3) METH receiving the EB diet. The rats were injected with METH at a dose of 20 mg/kg for 28 days during treatment with the EB diet. Then, the rats were euthanized, and their sperm samples were collected for sperm parameters analysis. Afterward, the testis samples were taken for histopathological experimentations, immunohistochemistry against TNF-α and caspase-3, and serum testosterone levels. Our findings indicated largely improved sperm and stereological parameters, like spermatogonia, primary spermatocyte, round spermatid, and Leydig cells, and an increased serum testosterone level in the METH group receiving the EB diet compared to the other METH group. The results also revealed a significantly decreased TNF-α and caspase-3 expression in the METH+EB group compared to the METH group. In conclusion, the EB diet is regarded as an alternative treatment for improving the spermatogenesis process in reproductive toxicity induced by METH exposure.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102732"},"PeriodicalIF":2.7,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012221","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-11DOI: 10.1016/j.tice.2025.102733
Mahmoud El Safadi, Tawaf Ali Shah, Syeda Sania Zahara, Yousef A Bin Jardan, Mohammed Bourhia
Bifenthrin (BFN) is a noxious insecticide which is reported to damage various body organs. Daidzein (DZN) is a natural flavone with excellent pharmacological properties. This research was conducted to evaluate the alleviative strength of DZN to counteract BFN prompted liver toxicity in male albino rats. Thirty-two rats were divided into 4 groups i.e., the control, BFN (7 mg /kg), BFN (7 mg/kg) + DZN (20 mg/kg) and DZN (20 mg/kg) alone group. The biochemical assessment was performed by using qRT PCR as well as standard ELISA protocols. The findings are validated by applying pharmacodynamic techniques including molecular simulation. It was observed that BFN reduced the gene expressions of phosphoinositide 3-kinase (PI3K), phosphatidylinositol-3, 4, 5-triphosphate (PIP3), Protein kinase B (Akt), nuclear factor erythroid 2-related factor 2 (Nrf-2) while promoting the gene expressions of Kelch-like ECH-associated protein 1 (Keap-1). Moreover, BFN notably reduced the activities of glutathione reductase (GSR), heme-oxygenase-1 (HO-1), glutathione peroxidase (GPx), superoxide dismutase (SOD), and catalase (CAT) while elevating the levels of reactive oxygen species (ROS) and malondialdehyde (MDA). BFN promoted the levels of matrix metallopeptidase 2 (MMP-2), Procollagen III N-terminal Pro-peptide (PIIINP), alkaline phosphatase (ALP), transforming growth factor-beta-1 (TGF-β1), aspartate aminotransferase (AST), tissue inhibitor of matrix metalloproteinases 1 (TIMP1), and alanine aminotransferase (ALT). The levels of nuclear factor- kappa B (NF-κB), interleukin-1 beta (IL-1β), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and cyclooxygenase-2 (COX-2) were increased following the BFN intoxication. BFN enhanced the expressions of cysteine-aspartic acid protease-3 (Caspase-3) and Bcl-2-associated X protein (Bax) while suppressing the gene expression of B-cell lymphoma-2 (Bcl-2). Moreover, BFN disrupted the normal histology of liver tissues. Nonetheless, DZN treatment remarkably alleviated hepatic damages owing to its antioxidative, anti-apoptotic as well as anti-inflammatory abilities. However, DZN supplementation remarkably safeguarded which is further confirmed by in-silico assessment.
{"title":"Regulation of TGF-β1, PI3K/PIP3/Akt, Nrf-2/Keap-1 and NF-κB signaling pathways to avert bifenthrin induced hepatic injury: A palliative role of daidzein.","authors":"Mahmoud El Safadi, Tawaf Ali Shah, Syeda Sania Zahara, Yousef A Bin Jardan, Mohammed Bourhia","doi":"10.1016/j.tice.2025.102733","DOIUrl":"https://doi.org/10.1016/j.tice.2025.102733","url":null,"abstract":"<p><p>Bifenthrin (BFN) is a noxious insecticide which is reported to damage various body organs. Daidzein (DZN) is a natural flavone with excellent pharmacological properties. This research was conducted to evaluate the alleviative strength of DZN to counteract BFN prompted liver toxicity in male albino rats. Thirty-two rats were divided into 4 groups i.e., the control, BFN (7 mg /kg), BFN (7 mg/kg) + DZN (20 mg/kg) and DZN (20 mg/kg) alone group. The biochemical assessment was performed by using qRT PCR as well as standard ELISA protocols. The findings are validated by applying pharmacodynamic techniques including molecular simulation. It was observed that BFN reduced the gene expressions of phosphoinositide 3-kinase (PI3K), phosphatidylinositol-3, 4, 5-triphosphate (PIP3), Protein kinase B (Akt), nuclear factor erythroid 2-related factor 2 (Nrf-2) while promoting the gene expressions of Kelch-like ECH-associated protein 1 (Keap-1). Moreover, BFN notably reduced the activities of glutathione reductase (GSR), heme-oxygenase-1 (HO-1), glutathione peroxidase (GPx), superoxide dismutase (SOD), and catalase (CAT) while elevating the levels of reactive oxygen species (ROS) and malondialdehyde (MDA). BFN promoted the levels of matrix metallopeptidase 2 (MMP-2), Procollagen III N-terminal Pro-peptide (PIIINP), alkaline phosphatase (ALP), transforming growth factor-beta-1 (TGF-β1), aspartate aminotransferase (AST), tissue inhibitor of matrix metalloproteinases 1 (TIMP1), and alanine aminotransferase (ALT). The levels of nuclear factor- kappa B (NF-κB), interleukin-1 beta (IL-1β), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and cyclooxygenase-2 (COX-2) were increased following the BFN intoxication. BFN enhanced the expressions of cysteine-aspartic acid protease-3 (Caspase-3) and Bcl-2-associated X protein (Bax) while suppressing the gene expression of B-cell lymphoma-2 (Bcl-2). Moreover, BFN disrupted the normal histology of liver tissues. Nonetheless, DZN treatment remarkably alleviated hepatic damages owing to its antioxidative, anti-apoptotic as well as anti-inflammatory abilities. However, DZN supplementation remarkably safeguarded which is further confirmed by in-silico assessment.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102733"},"PeriodicalIF":2.7,"publicationDate":"2025-01-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143024936","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-11DOI: 10.1016/j.tice.2025.102725
Mai A Samak, Yara M Elfakharany, Nancy Huessiny, Amira Ebrahim Alsemeh
Toxic-induced cerebellar syndrome (TOICS) poses substantial neurological challenges, given its diverse causes and complex manifestations. Gold nanoparticles (AuNPs) have gained significant attention owing to enhanced biocompatibility for therapeutic interventions. We aimed to investigate the impacts of AuNPs on cerebellar cytomolecular, immunohistochemical and ultrastructural alterations in the context of phenytoin-experimentally induced TOICS. Thirty male albino rats were assigned randomly to three equal groups; control, phenytoin (PHT) and PHT+ AuNPs groups. Cerebellar sections were examined histopathologically, ultra-structurally and immunohistochemically for GFAP and p-Tau. Cerebellar tissues were evaluated for TNF-α, IL-1β, MDA, CAT, SOD and CREB mRNA. Our data confirmed observable amelioration of histopathological and ultrastructural cerebellar alterations of Purkinje and granule cells after AuNPs cotreatment. Histomorphometric measures revealed AuNPs-induced significant downregulation of p-Tau and GFAP immune-expression. Concurrently, TNF-α, IL-1β, MDA were significantly quenched in cerebellar tissues after AuNPs cotreatment, on contrary to notable restoration of CAT, SOD and CREB mRNA levels. These outcomes confirm that AuNPs hold promise as a therapeutic strategy for TOICS, warranting further exploration of their mechanisms and clinical applications in cerebellar disorders.
{"title":"Gold nanoparticles \"AuNPs\" -mediated amelioration of experimentally toxic-induced cerebellar syndrome: Insights into cytomolecular and immuno-histochemical modifications, with a focus on CREB/ Tau modulation.","authors":"Mai A Samak, Yara M Elfakharany, Nancy Huessiny, Amira Ebrahim Alsemeh","doi":"10.1016/j.tice.2025.102725","DOIUrl":"https://doi.org/10.1016/j.tice.2025.102725","url":null,"abstract":"<p><p>Toxic-induced cerebellar syndrome (TOICS) poses substantial neurological challenges, given its diverse causes and complex manifestations. Gold nanoparticles (AuNPs) have gained significant attention owing to enhanced biocompatibility for therapeutic interventions. We aimed to investigate the impacts of AuNPs on cerebellar cytomolecular, immunohistochemical and ultrastructural alterations in the context of phenytoin-experimentally induced TOICS. Thirty male albino rats were assigned randomly to three equal groups; control, phenytoin (PHT) and PHT+ AuNPs groups. Cerebellar sections were examined histopathologically, ultra-structurally and immunohistochemically for GFAP and p-Tau. Cerebellar tissues were evaluated for TNF-α, IL-1β, MDA, CAT, SOD and CREB mRNA. Our data confirmed observable amelioration of histopathological and ultrastructural cerebellar alterations of Purkinje and granule cells after AuNPs cotreatment. Histomorphometric measures revealed AuNPs-induced significant downregulation of p-Tau and GFAP immune-expression. Concurrently, TNF-α, IL-1β, MDA were significantly quenched in cerebellar tissues after AuNPs cotreatment, on contrary to notable restoration of CAT, SOD and CREB mRNA levels. These outcomes confirm that AuNPs hold promise as a therapeutic strategy for TOICS, warranting further exploration of their mechanisms and clinical applications in cerebellar disorders.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102725"},"PeriodicalIF":2.7,"publicationDate":"2025-01-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012222","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-11DOI: 10.1016/j.tice.2025.102737
J F Raspeño-García, S González-Granero, V Herranz-Pérez, A Cózar-Cuesta, E Artacho-Pérula, R Insausti, J M García-Verdugo, C de la Rosa-Prieto
The mammalian olfactory system is responsible for processing environmental chemical stimuli and comprises several structures, including the olfactory epithelium, olfactory bulb, olfactory peduncle (OP), and olfactory cortices. Despite the critical role played by the OP in the conduction of olfactory information, it has remained understudied. In this work, optical, confocal, and electron microscopy were employed to examine the anatomy, histology, and ultrastructure of six human OP specimens (ages 37-84 years). Three concentric layers were identified in coronal sections: the external layer (EL), the axonal layer (AL), and the internal layer (IL). Immunohistochemistry revealed the distribution of neurons and glial cells throughout the OP. Two neuronal morphologies were observed: granule cells and larger pyramidal cells, the latter associated with projection neurons of the anterior olfactory nucleus. Astrocytes were uniformly distributed with a more radial morphology in the EL. Oligodendrocytes were mainly located in the AL. Blood vessels (BVs) were evenly distributed along the OP, with a mean luminal area of 82.9 µm² and a density of 1.26 %, with a significant increase in the IL. Corpora amylacea (CA) were abundant, with an average size of 49.3 µm² and a density of 3.23 %. CA clustered near BVs, particularly at tissue edges, with both size and density increasing with age. Notably, CA showed strong associations with astrocytes. This study provides the first detailed qualitative and quantitative data on the internal organization of the human OP, which may contribute to a better understanding of the pathophysiology of some neuropathological disorders.
{"title":"Anatomy, histology and ultrastructure of the adult human olfactory peduncle: Blood vessel and corpora amylacea assessment.","authors":"J F Raspeño-García, S González-Granero, V Herranz-Pérez, A Cózar-Cuesta, E Artacho-Pérula, R Insausti, J M García-Verdugo, C de la Rosa-Prieto","doi":"10.1016/j.tice.2025.102737","DOIUrl":"https://doi.org/10.1016/j.tice.2025.102737","url":null,"abstract":"<p><p>The mammalian olfactory system is responsible for processing environmental chemical stimuli and comprises several structures, including the olfactory epithelium, olfactory bulb, olfactory peduncle (OP), and olfactory cortices. Despite the critical role played by the OP in the conduction of olfactory information, it has remained understudied. In this work, optical, confocal, and electron microscopy were employed to examine the anatomy, histology, and ultrastructure of six human OP specimens (ages 37-84 years). Three concentric layers were identified in coronal sections: the external layer (EL), the axonal layer (AL), and the internal layer (IL). Immunohistochemistry revealed the distribution of neurons and glial cells throughout the OP. Two neuronal morphologies were observed: granule cells and larger pyramidal cells, the latter associated with projection neurons of the anterior olfactory nucleus. Astrocytes were uniformly distributed with a more radial morphology in the EL. Oligodendrocytes were mainly located in the AL. Blood vessels (BVs) were evenly distributed along the OP, with a mean luminal area of 82.9 µm² and a density of 1.26 %, with a significant increase in the IL. Corpora amylacea (CA) were abundant, with an average size of 49.3 µm² and a density of 3.23 %. CA clustered near BVs, particularly at tissue edges, with both size and density increasing with age. Notably, CA showed strong associations with astrocytes. This study provides the first detailed qualitative and quantitative data on the internal organization of the human OP, which may contribute to a better understanding of the pathophysiology of some neuropathological disorders.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102737"},"PeriodicalIF":2.7,"publicationDate":"2025-01-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012177","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}