Pub Date : 2025-01-07DOI: 10.1016/j.tice.2025.102722
Bingjie Fan, Lili Yin, A 'ni Wang, Fei Li, Shuguang Han
Background: Diabetes mellitus (DM), a chronic metabolic disease, is characterized by long-term hyperglycemia resulting from the defect of insulin production and insulin resistance. The damage and dysfunction of pancreatic β-cells is a main link in DM development.
Methods: In this work, pancreatic β-cell line INS-1E cells were exposed to 30 mM glucose for 48 h to construct an in vitro DM model. For gain-of-function experiments, HG-treated INS-1E cells were transfected with Oe-PIM1 to thoroughly discuss the biological role of PIM1 in HG-injured pancreatic β-cells. Furthermore, to probe into whether JNK/p38 signaling involved in the protective role of PIM1 in HG-injured pancreatic β-cells, HG-treated INS-1E cells were pre-treated with a JNK activator anisomycin (0.01 μM) for 1 h for rescue experiments.
Results: It was verified that HG treatment obviously downregulated PIM1 expression in INS-1E cells. PIM1 overexpression enhanced insulin secretion, inhibited ferroptosis and strengthened PINK1/Parkin-mediated mitophagy of HG-treated INS-1E cells. PIM1 overexpression inactivated JNK/p38 signaling pathway in HG-treated INS-1E cells. Activation of JNK/p38 signaling pathway partially abolished the strengthening effects of PIM1 overexpression on PINK1/Parkin-mediated mitophagy in HG-treated INS-1E cells. Upregulation of PIM1 strengthened PINK1/Parkin-mediated mitophagy in HG-injured pancreatic β-cells via inactivating JNK/p38 signaling pathway. Besides, activation of JNK/p38 signaling pathway partially abolished the enhancing effects of PIM1 overexpression on insulin secretion and the suppressive effects of PIM1 overexpression on ferroptosis in HG-treated INS-1E cells. Upregulation of PIM1 enhanced insulin secretion and inhibited ferroptosis in HG-injured pancreatic β-cells via inactivating JNK/p38 signaling pathway.
Conclusion: In a word, upregulation of PIM1 may alleviate HG-induced pancreatic β-cell injury through strengthening PINK1/Parkin-mediated mitophagy via inactivating JNK/p38 signaling pathway.
{"title":"PIM1 enhances insulin secretion and inhibits ferroptosis of high glucose-induced pancreatic β-cells through strengthening PINK1/Parkin-mediated mitophagy via inactivating JNK/p38 signaling pathway.","authors":"Bingjie Fan, Lili Yin, A 'ni Wang, Fei Li, Shuguang Han","doi":"10.1016/j.tice.2025.102722","DOIUrl":"https://doi.org/10.1016/j.tice.2025.102722","url":null,"abstract":"<p><strong>Background: </strong>Diabetes mellitus (DM), a chronic metabolic disease, is characterized by long-term hyperglycemia resulting from the defect of insulin production and insulin resistance. The damage and dysfunction of pancreatic β-cells is a main link in DM development.</p><p><strong>Methods: </strong>In this work, pancreatic β-cell line INS-1E cells were exposed to 30 mM glucose for 48 h to construct an in vitro DM model. For gain-of-function experiments, HG-treated INS-1E cells were transfected with Oe-PIM1 to thoroughly discuss the biological role of PIM1 in HG-injured pancreatic β-cells. Furthermore, to probe into whether JNK/p38 signaling involved in the protective role of PIM1 in HG-injured pancreatic β-cells, HG-treated INS-1E cells were pre-treated with a JNK activator anisomycin (0.01 μM) for 1 h for rescue experiments.</p><p><strong>Results: </strong>It was verified that HG treatment obviously downregulated PIM1 expression in INS-1E cells. PIM1 overexpression enhanced insulin secretion, inhibited ferroptosis and strengthened PINK1/Parkin-mediated mitophagy of HG-treated INS-1E cells. PIM1 overexpression inactivated JNK/p38 signaling pathway in HG-treated INS-1E cells. Activation of JNK/p38 signaling pathway partially abolished the strengthening effects of PIM1 overexpression on PINK1/Parkin-mediated mitophagy in HG-treated INS-1E cells. Upregulation of PIM1 strengthened PINK1/Parkin-mediated mitophagy in HG-injured pancreatic β-cells via inactivating JNK/p38 signaling pathway. Besides, activation of JNK/p38 signaling pathway partially abolished the enhancing effects of PIM1 overexpression on insulin secretion and the suppressive effects of PIM1 overexpression on ferroptosis in HG-treated INS-1E cells. Upregulation of PIM1 enhanced insulin secretion and inhibited ferroptosis in HG-injured pancreatic β-cells via inactivating JNK/p38 signaling pathway.</p><p><strong>Conclusion: </strong>In a word, upregulation of PIM1 may alleviate HG-induced pancreatic β-cell injury through strengthening PINK1/Parkin-mediated mitophagy via inactivating JNK/p38 signaling pathway.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102722"},"PeriodicalIF":2.7,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012286","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 : 2025-01-07DOI: 10.1016/j.tice.2025.102724
Sherein F El-Sayed, Samar Mortada Mahmoud, Walaa Samy, Reham M Wahid, Aliaa Talaat, Sara G Seada
Background: The prevalence of gastric ulcers has grown significantly in the modern era affecting 10 % of global population. Aspirin downregulates gastrokines 1(GKN1) expression in gastric mucosa and GKN1 down-regulation results in gastric cancer. Vitamin D3 (Vit.D3) has anti-inflammatory and antioxidant effects.
Aim: Study the gastroprotective impact of Vit.D3 following aspirin-induced gastric injury in relation to gastrokines and investigate the possible underlying mechanisms.
Materials and methods: 24 rats were divided into 4 groups: control, Vit.D3 supplemented normal, aspirin-induced gastric injury, and Vit.D3 supplemented gastric injury groups. Some oxidative stress markers with gene expression of GKN1&2, mucin 5AC (Muc5ac) and NLR family pyrin domain containing 3 (NLRP3) in the gastric tissue were done. Histopathological and immunohistochemical study of E-Cadherin, nuclear factor kappa beta (NFκB), and metalloproteinase-9 (MMP-9) in the stomach mucosa were identified.
Results: Vit.D3 supplementation significantly upregulated E-Cadherin, GSH, GKN1 and Muc5ac in the gastric tissue. Also, it improved the morphology, histology of gastric tissue, by alleviating oxidative stress and NFκB, MMP-9 and down regulation of inflammasome (NLRP3).
Conclusion: Vitamin D3 has a potential protective effect against aspirin -induced gastric injury via upregulating gastrokine1 and E-cadherin and down regulation of NFKB/MMP-9 signaling pathway and NLRP3 inflammasome.
{"title":"Vitamin D3 mitigates aspirin-induced gastric injury by modulating gastrokines, E-cadherin, and inhibiting NLRP3 and NF-κB/MMP-9 signaling pathway.","authors":"Sherein F El-Sayed, Samar Mortada Mahmoud, Walaa Samy, Reham M Wahid, Aliaa Talaat, Sara G Seada","doi":"10.1016/j.tice.2025.102724","DOIUrl":"https://doi.org/10.1016/j.tice.2025.102724","url":null,"abstract":"<p><strong>Background: </strong>The prevalence of gastric ulcers has grown significantly in the modern era affecting 10 % of global population. Aspirin downregulates gastrokines 1(GKN1) expression in gastric mucosa and GKN1 down-regulation results in gastric cancer. Vitamin D3 (Vit.D3) has anti-inflammatory and antioxidant effects.</p><p><strong>Aim: </strong>Study the gastroprotective impact of Vit.D3 following aspirin-induced gastric injury in relation to gastrokines and investigate the possible underlying mechanisms.</p><p><strong>Materials and methods: </strong>24 rats were divided into 4 groups: control, Vit.D3 supplemented normal, aspirin-induced gastric injury, and Vit.D3 supplemented gastric injury groups. Some oxidative stress markers with gene expression of GKN1&2, mucin 5AC (Muc5ac) and NLR family pyrin domain containing 3 (NLRP3) in the gastric tissue were done. Histopathological and immunohistochemical study of E-Cadherin, nuclear factor kappa beta (NFκB), and metalloproteinase-9 (MMP-9) in the stomach mucosa were identified.</p><p><strong>Results: </strong>Vit.D3 supplementation significantly upregulated E-Cadherin, GSH, GKN1 and Muc5ac in the gastric tissue. Also, it improved the morphology, histology of gastric tissue, by alleviating oxidative stress and NFκB, MMP-9 and down regulation of inflammasome (NLRP3).</p><p><strong>Conclusion: </strong>Vitamin D3 has a potential protective effect against aspirin -induced gastric injury via upregulating gastrokine1 and E-cadherin and down regulation of NFKB/MMP-9 signaling pathway and NLRP3 inflammasome.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102724"},"PeriodicalIF":2.7,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012292","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 : 2025-01-07DOI: 10.1016/j.tice.2025.102728
Mohammed A Alshehri, Mohammed Alissa, Abdullah Alghamdi
Cyclophosphamide (CP) is an alkylating chemotherapy agent that induces liver toxicity by cross-linking DNA, causing cell apoptosis. While CP is effective in cancer treatment, its side effects on the liver are significant. Recent studies have indicated that antioxidants, such as resveratrol, may reduce these toxic effects. In this study, we aimed to investigate the role of resveratrol in mitigating CP-induced hepatic apoptosis, oxidative stress, and inflammation in rats. Twenty male mature Sprague-Dawley rats were divided into 4 groups of equal size: control group, Resveratrol group which received resveratrol (20 mg/kg) for 15 consecutive days, CP group which received CP as a single dose (150 mg/kg) on day 16, and CP+Resveratrol group which was similar of the resveratrol and CP groups. Tissue samples were obtained for the histological, immunohistochemical, biochemical, and molecular evaluations. Findings showed that treatment with CP significantly decreased the total liver volume, numerical density of hepatocytes, length density of sinusoidals, and concentrations of antioxidative biomarkers (GSH and SOD). However, the CP+Resveratrol group exhibited notably greater values in these parameters compared to the CP group. Additionally, CP treatment resulted in a significant increase in serum levels of AST and ALT, higher numerical density of Kupffer cells, increased densities of apoptotic cells (increased Bax and caspase-3, and decreased Bcl-2 expression levels), elevated levels of MDA, and upregulated inflammatory genes (IL-1β and TNF-α). In contrast, co-treatment with resveratrol significantly reduced these parameters, suggesting its protective effects against CP-induced liver toxicity. We conclude that giving resveratrol can attenuate apoptosis, oxidative stress, inflammation, and histological alterations in the liver induced by CP toxicity.
{"title":"Resveratrol attenuates cyclophosphamide-induced hepatic apoptosis in association with the inhibition of oxidative stress and inflammation in a rat model of acute liver injury.","authors":"Mohammed A Alshehri, Mohammed Alissa, Abdullah Alghamdi","doi":"10.1016/j.tice.2025.102728","DOIUrl":"https://doi.org/10.1016/j.tice.2025.102728","url":null,"abstract":"<p><p>Cyclophosphamide (CP) is an alkylating chemotherapy agent that induces liver toxicity by cross-linking DNA, causing cell apoptosis. While CP is effective in cancer treatment, its side effects on the liver are significant. Recent studies have indicated that antioxidants, such as resveratrol, may reduce these toxic effects. In this study, we aimed to investigate the role of resveratrol in mitigating CP-induced hepatic apoptosis, oxidative stress, and inflammation in rats. Twenty male mature Sprague-Dawley rats were divided into 4 groups of equal size: control group, Resveratrol group which received resveratrol (20 mg/kg) for 15 consecutive days, CP group which received CP as a single dose (150 mg/kg) on day 16, and CP+Resveratrol group which was similar of the resveratrol and CP groups. Tissue samples were obtained for the histological, immunohistochemical, biochemical, and molecular evaluations. Findings showed that treatment with CP significantly decreased the total liver volume, numerical density of hepatocytes, length density of sinusoidals, and concentrations of antioxidative biomarkers (GSH and SOD). However, the CP+Resveratrol group exhibited notably greater values in these parameters compared to the CP group. Additionally, CP treatment resulted in a significant increase in serum levels of AST and ALT, higher numerical density of Kupffer cells, increased densities of apoptotic cells (increased Bax and caspase-3, and decreased Bcl-2 expression levels), elevated levels of MDA, and upregulated inflammatory genes (IL-1β and TNF-α). In contrast, co-treatment with resveratrol significantly reduced these parameters, suggesting its protective effects against CP-induced liver toxicity. We conclude that giving resveratrol can attenuate apoptosis, oxidative stress, inflammation, and histological alterations in the liver induced by CP toxicity.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102728"},"PeriodicalIF":2.7,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142984855","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 : 2025-01-06DOI: 10.1016/j.tice.2024.102719
Ludmila M Sencha, Maria A Karpova, Olga E Dobrynina, Irina V Balalaeva
The extracellular matrix (ECM) and its primary chemical components, including collagen, play a pivotal role in carcinogenesis and tumor progression. The ECM actively regulates cell proliferation, migration, and, importantly, resistance to various adverse factors. It is widely recognized as a key factor in modifying the resistance of tumor cells to various treatment modalities and cytotoxic compounds. However, the role of the ECM in tumor cell adaptation to nutritional deficiencies and hypoxic conditions remains significantly less studied. Since it is generally accepted that tumor cells resistance increases when cultured in a three-dimensional matrix, we sought to experimentally test the universality of this statement. In this work, we analyzed the responses of tumor cells with varying origins and proliferative activities, including human bladder carcinoma, epidermoid carcinoma, and ovarian carcinoma, to deprivation of serum, glucose and oxygen. We compared cell resistance to suboptimal conditions when cultured in a monolayer on tissue culture (TC)-treated polystyrene, on collagen-coated surfaces, or within a three-dimensional hydrogel composed of collagen type I. All three cell lines were stably transfected with fluorescent protein genes. To register the cell growth dynamics, we used a fluorescence-based technique that allows long-term quantitative observations without disrupting the hydrogel. The analyzed cell lines demonstrated different patterns of relative sensitivity to suboptimal conditions. We revealed that the direction and intensity of the collagen matrix effect depend on the cell type. Slowly proliferating ovarian carcinoma cells showed no noticeable changes in their behavior when cultured in a gel compared to a monolayer. In the case of bladder carcinoma, we registered predominantly resistance-stimulating effect of the collagen matrix, but it was significant only under serum deprivation. The most pronounced effect of collagen was registered for epidermoid carcinoma. Importantly, this effect was ambivalent: gel-embedded cells demonstrated significantly enhanced resistance to serum deprivation, but, at the same time, they were more responsive to glucose starvation and hypoxic conditions. We attribute the registered phenomenon to the individual characteristics of tumor cells with different origins and metabolic activities.
{"title":"Cell-type dependent effect of 3D collagen matrix on cancer cell resistance to suboptimal conditions: the case of serum deprivation, glucose starvation, and hypoxia.","authors":"Ludmila M Sencha, Maria A Karpova, Olga E Dobrynina, Irina V Balalaeva","doi":"10.1016/j.tice.2024.102719","DOIUrl":"https://doi.org/10.1016/j.tice.2024.102719","url":null,"abstract":"<p><p>The extracellular matrix (ECM) and its primary chemical components, including collagen, play a pivotal role in carcinogenesis and tumor progression. The ECM actively regulates cell proliferation, migration, and, importantly, resistance to various adverse factors. It is widely recognized as a key factor in modifying the resistance of tumor cells to various treatment modalities and cytotoxic compounds. However, the role of the ECM in tumor cell adaptation to nutritional deficiencies and hypoxic conditions remains significantly less studied. Since it is generally accepted that tumor cells resistance increases when cultured in a three-dimensional matrix, we sought to experimentally test the universality of this statement. In this work, we analyzed the responses of tumor cells with varying origins and proliferative activities, including human bladder carcinoma, epidermoid carcinoma, and ovarian carcinoma, to deprivation of serum, glucose and oxygen. We compared cell resistance to suboptimal conditions when cultured in a monolayer on tissue culture (TC)-treated polystyrene, on collagen-coated surfaces, or within a three-dimensional hydrogel composed of collagen type I. All three cell lines were stably transfected with fluorescent protein genes. To register the cell growth dynamics, we used a fluorescence-based technique that allows long-term quantitative observations without disrupting the hydrogel. The analyzed cell lines demonstrated different patterns of relative sensitivity to suboptimal conditions. We revealed that the direction and intensity of the collagen matrix effect depend on the cell type. Slowly proliferating ovarian carcinoma cells showed no noticeable changes in their behavior when cultured in a gel compared to a monolayer. In the case of bladder carcinoma, we registered predominantly resistance-stimulating effect of the collagen matrix, but it was significant only under serum deprivation. The most pronounced effect of collagen was registered for epidermoid carcinoma. Importantly, this effect was ambivalent: gel-embedded cells demonstrated significantly enhanced resistance to serum deprivation, but, at the same time, they were more responsive to glucose starvation and hypoxic conditions. We attribute the registered phenomenon to the individual characteristics of tumor cells with different origins and metabolic activities.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102719"},"PeriodicalIF":2.7,"publicationDate":"2025-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012219","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 : 2025-01-05DOI: 10.1016/j.tice.2024.102720
Reihaneh Zare, Rita ArabSolghar, Abbas Behzad Behbahni, Farahnaz Zare, Ali Kheirandish, Fatemeh Safari
Malignant pheochromocytomas are infrequent tumors that have a poorer prognosis compared to their benign counterparts. The administration of chemotherapy to patients with pheochromocytoma can result in adverse side effects and a reduced life quality. Alternative and more targeted treatment strategies, such as gene therapy significantly improve the patients' survival rate and life expectancy. Caspase-3 is a key apoptosis regulator activated by cancer treatments. Recent research shows it also influences tumor relapse and angiogenesis, complicating its role in cancer progression. Further exploration of Caspase-3's diverse functions is needed to clarify its impact on cancer development. In this study, we established Caspase-3 over expressed pheochromocytoma cell line by the use of lentiviral vector technology. Caspase 3 over expression by up to 3fold led to increase in cell proliferation by up to 12 %. Moreover, increasing in Caspase 3 level of expression resulted in more invasiveness and metastasis. By this way, the wound closure percentage for PC-12 Casp3 + cells reached 76.2 %, which is significantly higher compared to the 52.8 % observed in mock cells. Casp3 + cells were also significantly more sensitive to cisplatin than mock cells with Ic50 of 158.4 μM and 219.5uM respectively according to MTT assay which confirmed by apoptosis assay. Hence, targeting Caspase-3 as a therapeutic approach may enhance the cancer cell sensitivity to chemotherapy, but also increase the cancer cell proliferation, metastases and invasion which may works as a double edge sword. CONCLUSION: understanding the effects of Caspase 3 over expression on cancer cells could inspire innovative therapies targeting its non-apoptotic actions, potentially improving cancer treatment outcomes.
{"title":"Exploring Caspase-3 overexpression in pheochromocytoma cells: Implications for cancer therapy.","authors":"Reihaneh Zare, Rita ArabSolghar, Abbas Behzad Behbahni, Farahnaz Zare, Ali Kheirandish, Fatemeh Safari","doi":"10.1016/j.tice.2024.102720","DOIUrl":"https://doi.org/10.1016/j.tice.2024.102720","url":null,"abstract":"<p><p>Malignant pheochromocytomas are infrequent tumors that have a poorer prognosis compared to their benign counterparts. The administration of chemotherapy to patients with pheochromocytoma can result in adverse side effects and a reduced life quality. Alternative and more targeted treatment strategies, such as gene therapy significantly improve the patients' survival rate and life expectancy. Caspase-3 is a key apoptosis regulator activated by cancer treatments. Recent research shows it also influences tumor relapse and angiogenesis, complicating its role in cancer progression. Further exploration of Caspase-3's diverse functions is needed to clarify its impact on cancer development. In this study, we established Caspase-3 over expressed pheochromocytoma cell line by the use of lentiviral vector technology. Caspase 3 over expression by up to 3fold led to increase in cell proliferation by up to 12 %. Moreover, increasing in Caspase 3 level of expression resulted in more invasiveness and metastasis. By this way, the wound closure percentage for PC-12 Casp3 + cells reached 76.2 %, which is significantly higher compared to the 52.8 % observed in mock cells. Casp3 + cells were also significantly more sensitive to cisplatin than mock cells with Ic50 of 158.4 μM and 219.5uM respectively according to MTT assay which confirmed by apoptosis assay. Hence, targeting Caspase-3 as a therapeutic approach may enhance the cancer cell sensitivity to chemotherapy, but also increase the cancer cell proliferation, metastases and invasion which may works as a double edge sword. CONCLUSION: understanding the effects of Caspase 3 over expression on cancer cells could inspire innovative therapies targeting its non-apoptotic actions, potentially improving cancer treatment outcomes.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102720"},"PeriodicalIF":2.7,"publicationDate":"2025-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142955693","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 : 2025-01-04DOI: 10.1016/j.tice.2024.102717
Victoria Simões Bernardo, Flaviene Felix Torres, Ana Clara Albertin Zucão, Nayara Alves Chaves, Ilana Luize Rocha Santana, Danilo Grünig Humberto da Silva
Sickle cell disease (SCD) is a hereditary hemolytic anemia associated with the alteration of the membrane composition of the sickle erythrocytes, the loss of glycolysis, dysregulation of the pyruvate phosphatase pathway, and changes in nucleotide metabolism of the sickle red blood cell (RBC). This review provides a comprehensive overview of the impact of the presence of Hb S, which leads to the disruption of the normal RBC metabolism. The intricate interplay between the redox and energetic balance in erythrocytic cells, where the glycolysis, pentose phosphate pathway, and methemoglobin reductase pathways are all altered in sickle RBC, is a key focus. Moreover, this review summarizes the current knowledge about the disease-modifying agents and their action mechanisms based on the sickle RBC alterations previously mentioned (i.e., their association with beneficial effects on the sickle cells' membrane, to their RBCs' energy metabolism, and to their oxidative status). Therefore, providing a comprehensive understanding of how sickle cells cope with the disruption of metabolic homeostasis and the most promising therapeutic agents able to ameliorate the various consequences of abnormal sickle RBC alterations.
{"title":"Disrupted homeostasis in sickle cells: Expanding the comprehension of metabolism adaptation and related therapeutic strategies.","authors":"Victoria Simões Bernardo, Flaviene Felix Torres, Ana Clara Albertin Zucão, Nayara Alves Chaves, Ilana Luize Rocha Santana, Danilo Grünig Humberto da Silva","doi":"10.1016/j.tice.2024.102717","DOIUrl":"https://doi.org/10.1016/j.tice.2024.102717","url":null,"abstract":"<p><p>Sickle cell disease (SCD) is a hereditary hemolytic anemia associated with the alteration of the membrane composition of the sickle erythrocytes, the loss of glycolysis, dysregulation of the pyruvate phosphatase pathway, and changes in nucleotide metabolism of the sickle red blood cell (RBC). This review provides a comprehensive overview of the impact of the presence of Hb S, which leads to the disruption of the normal RBC metabolism. The intricate interplay between the redox and energetic balance in erythrocytic cells, where the glycolysis, pentose phosphate pathway, and methemoglobin reductase pathways are all altered in sickle RBC, is a key focus. Moreover, this review summarizes the current knowledge about the disease-modifying agents and their action mechanisms based on the sickle RBC alterations previously mentioned (i.e., their association with beneficial effects on the sickle cells' membrane, to their RBCs' energy metabolism, and to their oxidative status). Therefore, providing a comprehensive understanding of how sickle cells cope with the disruption of metabolic homeostasis and the most promising therapeutic agents able to ameliorate the various consequences of abnormal sickle RBC alterations.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102717"},"PeriodicalIF":2.7,"publicationDate":"2025-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142979964","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 : 2025-01-03DOI: 10.1016/j.tice.2024.102718
Aylar Borhan, Ali Bagherlou, Mohammad B Ghayour
Background: Carnosic acid (CA) has potential anti-cancer properties, but its effectiveness can be improved by combining it with Folic acid (FA). This research aimed to evaluate the impact of CA and CA-FA conjugate on breast cancer cell lines (MCF-7, MDA-MB-231, and MCA10).
Materials and methods: The viability of the cell lines was measured using the MTT assay, and the IC₅₀ was determined to compare the cytotoxicity of CA and CA-FA. The process of programmed cell death was investigated by utilizing Annexin V/PI staining, measuring caspase-3/7 activity, and real-time PCR for apoptotic gene expression. Reactive oxygen species (ROS) were also assessed to determine the extent of oxidative stress.
Results: CA significantly decreased the viability of MCF-7 and MDA-MB-231 cells depending on the dosage, with CA-FA exhibiting enhanced cytotoxicity, particularly in MDA-MB-231 cells. The evaluation of IC₅₀ confirmed that conjugation with FA reduced the IC₅₀ of CA. Apoptosis analysis demonstrated increased apoptosis rates in MCF-7 and MDA-MB-231 cells exposed to treatment with CA and CA-FA, while MCA10 cells showed minimal effects. Caspase-3/7 activity was notably higher in CA-FA-treated cells. Gene expression analysis revealed elevated pro-apoptotic gene activity and reduced anti-apoptotic gene activity, with CA-FA having a more pronounced effect. Cells subjected to CA-FA treatment exhibited a significant increase in ROS levels.
Conclusion: These findings suggest that CA conjugation with FA enhances its cytotoxic effects and promotes apoptosis through increased apoptosis and ROS production. The research emphasizes the promise of CA-FA as a focused treatment approach for aggressive forms of breast cancer, underscoring the need for additional exploration of its practical uses in clinical settings.
{"title":"Evaluating the anticancer effects of carnosic acid against breast cancer: An In Vitro investigation.","authors":"Aylar Borhan, Ali Bagherlou, Mohammad B Ghayour","doi":"10.1016/j.tice.2024.102718","DOIUrl":"https://doi.org/10.1016/j.tice.2024.102718","url":null,"abstract":"<p><strong>Background: </strong>Carnosic acid (CA) has potential anti-cancer properties, but its effectiveness can be improved by combining it with Folic acid (FA). This research aimed to evaluate the impact of CA and CA-FA conjugate on breast cancer cell lines (MCF-7, MDA-MB-231, and MCA10).</p><p><strong>Materials and methods: </strong>The viability of the cell lines was measured using the MTT assay, and the IC₅₀ was determined to compare the cytotoxicity of CA and CA-FA. The process of programmed cell death was investigated by utilizing Annexin V/PI staining, measuring caspase-3/7 activity, and real-time PCR for apoptotic gene expression. Reactive oxygen species (ROS) were also assessed to determine the extent of oxidative stress.</p><p><strong>Results: </strong>CA significantly decreased the viability of MCF-7 and MDA-MB-231 cells depending on the dosage, with CA-FA exhibiting enhanced cytotoxicity, particularly in MDA-MB-231 cells. The evaluation of IC₅₀ confirmed that conjugation with FA reduced the IC₅₀ of CA. Apoptosis analysis demonstrated increased apoptosis rates in MCF-7 and MDA-MB-231 cells exposed to treatment with CA and CA-FA, while MCA10 cells showed minimal effects. Caspase-3/7 activity was notably higher in CA-FA-treated cells. Gene expression analysis revealed elevated pro-apoptotic gene activity and reduced anti-apoptotic gene activity, with CA-FA having a more pronounced effect. Cells subjected to CA-FA treatment exhibited a significant increase in ROS levels.</p><p><strong>Conclusion: </strong>These findings suggest that CA conjugation with FA enhances its cytotoxic effects and promotes apoptosis through increased apoptosis and ROS production. The research emphasizes the promise of CA-FA as a focused treatment approach for aggressive forms of breast cancer, underscoring the need for additional exploration of its practical uses in clinical settings.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102718"},"PeriodicalIF":2.7,"publicationDate":"2025-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142955692","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}
Mechanical loading plays a pivotal role in regulating bone anabolic processes. Understanding the optimal mechanical loading parameters for cellular responses is critical for advancing strategies in orthopedic bioreactor-based bone tissue engineering. This study developed a poly (sorbitol sebacate) (PSS) filmscaffold with a sorbitol-to-sebacic acid molar ratio of 1:4. The scaffold underwent extensive characterization, including physical and mechanical property evaluations, biocompatibility assessments, and cell adhesion analysis. The Young's modulus of the PSS polymer was determined to be 6.81 ± 0.44 MPa under dry conditions, 6.37 ± 1.09 MPa in a wet state, and 6.38 ± 0.71 MPa after ethanol washing (70 %). The average contact angle of the PSS film was measured at 88.806 ± 1.644°, indicating moderate hydrophilicity. To investigate the osteogenic potential, a fluid flow inducing a shear stress of 1 Pa at a frequency of 1 Hz was applied to mesenchymal stem cells (MSCs) cultured on the PSS scaffold. Cells were exposed to dynamic fluid flow for one hour daily on days 4, 5, 6, and 7 of culture, followed by a static culture period of 14 days. The expression of osteogenic differentiation markers, including osteopontin (OPN), osteocalcin (OCN), type I collagen, and calcium deposition, was significantly elevated under dynamic conditions compared to static culture. This study highlights the importance of mechanical stimulation in enhancing MSC osteogenesis and underscores the osteoconductive properties of the PSS scaffold. These findings provide valuable insights into scaffold design and mechanical loading strategies for laboratory-based bone tissue engineering applications.
{"title":"Osteogenic differentiation of mesenchymal stem cell on poly sorbitol sebacate scaffold under shear stress in a bioreactor.","authors":"Fatemeh Abbasloo, Bahman Vahidi, Mohammad-Mehdi Khani, Faraz Sigaroodi, Reza Ramezani Sarbandi","doi":"10.1016/j.tice.2024.102715","DOIUrl":"https://doi.org/10.1016/j.tice.2024.102715","url":null,"abstract":"<p><p>Mechanical loading plays a pivotal role in regulating bone anabolic processes. Understanding the optimal mechanical loading parameters for cellular responses is critical for advancing strategies in orthopedic bioreactor-based bone tissue engineering. This study developed a poly (sorbitol sebacate) (PSS) filmscaffold with a sorbitol-to-sebacic acid molar ratio of 1:4. The scaffold underwent extensive characterization, including physical and mechanical property evaluations, biocompatibility assessments, and cell adhesion analysis. The Young's modulus of the PSS polymer was determined to be 6.81 ± 0.44 MPa under dry conditions, 6.37 ± 1.09 MPa in a wet state, and 6.38 ± 0.71 MPa after ethanol washing (70 %). The average contact angle of the PSS film was measured at 88.806 ± 1.644°, indicating moderate hydrophilicity. To investigate the osteogenic potential, a fluid flow inducing a shear stress of 1 Pa at a frequency of 1 Hz was applied to mesenchymal stem cells (MSCs) cultured on the PSS scaffold. Cells were exposed to dynamic fluid flow for one hour daily on days 4, 5, 6, and 7 of culture, followed by a static culture period of 14 days. The expression of osteogenic differentiation markers, including osteopontin (OPN), osteocalcin (OCN), type I collagen, and calcium deposition, was significantly elevated under dynamic conditions compared to static culture. This study highlights the importance of mechanical stimulation in enhancing MSC osteogenesis and underscores the osteoconductive properties of the PSS scaffold. These findings provide valuable insights into scaffold design and mechanical loading strategies for laboratory-based bone tissue engineering applications.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102715"},"PeriodicalIF":2.7,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142955694","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 : 2024-12-31DOI: 10.1016/j.tice.2024.102712
Liang Wu, Juan Wang
Allergic rhinitis (AR), common in children and adolescents, involves Lysophosphatidylcholine acyltransferase 1 (LPCAT1) catalyzing surfactant lipid biosynthesis and suppressing endoplasmic reticulum expression. However, the precise mechanism underlying the impact of LPCAT1 on epithelial cell damage in AR remains elusive. Hence, the present investigation elucidated the potential effect of LPCAT1 on epithelial cell damage in AR by inhibiting endoplasmic reticulum stress. To assess cell viability, CCK8 assay was employed. Additionally, western blotting was utilized to evaluate the expression of endoplasmic reticulum stress-associated proteins ATF6, CHOP, p-eIF2α, p-IRE1, and LPCAT1. Subsequently, an interference plasmid targeting LPCAT1 was constructed, and western blot analysis was conducted to determine interference level of LPCAT1. An ELISA assay was employed to quantify the concentrations of TNFα, IL-1β, IL-6, GM-CSF, and eotaxin. Additionally, flow cytometry and western blotting techniques were utilized to evaluate cellular apoptosis, whereas immunofluorescence staining was applied to detect the expression levels of ZO-1. Our findings indicated that IL-13 stimulation resulted in an elevated expression of ER stress proteins and LPCAT1 in nasal mucosal epithelial cells. Furthermore, LPCAT1 interference diminished the expression of inflammatory mediators, apoptosis markers, barrier disruption indicators, and ER stress proteins in IL-13-stimulated nasal mucosal epithelial cells. Further, by inhibiting ER stress, LPCAT1 interference diminished the expression of inflammatory factors, apoptosis, and barrier damage in nasal mucosal epithelial cells stimulated by IL-13. Concisely, LPCAT1 ameliorates AR-induced inflammation, apoptosis, and barrier impairment in nasal mucosal epithelial cells by modulating ER stress, implying its potential as a novel therapeutic target for AR.
{"title":"LPCAT1 reduces inflammatory response, apoptosis and barrier damage of nasal mucosal epithelial cells caused by allergic rhinitis through endoplasmic reticulum stress.","authors":"Liang Wu, Juan Wang","doi":"10.1016/j.tice.2024.102712","DOIUrl":"https://doi.org/10.1016/j.tice.2024.102712","url":null,"abstract":"<p><p>Allergic rhinitis (AR), common in children and adolescents, involves Lysophosphatidylcholine acyltransferase 1 (LPCAT1) catalyzing surfactant lipid biosynthesis and suppressing endoplasmic reticulum expression. However, the precise mechanism underlying the impact of LPCAT1 on epithelial cell damage in AR remains elusive. Hence, the present investigation elucidated the potential effect of LPCAT1 on epithelial cell damage in AR by inhibiting endoplasmic reticulum stress. To assess cell viability, CCK8 assay was employed. Additionally, western blotting was utilized to evaluate the expression of endoplasmic reticulum stress-associated proteins ATF6, CHOP, p-eIF2α, p-IRE1, and LPCAT1. Subsequently, an interference plasmid targeting LPCAT1 was constructed, and western blot analysis was conducted to determine interference level of LPCAT1. An ELISA assay was employed to quantify the concentrations of TNFα, IL-1β, IL-6, GM-CSF, and eotaxin. Additionally, flow cytometry and western blotting techniques were utilized to evaluate cellular apoptosis, whereas immunofluorescence staining was applied to detect the expression levels of ZO-1. Our findings indicated that IL-13 stimulation resulted in an elevated expression of ER stress proteins and LPCAT1 in nasal mucosal epithelial cells. Furthermore, LPCAT1 interference diminished the expression of inflammatory mediators, apoptosis markers, barrier disruption indicators, and ER stress proteins in IL-13-stimulated nasal mucosal epithelial cells. Further, by inhibiting ER stress, LPCAT1 interference diminished the expression of inflammatory factors, apoptosis, and barrier damage in nasal mucosal epithelial cells stimulated by IL-13. Concisely, LPCAT1 ameliorates AR-induced inflammation, apoptosis, and barrier impairment in nasal mucosal epithelial cells by modulating ER stress, implying its potential as a novel therapeutic target for AR.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102712"},"PeriodicalIF":2.7,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012171","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 : 2024-12-31DOI: 10.1016/j.tice.2024.102711
Larissa Dos Santos Sebould Marinho, Márcia Cristina Ribeiro Andrade, Cláudia Andréa de Araújo Lopes, Kassia Valéria Gomes Coelho da Silva, Kauet de Matos Gama E Souza, Clarice Machado-Santos
SARS-Cov-2 is a corona virus that causes COVID-19 disease, a viral infection responsible for the pandemic decreed by the World Health Organization in March 2020. Angiotensin-converting enzyme 2 (ACE-2) functions as the main receptor for SARS-Cov-2. The study aimed to detect the expression of ACE-2 in the gastrointestinal tract, kidney, and lung in the rhesus monkeys and squirrel monkeys. The sections from 18 rhesus monkey and 17 squirrel monkeys were incubated with rabbit polyclonal antibody to ACE2 (ab65863). In the lung of the rhesus monkeys, the presence of ACE-2 was noted in the bronchial mucosa of the respiratory epithelium. In the kidney, there was irregular in the proximal convoluted tubules. In the pyloric stomach, duodenum and in the large intestine it was observed on the surface of the lining epithelium. In the lung of the squirrel monkeys, this marking was present in both the ciliated cylindrical and goblet cell sof the bronchi. In the kidney light marking was observed along the surfasse of the cubic epithelium of the proximal convoluted tubules and in the renal glomerulus. No markings were observed throughout the stomach and intense staining was observed along the surfasse of the intestinal epithelium of the duodenum, jejunum and ileum, as well as in the intestinal glands. In our study, we can observe not able differences in the distribution of ACE2 between the two species of primates analysed. These differences must be considered in experimental studies on this disease, which continues to be a topic of notable importance for Public Health.
{"title":"Immunohistochemical identification of ACE-2 (SARS-COV II entry mechanism) in the gastrointestinal tract, kidney and lung of rhesus monkeys (Macaca mulatta) and squirrel monkeys (Saimiri sciureus).","authors":"Larissa Dos Santos Sebould Marinho, Márcia Cristina Ribeiro Andrade, Cláudia Andréa de Araújo Lopes, Kassia Valéria Gomes Coelho da Silva, Kauet de Matos Gama E Souza, Clarice Machado-Santos","doi":"10.1016/j.tice.2024.102711","DOIUrl":"https://doi.org/10.1016/j.tice.2024.102711","url":null,"abstract":"<p><p>SARS-Cov-2 is a corona virus that causes COVID-19 disease, a viral infection responsible for the pandemic decreed by the World Health Organization in March 2020. Angiotensin-converting enzyme 2 (ACE-2) functions as the main receptor for SARS-Cov-2. The study aimed to detect the expression of ACE-2 in the gastrointestinal tract, kidney, and lung in the rhesus monkeys and squirrel monkeys. The sections from 18 rhesus monkey and 17 squirrel monkeys were incubated with rabbit polyclonal antibody to ACE2 (ab65863). In the lung of the rhesus monkeys, the presence of ACE-2 was noted in the bronchial mucosa of the respiratory epithelium. In the kidney, there was irregular in the proximal convoluted tubules. In the pyloric stomach, duodenum and in the large intestine it was observed on the surface of the lining epithelium. In the lung of the squirrel monkeys, this marking was present in both the ciliated cylindrical and goblet cell sof the bronchi. In the kidney light marking was observed along the surfasse of the cubic epithelium of the proximal convoluted tubules and in the renal glomerulus. No markings were observed throughout the stomach and intense staining was observed along the surfasse of the intestinal epithelium of the duodenum, jejunum and ileum, as well as in the intestinal glands. In our study, we can observe not able differences in the distribution of ACE2 between the two species of primates analysed. These differences must be considered in experimental studies on this disease, which continues to be a topic of notable importance for Public Health.</p>","PeriodicalId":23201,"journal":{"name":"Tissue & cell","volume":"93 ","pages":"102711"},"PeriodicalIF":2.7,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142955691","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}