1
|
Quintal Bojórquez NDC, Segura Campos MR. Novel peptides derived from S. hispanica seeds induce selective cytotoxicity on human cancer cells. Food Chem 2024; 460:140470. [PMID: 39032303 DOI: 10.1016/j.foodchem.2024.140470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/04/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
Cancer prevails as one of the major health concerns worldwide due to the consistent rise in incidence and lack of effective therapies. Previous studies identified the peptides KLKKNL, MLKSKR, and KKYRVF from Salvia hispanica seeds and stated their selective anticancer activity. Thus, this study aimed to determine the cell death pathway induced by these peptides on five cancer cell lines (MCF-7, Caco2, HepG2, DU145, and HeLa). Based on the results of this work, it is possible to suggest that KLKKNL primarily induces selective cancer cell death through the apoptotic pathway in the Caco2 and HeLa lines. On the other hand, the peptide KKYRVF reported the highest statistical (p < 0.05) selective cytotoxic effect on the MCF-7, Caco2, HepG2, and DU145 cancer cell lines by induction of the necrotic pathway. These findings offer some understanding of the selective anticancer effect of KLKKNL, MLKSKR, and KKYRVF.
Collapse
Affiliation(s)
- Nidia Del Carmen Quintal Bojórquez
- Facultad de Ingeniería Química, Universidad Autónoma de Yucatán, Periférico Norte Km. 33.5, Tablaje Catastral 13615, Colonia Chuburná de Hidalgo Inn. Mérida, Yucatán, C.P., Mexico
| | - Maira Rubi Segura Campos
- Facultad de Ingeniería Química, Universidad Autónoma de Yucatán, Periférico Norte Km. 33.5, Tablaje Catastral 13615, Colonia Chuburná de Hidalgo Inn. Mérida, Yucatán, C.P., Mexico.
| |
Collapse
|
2
|
Lotfi MS, Rassouli FB. Navigating the complexities of cell death: Insights into accidental and programmed cell death. Tissue Cell 2024; 91:102586. [PMID: 39426124 DOI: 10.1016/j.tice.2024.102586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Cell death is a critical biological phenomenon that can be categorized into accidental cell death (ACD) and programmed cell death (PCD), each exhibiting distinct signaling, mechanistic and morphological characteristics. This paper provides a comprehensive overview of seven types of ACD, including coagulative, liquefactive, caseous, fat, fibrinoid, gangrenous and secondary necrosis, discussing their pathological implications in conditions such as ischemia and inflammation. Additionally, we review eighteen forms of PCD, encompassing autophagy, apoptosis, necroptosis, pyroptosis, paraptosis, ferroptosis, anoikis, entosis, NETosis, eryptosis, parthanatos, mitoptosis, and newly recognized types such as methuosis, autosis, alkaliptosis, oxeiptosis, cuprotosis and erebosis. The implications of these cell death modalities for cellular processes, development, and disease-particularly in the context of neoplastic and neurodegenerative disorders-are also covered. Furthermore, we explore the crosstalk between various forms of PCD, emphasizing how apoptotic mechanisms can influence pathways like necroptosis and pyroptosis. Understanding this interplay is crucial for elucidating cellular responses to stress, as well as for its potential relevance in clinical applications and therapeutic strategies. Future research should focus on clarifying the molecular mechanisms that govern different forms of PCD and their interactions.
Collapse
Affiliation(s)
- Mohammad-Sadegh Lotfi
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Fatemeh B Rassouli
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
3
|
Wulan SN, Anggraini JA, Hidayat W. In Vitro Anticancer Potential of Eugenol on Oral Cancer Cell Lines: A Systematic Review. Malays J Med Sci 2024; 31:10-31. [PMID: 39416730 PMCID: PMC11477475 DOI: 10.21315/mjms2024.31.5.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 06/20/2024] [Indexed: 10/19/2024] Open
Abstract
Oral cancer is one of the most common types of cancer and has become a global health concern. Oral squamous cell carcinoma (OSCC) is the most prevalent form of oral cancer worldwide. Eugenol, an aromatic phenolic compound, exhibits various pharmacological activities, including anticancer effects. Several studies have reported the anticancer activity of eugenol against OSCC via different pathways. However, no current review has discussed the extent of eugenol anticancer research on oral cancer cell lines using in vitro studies. This systematic review aimed to discuss the anticancer potential of eugenol against oral cancer cell lines in vitro. Articles were selected from PubMed, ScienceDirect, SpringerLink and EBSCOhost databases based on specified inclusion and exclusion criteria. Additional articles were identified through manual hand searching. The selection process followed PRISMA guidelines. A risk-of-bias assessment was performed to evaluate the reliability and relevance of the in vitro studies. Thirteen articles with high-quality results were assessed and analysed for further investigation. These studies investigated the ability of eugenol to induce cell death through apoptotic and non-apoptotic pathways, inhibit cell proliferation and affect oxidative stress, contributing to cell death in several oral cancer cell lines. Therefore, eugenol is a potential anticancer agent for OSCC, as it exhibited anticancer activity in oral cancer cell lines in vitro studies.
Collapse
Affiliation(s)
- Shafa Nafisa Wulan
- Undergraduate Dentistry Study Program, Faculty of Dentistry, Universitas Padjadjaran, Indonesia
| | - Jamas Ari Anggraini
- Department of Oral Biology, Faculty of Dentistry, Universitas Padjadjaran, Indonesia
| | - Wahyu Hidayat
- Department of Oral Medicine, Faculty of Dentistry, Universitas Padjadjaran, Indonesia
| |
Collapse
|
4
|
Zheng M, Kong X, Jiang X, Yang Y, Fu S, Wen C, Zhang W, Di W. Qualitative analysis of Fasciola gigantica excretory and secretory products coimmunoprecipitated with buffalo secondary infection sera shows dissimilar components from primary infection sera. Acta Trop 2024; 260:107391. [PMID: 39278520 DOI: 10.1016/j.actatropica.2024.107391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/30/2024] [Accepted: 09/07/2024] [Indexed: 09/18/2024]
Abstract
Buffaloes cannot mount a robust adaptive immune response to secondary infection by Fasciola gigantica. Even if excretory and secretory products (ESPs) exhibit potent immunoregulatory effects during primary infection, research on ESPs in secondary infection is lacking, even though the ESP components that are excreted/secreted during secondary infection are unknown. Therefore, qualitative analysis of ESP during secondary infection was performed and compared with that of primary infection to deepen the recognition of secondary infection and facilitate immunoregulatory molecules screening. Buffaloes were divided into three groups: A (n = 3, noninfected), B (n = 3, primary infection) and C (n = 3, secondary infection). Buffaloes in the primary (0 weeks post infection; wpi) and secondary (-4 and 0 wpi) infection groups were infected with 250 metacercariae by oral administration. Then, sera were collected from groups at different wpi, and interacting proteins were precipitated by coimmunoprecipitation (Co-IP), qualitatively analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS), and annotated by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses to infer their potential functions. In group C, 324 proteins were identified, of which 76 proteins were consistently identified across 7 time points (1, 3, 6, 8, 10, 13, and 16 wpi). Compared with 87 proteins consistently identified in group B, 22 proteins were identified in group C. Meanwhile, 34 proteins were only identified in group C compared to 200 proteins identified in group B. Protein pathway analysis indicated that these proteins were mainly involved in the cellular processes and metabolism of F. gigantica. Among them, 14-3-3θ was consistently identified in group C and may be involved in various cellular processes and innate immune signalling pathways. Members of the HSP family were identified in both groups B and C and may function in both primary and secondary infection processes. The proteins discovered in the present study will help to deepen the understanding of the molecular interactions between F. gigantica and buffalo during secondary infection and facilitate the identification of new potential immunoregulatory molecules.
Collapse
Affiliation(s)
- Mengwei Zheng
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, PR China
| | - Xinping Kong
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, PR China
| | - Xuelian Jiang
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, PR China
| | - Yankun Yang
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, PR China
| | - Shishi Fu
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, PR China
| | - Chongli Wen
- Guangxi Buffalo Research Institute, Chinese Academy Agricultural Sciences, Nanning, 530001, PR China.
| | - Weiyu Zhang
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, PR China.
| | - Wenda Di
- College of Animal Science and Technology, Guangxi University, Nanning, 530005, PR China.
| |
Collapse
|
5
|
Sengupta A, Chakraborty S, Biswas S, Patra SK, Ghosh S. S-nitrosoglutathione (GSNO) induces necroptotic cell death in K562 cells: Involvement of p73, TSC2 and SIRT1. Cell Signal 2024; 124:111377. [PMID: 39222864 DOI: 10.1016/j.cellsig.2024.111377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 08/21/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Nitric oxide and Reactive Nitrogen Species are known to effect tumorigenicity. GSNO is one of the main NO carrying signalling moiety in cell. In the current study, we tried to delve into the effect of GSNO induced nitrosative stress in three different myelogenous leukemic K562, U937 and THP-1 cell lines. METHOD WST-8 assay was performed to investigate cell viability. RT-PCR and western-blot analysis were done to investigate mRNA and protein expression. Spectrophotometric and fluorimetric assays were done to investigate enzyme activities. RESULT We found that GSNO exposure led to reduced cell viability and the mode of cell death in K562 was non apoptotic in nature. GSNO promoted impaired autophagic flux and necroptosis. GSNO treatment heightened phosphorylation of AMPK and TSC2 and inhibited mTOR pathway. We observed increase in NAD+/ NADH ratio following GSNO treatment. Increase in both SIRT1 m-RNA and protein expression was observed. While total SIRT activity remained unaltered. GSNO increased tumor suppressor TAp73/ oncogenic ∆Np73 ratio in K562 cells which was correlated with cell mortality. Surprisingly, GSNO did not alter cellular redox status or redox associated protein expression. However, steep increase in total SNO and PSNO content was observed. Furthermore, inhibition of autophagy, AMPK phosphorylation or SIRT1 exacerbated the effect of GSNO. Altogether our work gives insights into GSNO mediated necroptotic event in K562 cells which can be excavated to develop NO based anticancer therapeutics. CONCLUSION Our data suggests that GSNO could induce necroptotic cell death in K562 through mitochondrial dysfunctionality and PTM of different cellular proteins.
Collapse
Affiliation(s)
- Ayantika Sengupta
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Subhamoy Chakraborty
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Sanchita Biswas
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Sourav Kumar Patra
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Sanjay Ghosh
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| |
Collapse
|
6
|
Cristian PM, Aarón VJ, Armando EHD, Estrella MLY, Daniel NR, David GV, Edgar M, Paul SCJ, Osbaldo RA. Diffusion on PCA-UMAP Manifold: The Impact of Data Structure Preservation to Denoise High-Dimensional Single-Cell RNA Sequencing Data. BIOLOGY 2024; 13:512. [PMID: 39056705 PMCID: PMC11274112 DOI: 10.3390/biology13070512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024]
Abstract
Single-cell transcriptomics (scRNA-seq) is revolutionizing biological research, yet it faces challenges such as inefficient transcript capture and noise. To address these challenges, methods like neighbor averaging or graph diffusion are used. These methods often rely on k-nearest neighbor graphs from low-dimensional manifolds. However, scRNA-seq data suffer from the 'curse of dimensionality', leading to the over-smoothing of data when using imputation methods. To overcome this, sc-PHENIX employs a PCA-UMAP diffusion method, which enhances the preservation of data structures and allows for a refined use of PCA dimensions and diffusion parameters (e.g., k-nearest neighbors, exponentiation of the Markov matrix) to minimize noise introduction. This approach enables a more accurate construction of the exponentiated Markov matrix (cell neighborhood graph), surpassing methods like MAGIC. sc-PHENIX significantly mitigates over-smoothing, as validated through various scRNA-seq datasets, demonstrating improved cell phenotype representation. Applied to a multicellular tumor spheroid dataset, sc-PHENIX identified known extreme phenotype states, showcasing its effectiveness. sc-PHENIX is open-source and available for use and modification.
Collapse
Affiliation(s)
- Padron-Manrique Cristian
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Periferico Sur 4809, Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico; (P.-M.C.); (V.-J.A.); (E.-H.D.A.); (N.-R.D.); (G.-V.D.); (M.E.)
- Programa de Doctorado en Ciencias Biomédicas, Circuito Posgrados, Ciudad Universitaria, Alcaldía Coyoacán Unidad de Posgrado Edificio B primer Piso, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico
| | - Vázquez-Jiménez Aarón
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Periferico Sur 4809, Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico; (P.-M.C.); (V.-J.A.); (E.-H.D.A.); (N.-R.D.); (G.-V.D.); (M.E.)
| | - Esquivel-Hernandez Diego Armando
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Periferico Sur 4809, Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico; (P.-M.C.); (V.-J.A.); (E.-H.D.A.); (N.-R.D.); (G.-V.D.); (M.E.)
| | - Martinez-Lopez Yoscelina Estrella
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Periferico Sur 4809, Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico; (P.-M.C.); (V.-J.A.); (E.-H.D.A.); (N.-R.D.); (G.-V.D.); (M.E.)
- Programa de Doctorado en Ciencias Médicas, Odontológicas y de la Salud, Unidad de Posgrado, Edificio A, 1er Piso, Circuito Posgrados, Ciudad Universitaria, Alcaldía Coyoacán, Universidad Nacional Autónoma de México (UNAM), Mexico City 04510, Mexico
| | - Neri-Rosario Daniel
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Periferico Sur 4809, Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico; (P.-M.C.); (V.-J.A.); (E.-H.D.A.); (N.-R.D.); (G.-V.D.); (M.E.)
- Programa de Maestría en Ciencias Bioquímicas, Unidad de Posgrado, Edificio B, 1er Piso, Circuito de los Posgrados, Ciudad Universitaria, Universidad Nacional Autónoma de México (UNAM), Alcaldía Coyoacán, Ciudad de México 04510, Mexico
| | - Giron-Villalobos David
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Periferico Sur 4809, Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico; (P.-M.C.); (V.-J.A.); (E.-H.D.A.); (N.-R.D.); (G.-V.D.); (M.E.)
- Programa de Maestría en Ciencias Bioquímicas, Unidad de Posgrado, Edificio B, 1er Piso, Circuito de los Posgrados, Ciudad Universitaria, Universidad Nacional Autónoma de México (UNAM), Alcaldía Coyoacán, Ciudad de México 04510, Mexico
| | - Mixcoha Edgar
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Periferico Sur 4809, Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico; (P.-M.C.); (V.-J.A.); (E.-H.D.A.); (N.-R.D.); (G.-V.D.); (M.E.)
- CONAHCYT-INMEGEN, Periferico Sur 4809, Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico
| | - Sánchez-Castañeda Jean Paul
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Periferico Sur 4809, Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico; (P.-M.C.); (V.-J.A.); (E.-H.D.A.); (N.-R.D.); (G.-V.D.); (M.E.)
- Programa de Maestría en Ciencias Bioquímicas, Unidad de Posgrado, Edificio B, 1er Piso, Circuito de los Posgrados, Ciudad Universitaria, Universidad Nacional Autónoma de México (UNAM), Alcaldía Coyoacán, Ciudad de México 04510, Mexico
| | - Resendis-Antonio Osbaldo
- Human Systems Biology Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), Periferico Sur 4809, Arenal Tepepan, Tlalpan, Mexico City 14610, Mexico; (P.-M.C.); (V.-J.A.); (E.-H.D.A.); (N.-R.D.); (G.-V.D.); (M.E.)
- Coordinación de la Investigación Científica-Red de Apoyo a la Investigación, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga, 14, Belisario Dominguez Sección XVI, Tlalpan, Mexico City 14080, Mexico
- Centro de Ciencias de la Complejidad, Unversidad Nacional Autónoma de México (UNAM), Circuito Centro Cultural, Coyoacán, Mexico City 04510, Mexico
| |
Collapse
|
7
|
Noh MR, Padanilam BJ. Cell death induced by acute renal injury: a perspective on the contributions of accidental and programmed cell death. Am J Physiol Renal Physiol 2024; 327:F4-F20. [PMID: 38660714 PMCID: PMC11390133 DOI: 10.1152/ajprenal.00275.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/11/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
The involvement of cell death in acute kidney injury (AKI) is linked to multiple factors including energy depletion, electrolyte imbalance, reactive oxygen species, inflammation, mitochondrial dysfunction, and activation of several cell death pathway components. Since our review in 2003, discussing the relative contributions of apoptosis and necrosis, several other forms of cell death have been identified and are shown to contribute to AKI. Currently, these various forms of cell death can be fundamentally divided into accidental cell death and regulated or programmed cell death based on functional aspects. Several death initiator and effector molecules switch molecules that may act as signaling components triggering either death or protective mechanisms or alternate cell death pathways have been identified as part of the machinery. Intriguingly, several of these cell death pathways share components and signaling pathways suggesting complementary or compensatory functions. Thus, defining the cross talk between distinct cell death pathways and identifying the unique molecular effectors for each type of cell death may be required to develop novel strategies to prevent cell death. Furthermore, depending on the multiple forms of cell death simultaneously induced in different AKI settings, strategies for combination therapies that block multiple cell death pathways need to be developed to completely prevent injury, cell death, and renal function. This review highlights the various cell death pathways, cross talk, and interactions between different cell death modalities in AKI.
Collapse
Affiliation(s)
- Mi Ra Noh
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Babu J Padanilam
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| |
Collapse
|
8
|
Joshi M, Bhatt P. Ameliorated in vitroanti-cancer efficacy of methotrexate loaded zinc oxide nanoparticles in breast cancer cell lines MCF-7 & MDA-MB-231 and its acute toxicity study. NANOTECHNOLOGY 2024; 35:335101. [PMID: 38746972 DOI: 10.1088/1361-6528/ad4b24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024]
Abstract
Traditional therapies often struggle with specificity and resistance in case of cancer treatments. It is therefore important to investigate new approaches for cancer treatment based on nanotechnology. Zinc oxide nanoparticles (ZnONPs) are known to exhibit anti-cancer properties by inducing oxidative stress, apoptosis, and cell cycle arrest. Methotrexate (MTX) a known anti-folate shows specificity to folate receptors and interrupts healthy functioning of cells. This study proposes the use of previously characterized biocompatible Methotrexate loaded Zinc oxide nanoparticles (MTX-ZnONPs) as a dual action therapeutic strategy against breast cancer cell lines, MCF-7 (MTX-sensitive) and MDA-MB-231 (MTX-resistant). To elucidate the cytotoxicity mechanism of MTX-ZnONPs an in depthIn vitrostudy was carried out.In vitroassays, including cell cycle analysis, apoptosis assay, and western blot analysis to study the protein expression were performed. Results of these assays, further supported the anti-cancer activity of MTX-ZnONPs showing apoptotic and necrotic activity in MCF-7 and MDA-MB-231 cell line respectively.In vivoacute oral toxicity study to identify the LD50in animals revealed no signs of toxicity and mortality up to 550 mg kg-1body weight of animal, significantly higher LD50values than anticipated therapeutic levels and safety of the synthesized nanosystem. The study concludes that MTX-ZnONPs exhibit anti-cancer potential against breast cancer cells offering a promising strategy for overcoming resistance.
Collapse
Affiliation(s)
- Mitesh Joshi
- Department of Biological Sciences, SVKM's NMIMS (Deemed-to-be University), Sunandan Divatia School of Science, Vile Parle (West), Mumbai 400056, India
| | - Purvi Bhatt
- Department of Biological Sciences, SVKM's NMIMS (Deemed-to-be University), Sunandan Divatia School of Science, Vile Parle (West), Mumbai 400056, India
| |
Collapse
|
9
|
Tokmakov AA, Teranishi R, Sato KI. Spontaneous Overactivation of Xenopus Frog Eggs Triggers Necrotic Cell Death. Int J Mol Sci 2024; 25:5321. [PMID: 38791359 PMCID: PMC11121189 DOI: 10.3390/ijms25105321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
The excessive activation of frog eggs, referred to as overactivation, can be initiated by strong oxidative stress, leading to expedited calcium-dependent non-apoptotic cell death. Overactivation also occurs spontaneously, albeit at a low frequency, in natural populations of spawned frog eggs. Currently, the cytological and biochemical events of the spontaneous process have not been characterized. In the present study, we demonstrate that the spontaneous overactivation of Xenopus frog eggs, similarly to oxidative stress- and mechanical stress-induced overactivation, is characterized by the fast and irreversible contraction of the egg's cortical layer, an increase in egg size, the depletion of intracellular ATP, a drastic increase in the intracellular ADP/ATP ratio, and the degradation of M phase-specific cyclin B2. These events manifest in eggs in the absence of caspase activation within one hour of triggering overactivation. Importantly, substantial amounts of ATP and ADP leak from the overactivated eggs, indicating that plasma membrane integrity is compromised in these cells. The rupture of the plasma membrane and acute depletion of intracellular ATP explicitly define necrotic cell death. Finally, we report that egg overactivation can occur in the frog's genital tract. Our data suggest that mechanical stress may be a key factor promoting egg overactivation during oviposition in frogs.
Collapse
Affiliation(s)
- Alexander A. Tokmakov
- Institute of Advanced Technology, Faculty of Biology-Oriented Science and Technology, KinDai University, 930 Nishimitani, Kinokawa City 649-6493, Japan
| | - Ryuga Teranishi
- Faculty of Life Sciences, Kyoto Sangyo University, Kamigamo-Motoyama, Kita-ku, Kyoto 603-8555, Japan;
| | - Ken-Ichi Sato
- Faculty of Life Sciences, Kyoto Sangyo University, Kamigamo-Motoyama, Kita-ku, Kyoto 603-8555, Japan;
| |
Collapse
|
10
|
Dong L, Chen M, Huang Z, Tan Y, Zhang C, Zhang S, Zhang Y, Zhang X. A new labdane diterpenoid from Scoparia dulcis improving pancreatic function against islets cell apoptotic by Bax/Bcl-2/Caspase-3 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117571. [PMID: 38103847 DOI: 10.1016/j.jep.2023.117571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Scoparia dulcis has been identified as a significant ethnopharmacological substance in the Li, Zhuang, and Dai ethnic groups of China. Traditional medicine use S. dulcis to treat numerous illnesses, most notably diabetes. The considerable antidiabetic properties of this herbal remedy have been established by several clinical investigations and animal experiments. The islet is the intended target of S. dulcis, although the cause of its activity and mechanism for diabetes treatment is unclear. The diterpenoids from S. dulcis have been shown in the literature to have significant hypoglycemic efficacy and to protect islet cells in vitro. Diterpenoids may be the components of this herbal remedy that preserve islets, but further research is needed. AIM OF THE STUDY This study was projected to investigate the new diterpenoid scoparicol E from S. dulcis and examined its islet-protective effect and the potential mechanism both in vitro and in vivo. METHODS The structure of the novel diterpenoid scoparicol E was clarified by employing a wide range of spectroscopic methods. Using CCK-8 tests, cytotoxicity and antiapoptotic activity of scoparicol E were detected. Serum biochemical analysis and pathologic examination were performed to study the protective effect of scoparicol E against islet damage. The specific mechanism of action of scoparicol E was investigated through the mitochondrial membrane potential, Annexin V-FITC flow cytometry, and western blotting. RESULTS Scoparicol E reduced MLD-STZ-induced hyperglycemia in mice and increased insulin and islet apoptosis. Scoparicol E effectively suppressed the Bax/Bcl-2/Caspase-3 pathway, according to the in vivo western blot investigation. Scoparicol E showed significant antiapoptotic action in vitro. We also showed that scoparicol E might prevent islet cells from dying by inhibiting the Bax/Bcl-2/Caspase-3 pathway. The Annexin V-FITC flow cytometry results revealed that MIN6 cell apoptosis was considerably decreased following scoparicol E intervention, showing anti-islet cell apoptosis action. Furthermore, the Caspase-3-mediated apoptosis pathway depends on cytochrome c and the potential of the mitochondrial membrane. Scoparicol E prevented the release of cytochrome c, restored the mitochondrial membrane potential, and prevented MIN6 cell apoptosis. CONCLUSION We demonstrated the new diterpenoid scoparicol E could protect islet cells apoptosis by modulating the Bax/Bcl-2/Caspase-3 pathway.
Collapse
Affiliation(s)
- Lin Dong
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Mimi Chen
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Zibao Huang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Yinfeng Tan
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China
| | - Caiyun Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China; Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou, 571101, China
| | - Shouwen Zhang
- Research Center for Traditional Chinese Medicine Resources and Ethnic Minority Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Yong Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China.
| | - Xiaopo Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, 571199, China; Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou, 571101, China.
| |
Collapse
|
11
|
Cai M, Wu P, Ni W, Huang D, Wang X. mTORC1 hyperactivation and resultant suppression of macroautophagy contribute to the induction of cardiomyocyte necroptosis by catecholamine surges. Physiol Rep 2024; 12:e15966. [PMID: 38444056 PMCID: PMC10915131 DOI: 10.14814/phy2.15966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/17/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024] Open
Abstract
Previous studies revealed a controversial role of mechanistic target of rapamycin complex 1 (mTORC1) and mTORC1-regulated macroautophagy in isoproterenol (ISO)-induced cardiac injury. Here we investigated the role of mTORC1 and potential underlying mechanisms in ISO-induced cardiomyocyte necrosis. Two consecutive daily injections of ISO (85 mg/kg, s.c.) or vehicle control (CTL) were administered to C57BL/6J mice with or without rapamycin (RAP, 5 mg/kg, i.p.) pretreatment. Western blot analyses showed that myocardial mTORC1 signaling and the RIPK1-RIPK3-MLKL necroptotic pathway were activated, mRNA expression analyses revealed downregulation of representative TFEB target genes, and Evan's blue dye uptake assays detected increased cardiomyocyte necrosis in ISO-treated mice. However, RAP pretreatment prevented or significantly attenuated the ISO-induced cardiomyocyte necrosis, myocardial inflammation, downregulation of TFEB target genes, and activation of the RIPK1-RIPK3-MLKL pathway. LC3-II flux assays confirmed the impairment of myocardial autophagic flux in the ISO-treated mice. In cultured neonatal rat cardiomyocytes, mTORC1 signaling was also activated by ISO, and inhibition of mTORC1 by RAP attenuated ISO-induced cytotoxicity. These findings suggest that mTORC1 hyperactivation and resultant suppression of macroautophagy play a major role in the induction of cardiomyocyte necroptosis by catecholamine surges, identifying mTORC1 inhibition as a potential strategy to treat heart diseases with catecholamine surges.
Collapse
Affiliation(s)
- Mingqi Cai
- Heart CenterShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Division of Basic Biomedical SciencesSanford School of Medicine of the University of South DakotaVermillionSouth DakotaUSA
| | - Penglong Wu
- Division of Basic Biomedical SciencesSanford School of Medicine of the University of South DakotaVermillionSouth DakotaUSA
- Department of Cardiology, Xiamen Cardiovascular Hospital of Xiamen University, School of MedicineXiamen UniversityXiamenChina
| | - Wei Ni
- Department of Cardiology, Xiamen Cardiovascular Hospital of Xiamen University, School of MedicineXiamen UniversityXiamenChina
| | - Dong Huang
- Heart CenterShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xuejun Wang
- Division of Basic Biomedical SciencesSanford School of Medicine of the University of South DakotaVermillionSouth DakotaUSA
| |
Collapse
|
12
|
Ganguly U, Singh S, Bir A, Ghosh A, Chakrabarti SS, Saini RV, Saso L, Bisaglia M, Chakrabarti S. Alpha-synuclein interaction with mitochondria is the final mechanism of ferroptotic death induced by erastin in SH-SY5Y cells. Free Radic Res 2024; 58:217-228. [PMID: 38572725 DOI: 10.1080/10715762.2024.2336563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 02/16/2024] [Indexed: 04/05/2024]
Abstract
Ferroptosis has been characterized as a form of iron-dependent regulated cell death accompanied by an accumulation of reactive oxygen species and lipid oxidation products along with typical morphological alterations in mitochondria. Ferroptosis is activated by diverse triggers and inhibited by ferrostatin-1 and liproxstatin-1, apart from iron chelators and several antioxidants, and the process is implicated in multiple pathological conditions. There are, however, certain ambiguities about ferroptosis, especially regarding the final executioner of cell death subsequent to the accumulation of ROS. This study uses a typical inducer of ferroptosis such as erastin on SH-SY5Y cells, and shows clearly that ferroptotic death of cells is accompanied by the loss of mitochondrial membrane potential and intracellular ATP content along with an accumulation of oxidative stress markers. All these are prevented by ferrostatin-1 and liproxstatin-1. Additionally, cyclosporine A prevents mitochondrial alterations and cell death induced by erastin implying the crucial role of mitochondrial permeability transition pore (mPTP) activation in ferroptotic death. Furthermore, an accumulation of α-synuclein occurs during erastin induced ferroptosis which can be inhibited by ferrostatin-1 and liproxstatin-1. When the knock-down of α-synuclein expression is performed by specific siRNA treatment of SH-SY5Y cells, the mitochondrial impairment and ferroptotic death of the cells induced by erastin are markedly prevented. Thus, α-synuclein through the involvement of mPTP appears to be the key executioner protein of ferroptosis induced by erastin, but it needs to be verified if it is a generalized mechanism of ferroptosis by using other inducers and cell lines.
Collapse
Affiliation(s)
- Upasana Ganguly
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Ambala, India
| | - Sukhpal Singh
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Ambala, India
| | - Aritri Bir
- Department of Biochemistry, Dr B. C. Roy Multi-Speciality Medical Research Centre, IIT Kharagpur, India
| | - Arindam Ghosh
- Department of Biochemistry, Dr B. C. Roy Multi-Speciality Medical Research Centre, IIT Kharagpur, India
| | - Sankha Shubhra Chakrabarti
- Department of Geriatric Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Reena V Saini
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar University (Deemed to be), Ambala, India
| | - Luciano Saso
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Marco Bisaglia
- Department of Biology, University of Padova, Padova, Italy
| | - Sasanka Chakrabarti
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Ambala, India
| |
Collapse
|
13
|
Ahmad S, Singh V, Gautam HK, Raza K. Multisampling-based docking reveals Imidazolidinyl urea as a multitargeted inhibitor for lung cancer: an optimisation followed multi-simulation and in-vitro study. J Biomol Struct Dyn 2024; 42:2494-2511. [PMID: 37154501 DOI: 10.1080/07391102.2023.2209673] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/16/2023] [Indexed: 05/10/2023]
Abstract
Lung Cancer is one of the deadliest cancers, responsible for more than 1.80 million deaths annually worldwide, and it is on the priority list of WHO. In the current scenario, when cancer cells become resistant to the drug, making it less effective leaves the patient in vulnerable conditions. To overcome this situation, researchers are constantly working on new drugs and medications that can help fight drug resistance and improve patients' outcomes. In this study, we have taken five main proteins of lung cancer, namely RSK4 N-terminal kinase, guanylate kinase, cyclin-dependent kinase 2, kinase CK2 holoenzyme, tumour necrosis factor-alpha and screened the prepared Drug Bank library with 1,55,888 compounds against all using three Glide-based docking algorithms namely HTVS, standard precision and extra precise with a docking score ranging from -5.422 to -8.432 Kcal/mol. The poses were filtered with the MM\GBSA calculations, which helped to identify Imidazolidinyl urea C11H16N8O8 (DB14075) as a multitargeted inhibitor for lung cancer, validated with advanced computations like ADMET, interaction pattern fingerprints, and optimised the compound with Jaguar, producing satisfied relative energy. All five complexes were performed with MD Simulation for 100 ns with NPT ensemble class, producing cumulative deviation and fluctuations < 2 Å and a web of intermolecular interaction, making the complexes stable. Further, the in-vitro analysis for morphological imaging, Annexin V/PI FACS assay, ROS and MMP analysis caspase3//7 activity were performed on the A549 cell line producing promising results and can be an option to treat lung cancer at a significantly cheaper state.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shaban Ahmad
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Vijay Singh
- Immunology and Infectious Disease, Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Hemant K Gautam
- Immunology and Infectious Disease, Institute of Genomics and Integrative Biology (IGIB), New Delhi, India
| | - Khalid Raza
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
14
|
Kasica N, Kaleczyc J. Xanthohumol, a prenylated flavonoid from hops (Humulus lupulus L.) exerts multidirectional pro-healing properties towards damaged zebrafish hair cells by regulating the innate immune response. Toxicol Appl Pharmacol 2024; 483:116809. [PMID: 38211931 DOI: 10.1016/j.taap.2024.116809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/18/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
Xanthohumol (XN) is a prominent prenylated flavonoid present in the hop plant (Humulus lupulus L.). Despite undoubted pro-healing properties of hop plant, there is still a need for clinical investigations confirming these effects as well as the underlying molecular mechanisms. The present study was designed to (1) establish the role of XN in non-invasive inflammation induced by chemical damage to zebrafish hair cells, (2) clarify if it influences cell injury severity, neutrophil migration, macrophage activation, cell regeneration, and (3) find out whether it modulates the gene expression profile of chosen immune and stress response markers. All experiments were performed on 3 dpf zebrafish larvae. After fertilization the embryos were transferred to appropriate XN solutions (0.1 μM, 0.3 μM and 0.5 μM). The 40 min 10 μM CuSO4 exposure evoked severe damage to posterior lateral line hair cells triggering a robust acute inflammatory response. Four readouts were selected as the indicators of XN role in the process of inflammation: 1) hair cell death, 2) neutrophil migration towards damaged hair cells, 3) macrophage activation and recruitment to damaged hair cells, 4) hair cell regeneration. The assessments involved in vivo confocal microscopy imaging and qPCR based molecular analysis. It was demonstrated that XN (1) influences death pathway of damaged hair cells by redirecting their severe necrotic phenotype into apoptotic one, (2) impacts the immune response via regulating neutrophil migration, macrophage recruitment and activation (3) modulates gene expression of immune system markers and (4) accelerates hair cell regeneration.
Collapse
Affiliation(s)
- Natalia Kasica
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland.
| | - Jerzy Kaleczyc
- Department of Animal Anatomy, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| |
Collapse
|
15
|
Bessone F, Hillotte GL, Ahumada N, Jaureguizahar F, Medeot AC, Roma MG. UDCA for Drug-Induced Liver Disease: Clinical and Pathophysiological Basis. Semin Liver Dis 2024; 44:1-22. [PMID: 38378025 DOI: 10.1055/s-0044-1779520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Drug-induced liver injury (DILI) is an adverse reaction to medications and other xenobiotics that leads to liver dysfunction. Based on differential clinical patterns of injury, DILI is classified into hepatocellular, cholestatic, and mixed types; although hepatocellular DILI is associated with inflammation, necrosis, and apoptosis, cholestatic DILI is associated with bile plugs and bile duct paucity. Ursodeoxycholic acid (UDCA) has been empirically used as a supportive drug mainly in cholestatic DILI, but both curative and prophylactic beneficial effects have been observed for hepatocellular DILI as well, according to preliminary clinical studies. This could reflect the fact that UDCA has a plethora of beneficial effects potentially useful to treat the wide range of injuries with different etiologies and pathomechanisms occurring in both types of DILI, including anticholestatic, antioxidant, anti-inflammatory, antiapoptotic, antinecrotic, mitoprotective, endoplasmic reticulum stress alleviating, and immunomodulatory properties. In this review, a revision of the literature has been performed to evaluate the efficacy of UDCA across the whole DILI spectrum, and these findings were associated with the multiple mechanisms of UDCA hepatoprotection. This should help better rationalize and systematize the use of this versatile and safe hepatoprotector in each type of DILI scenarios.
Collapse
Affiliation(s)
- Fernando Bessone
- Hospital Provincial del Centenario, Facultad de Ciencias Médicas, Servicio de Gastroenterología y Hepatología, Universidad Nacional de Rosario, Rosario, Argentina
| | - Geraldine L Hillotte
- Instituto de Fisiología Experimental (IFISE-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Natalia Ahumada
- Hospital Provincial del Centenario, Facultad de Ciencias Médicas, Servicio de Gastroenterología y Hepatología, Universidad Nacional de Rosario, Rosario, Argentina
| | - Fernanda Jaureguizahar
- Hospital Provincial del Centenario, Facultad de Ciencias Médicas, Servicio de Gastroenterología y Hepatología, Universidad Nacional de Rosario, Rosario, Argentina
| | | | - Marcelo G Roma
- Instituto de Fisiología Experimental (IFISE-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
| |
Collapse
|
16
|
Verma P, Chauhan A, Thakur R, Lata K, Sharma A, Chattopadhyay K, Mukhopadhaya A. Vibrio parahaemolyticus thermostable direct haemolysin induces non-classical programmed cell death despite caspase activation. Mol Microbiol 2023; 120:845-873. [PMID: 37818865 DOI: 10.1111/mmi.15180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 09/02/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023]
Abstract
Thermostable direct haemolysin (TDH) is the key virulence factor secreted by the human gastroenteric bacterial pathogen Vibrio parahaemolyticus. TDH is a membrane-damaging pore-forming toxin. It evokes potent cytotoxicity, the mechanism of which still remains under-explored. Here, we have elucidated the mechanistic details of cell death response elicited by TDH. Employing Caco-2 intestinal epithelial cells and THP-1 monocytic cells, we show that TDH induces some of the hallmark features of apoptosis-like programmed cell death. TDH triggers caspase-3 and 7 activations in the THP-1 cells, while caspase-7 activation is observed in the Caco-2 cells. Interestingly, TDH appears to induce caspase-independent cell death. Higher XIAP level and lower Smac/Diablo level upon TDH intoxication provide plausible explanation for the functional inability of caspases in the THP-1 cells, in particular. Further exploration reveals that mitochondria play a central role in the TDH-induced cell death. TDH triggers mitochondrial damage, resulting in the release of AIF and endonuclease G, responsible for the execution of caspase-independent cell death. Among the other critical mediators of cell death, ROS is found to play an important role in the THP-1 cells, while PARP-1 appears to play a critical role in the Caco-2 cells. Altogether, our work provides critical new insights into the mechanism of cell death induction by TDH, showing a common central theme of non-classical programmed cell death. Our study also unravels the interplay of crucial molecules in the underlying signalling processes. Our findings add valuable insights into the role of TDH in the context of the host-pathogen interaction processes.
Collapse
Affiliation(s)
- Pratima Verma
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Aakanksha Chauhan
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Reena Thakur
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Kusum Lata
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Arpita Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Kausik Chattopadhyay
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| | - Arunika Mukhopadhaya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, Punjab, India
| |
Collapse
|
17
|
Tajvar Nasab N, Jalili-Nik M, Afshari AR, Rezaei Farimani A, Soukhtanloo M. Urolithin B inhibits proliferation and migration and promotes apoptosis and necrosis by inducing G2/M arrest and targeting MMP-2/-9 expression in osteosarcoma cells. J Biochem Mol Toxicol 2023; 37:e23486. [PMID: 37555500 DOI: 10.1002/jbt.23486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/14/2023] [Accepted: 07/28/2023] [Indexed: 08/10/2023]
Abstract
Osteosarcoma (OS) is the most prevalent primary bone cancer, with a high morbidity and mortality rate. Over the past decades, therapeutic approaches have not considerably improved patients' survival rates, and further research is required to find efficient treatments for OS. Data from several studies have shown that urolithin B (UB), the intestinal metabolite of polyphenolic ellagitannins, is emerging as a new class of anticancer compounds, yet its effect on OS cancer cells remains elusive. Herein, we investigated UB's antimetastatic, antiproliferative, and apoptotic effects on the MG-63 OS cell line. Cell viability assay, annexin V/propidium iodide staining, cell cycle arrest analysis, determination of the gene expression of MMP-2, MMP-9, Bax, Bcl-2, and p53 messenger RNA (mRNA), evaluation of reactive oxygen species (ROS) generation and migration, and MMP-2 and MMP-9 protein expression assessments were performed. UB caused late apoptosis, necrosis, G2/M arrest, and ROS generation in MG-63 cells. It increased the mRNA expression of the p53 tumor suppressor and Bax proapoptotic genes. UB also inhibited the migration and metastatic behavior of MG-63 OS cells by downregulating mRNA and MMP-2 and MMP-9 protein expression. In general, although further in vivo investigations are warranted, the current results showed that UB might be utilized as a potential novel natural compound for OS therapy due to its nontoxic, antiproliferative, and antimetastatic nature.
Collapse
Affiliation(s)
- Nahid Tajvar Nasab
- Department of Clinical Biochemistry, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Jalili-Nik
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Azam Rezaei Farimani
- Department of Clinical Biochemistry, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmacological Research Center of Medicinal Plants, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Kheyar A, Ahnou N, Ahmed-Belkacem A, Hulin A, Pressiat C, Ghaleh B, Guichou JF, Morin D, Pawlotsky JM, Teixeira-Clerc F. The novel cyclophilin inhibitor C105SR reduces hepatic ischaemia-reperfusion injury via mitoprotection. JHEP Rep 2023; 5:100876. [PMID: 37860051 PMCID: PMC10582583 DOI: 10.1016/j.jhepr.2023.100876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/07/2023] [Accepted: 07/27/2023] [Indexed: 10/21/2023] Open
Abstract
Background & Aims Mitochondrial permeability transition pore (mPTP) opening is critical for mediating cell death during hepatic ischaemia-reperfusion injury (IRI). Blocking mPTP opening by inhibiting cyclophilin D (CypD) is a promising pharmacological approach for the treatment of IRI. Here, we show that diastereoisomers of a new class of small-molecule cyclophilin inhibitors (SMCypIs) have properties that make them attractive candidates for the development of therapeutic agents against liver IRI. Methods Derivatives of the parent SMCypI were synthesised and evaluated for their ability to inhibit CypD peptidyl-prolyl cis-trans isomerase (PPIase) activity and for their mitoprotective properties, evaluated by measuring mitochondrial swelling and calcium retention capacity in liver mitochondria. The ability of the selected compounds to inhibit mPTP opening was evaluated in cells subjected to hypoxia/reoxygenation using a calcein/cobalt assay. Their ability to inhibit cell death was evaluated in cells subjected to hypoxia/reoxygenation by measuring lactate dehydrogenase (LDH) release, propidium iodide staining, and cell viability. The compound performing best in vitro was selected for in vivo efficacy evaluation in a mouse model of hepatic IRI. Results The two compounds that showed the strongest inhibition of CypD PPIase activity and mPTP opening, C105 and C110, were selected. Their SR diastereoisomers carried the activity of the racemic mixture and exhibited mitoprotective properties superior to those of the known macrocyclic cyclophilin inhibitors cyclosporin A and alisporivir. C105SR was more potent than C110SR in inhibiting mPTP opening and prevented cell death in a model of hypoxia/reoxygenation. Finally, C105SR substantially protected against hepatic IRI in vivo by reducing hepatocyte necrosis and apoptosis. Conclusions We identified a novel cyclophilin inhibitor with strong mitoprotective properties both in vitro and in vivo that represents a promising candidate for cellular protection in hepatic IRI. Impact and Implications Hepatic ischaemia-reperfusion injury (IRI) is one of the main causes of morbidity and mortality during or after liver surgery. However, no effective therapies are available to prevent or treat this devastating syndrome. An attractive strategy to prevent hepatic IRI aims at reducing cell death by targeting mitochondrial permeability transition pore opening, a phenomenon regulated by cyclophilin D. Here, we identified a new small-molecule cyclophilin inhibitor, and demonstrated the enhanced mitoprotective and hepatoprotective properties of one of its diastereoisomers both in vitro and in vivo, making it an attractive lead compound for subsequent clinical development.
Collapse
Affiliation(s)
- Amel Kheyar
- Équipe “Virus, Hépatologie, Cancer”, INSERM U955, IMRB, Université Paris-Est, Créteil, France
| | - Nazim Ahnou
- Équipe “Virus, Hépatologie, Cancer”, INSERM U955, IMRB, Université Paris-Est, Créteil, France
| | | | - Anne Hulin
- Laboratoire de Pharmacologie, DMU de Biologie et Pathologie, Hôpitaux Universitaires Henri Mondor, AP-HP, Créteil, France
- Équipe “Pharmacologie et Technologies pour les Maladies Cardiovasculaires”, INSERM U955, IMRB, Université Paris-Est, Créteil, France
| | - Claire Pressiat
- Laboratoire de Pharmacologie, DMU de Biologie et Pathologie, Hôpitaux Universitaires Henri Mondor, AP-HP, Créteil, France
- Équipe “Pharmacologie et Technologies pour les Maladies Cardiovasculaires”, INSERM U955, IMRB, Université Paris-Est, Créteil, France
| | - Bijan Ghaleh
- Équipe “Pharmacologie et Technologies pour les Maladies Cardiovasculaires”, INSERM U955, IMRB, Université Paris-Est, Créteil, France
| | - Jean-François Guichou
- Centre de Biologie Structurale (CBS), Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Didier Morin
- Équipe “Pharmacologie et Technologies pour les Maladies Cardiovasculaires”, INSERM U955, IMRB, Université Paris-Est, Créteil, France
| | - Jean-Michel Pawlotsky
- Équipe “Virus, Hépatologie, Cancer”, INSERM U955, IMRB, Université Paris-Est, Créteil, France
- Département Prévention, Diagnostic et Traitement des Infections, DMU de Biologie et Pathologie, Hôpitaux Universitaires Henri Mondor, AP-HP, Créteil, France
| | - Fatima Teixeira-Clerc
- Équipe “Virus, Hépatologie, Cancer”, INSERM U955, IMRB, Université Paris-Est, Créteil, France
| |
Collapse
|
19
|
Zhang M, Chen Y, Qiu Y, Sun J, He J, Liu Z, Shi J, Wei W, Wu G, Liang J. PCSK9 Promotes Hypoxia-Induced EC Pyroptosis by Regulating Smac Mitochondrion-Cytoplasm Translocation in Critical Limb Ischemia. JACC Basic Transl Sci 2023; 8:1060-1077. [PMID: 37791316 PMCID: PMC10544082 DOI: 10.1016/j.jacbts.2023.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 10/05/2023]
Abstract
Hypoxia-induced endothelial cell death and impaired angiogenesis are the main pathophysiological features of critical limb ischemia. Mechanistically, proprotein convertase subtilisin/kexin type 9 (PCSK9) promoted Smac translocation from mitochondria to the cytoplasm. Inhibition of Smac release into the cytoplasm attenuated PCSK9-mediated hypoxia-induced pyroptosis. Functionally, PCSK9 overexpression impaired angiogenesis in vitro and reduced blood perfusion in mice with lower limb ischemia, but the effect was reversed by PCSK9 inhibition. This study demonstrates that PCSK9 aggravates pyroptosis by regulating Smac mitochondrion-cytoplasm translocation in the vascular endothelium, providing novel insights into PCSK9 as a potential therapeutic target in critical limb ischemia.
Collapse
Affiliation(s)
- Meixin Zhang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yixi Chen
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yumin Qiu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiapan Sun
- Department of Geriatrics, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Jiang He
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhefu Liu
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian Shi
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Wenbin Wei
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Guifu Wu
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Jianwen Liang
- Department of Cardiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
20
|
Armstrong-Jr R, Ricardo-da-Silva FY, Vidal-Dos-Santos M, da Anunciação LF, Ottens PJ, Correia CJ, Moreira LFP, Leuvenink HGD, Breithaupt-Faloppa AC. Comparison of acute kidney injury following brain death between male and female rats. Clinics (Sao Paulo) 2023; 78:100222. [PMID: 37257364 DOI: 10.1016/j.clinsp.2023.100222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/27/2023] [Accepted: 05/16/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND Clinical reports associate kidneys from female donors with worse prognostic in male recipients. Brain Death (BD) produces immunological and hemodynamic disorders that affect organ viability. Following BD, female rats are associated with increased renal inflammation interrelated with female sex hormone reduction. Here, the aim was to investigate the effects of sex on BD-induced Acute Kidney Injury (AKI) using an Isolated Perfused rat Kidney (IPK) model. METHODS Wistar rats, females, and males (8 weeks old), were maintained for 4h after BD. A left nephrectomy was performed and the kidney was preserved in a cold saline solution (30 min). IPK was performed under normothermic temperature (37°C) for 90 min using WME as perfusion solution. AKI was assessed by morphological analyses, staining of complement system components and inflammatory cell markers, perfusion flow, and creatinine clearance. RESULTS BD-male kidneys had decreased perfusion flow on IPK, a phenomenon that was not observed in the kidneys of BD-females (p < 0.0001). BD-male kidneys presented greater proximal (p = 0.0311) and distal tubule (p = 0.0029) necrosis. However, BD-female kidneys presented higher expression of eNOS (p = 0.0060) and greater upregulation of inflammatory mediators, iNOS (p = 0.0051), and Caspase-3 (p = 0.0099). In addition, both sexes had increased complement system formation (C5b-9) (p=0.0005), glomerular edema (p = 0.0003), and nNOS (p = 0.0051). CONCLUSION The present data revealed an important sex difference in renal perfusion in the IPK model, evidenced by a pronounced reduction in perfusate flow and low eNOS expression in the BD-male group. Nonetheless, the upregulation of genes related to the proinflammatory cascade suggests a progressive inflammatory process in BD-female kidneys.
Collapse
Affiliation(s)
- Roberto Armstrong-Jr
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Fernanda Yamamoto Ricardo-da-Silva
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brasil
| | - Marina Vidal-Dos-Santos
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands; Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brasil
| | - Lucas Ferreira da Anunciação
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brasil
| | - Petra J Ottens
- Department of Surgery, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Cristiano Jesus Correia
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brasil
| | - Luiz Felipe Pinho Moreira
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brasil
| | | | - Ana Cristina Breithaupt-Faloppa
- Laboratório de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brasil.
| |
Collapse
|
21
|
Deragon MA, McCaig WD, Truong PV, Metz KR, Carron KA, Hughes KJ, Knapp AR, Dougherty MJ, LaRocca TJ. Mitochondrial Trafficking of MLKL, Bak/Bax, and Drp1 Is Mediated by RIP1 and ROS which Leads to Decreased Mitochondrial Membrane Integrity during the Hyperglycemic Shift to Necroptosis. Int J Mol Sci 2023; 24:ijms24108609. [PMID: 37239951 DOI: 10.3390/ijms24108609] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Apoptosis and necroptosis overlap in their initial signaling but diverge to produce non-inflammatory and pro-inflammatory outcomes, respectively. High glucose pushes signaling in favor of necroptosis producing a hyperglycemic shift from apoptosis to necroptosis. This shift depends on receptor-interacting protein 1 (RIP1) and mitochondrial reactive oxygen species (ROS). Here, we show that RIP1, mixed lineage kinase domain-like (MLKL) protein, Bcl-2 agonist/killer (Bak), Bcl-2 associated x (Bax) protein, and dynamin-related protein 1 (Drp1) traffic to the mitochondria in high glucose. RIP1 and MLKL appear in the mitochondria in their activated, phosphorylated states while Drp1 appears in its activated, dephosphorylated state in high glucose. Mitochondrial trafficking is prevented in rip1 KO cells and upon treatment with N-acetylcysteine. Induction of ROS replicated the mitochondrial trafficking seen in high glucose. MLKL forms high MW oligomers in the outer and inner mitochondrial membranes while Bak and Bax form high MW oligomers in the outer mitochondrial membrane in high glucose, suggesting pore formation. MLKL, Bax, and Drp1 promoted cytochrome c release from the mitochondria as well as a decrease in mitochondrial membrane potential in high glucose. These results indicate that mitochondrial trafficking of RIP1, MLKL, Bak, Bax, and Drp1 are key events in the hyperglycemic shift from apoptosis to necroptosis. This is also the first report to show oligomerization of MLKL in the inner and outer mitochondrial membranes and dependence of mitochondrial permeability on MLKL.
Collapse
Affiliation(s)
- Matthew A Deragon
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| | - William D McCaig
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| | - Phillip V Truong
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| | - Kevin R Metz
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| | - Katherine A Carron
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| | - Keven J Hughes
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| | - Angeleigh R Knapp
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| | - Molly J Dougherty
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| | - Timothy J LaRocca
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA
| |
Collapse
|
22
|
Zhou Q, Quirk JD, Hu Y, Yan H, Gaut JP, Pham CTN, Wickline SA, Pan H. Rapamycin Perfluorocarbon Nanoparticle Mitigates Cisplatin-Induced Acute Kidney Injury. Int J Mol Sci 2023; 24:6086. [PMID: 37047059 PMCID: PMC10093942 DOI: 10.3390/ijms24076086] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
For nearly five decades, cisplatin has played an important role as a standard chemotherapeutic agent and been prescribed to 10-20% of all cancer patients. Although nephrotoxicity associated with platinum-based agents is well recognized, treatment of cisplatin-induced acute kidney injury is mainly supportive and no specific mechanism-based prophylactic approach is available to date. Here, we postulated that systemically delivered rapamycin perfluorocarbon nanoparticles (PFC NP) could reach the injured kidneys at sufficient and sustained concentrations to mitigate cisplatin-induced acute kidney injury and preserve renal function. Using fluorescence microscopic imaging and fluorine magnetic resonance imaging/spectroscopy, we illustrated that rapamycin-loaded PFC NP permeated and were retained in injured kidneys. Histologic evaluation and blood urea nitrogen (BUN) confirmed that renal structure and function were preserved 48 h after cisplatin injury. Similarly, weight loss was slowed down. Using western blotting and immunofluorescence staining, mechanistic studies revealed that rapamycin PFC NP significantly enhanced autophagy in the kidney, reduced the expression of intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1), as well as decreased the expression of the apoptotic protein Bax, all of which contributed to the suppression of apoptosis that was confirmed with TUNEL staining. In summary, the delivery of an approved agent such as rapamycin in a PFC NP format enhances local delivery and offers a novel mechanism-based prophylactic therapy for cisplatin-induced acute kidney injury.
Collapse
Affiliation(s)
- Qingyu Zhou
- Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA
| | - James D. Quirk
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ying Hu
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Huimin Yan
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph P. Gaut
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christine T. N. Pham
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel A. Wickline
- Morsani College of Medicine, University of South Florida, Tampa, FL 33620, USA
| | - Hua Pan
- Division of Rheumatology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| |
Collapse
|
23
|
Gulka SMD, Gowen B, Litke AM, Delaney KR, Chow RL. Laser-induced microinjury of the corneal basal epithelium and imaging of resident macrophage responses in a live, whole-eye preparation. Front Immunol 2023; 14:1050594. [PMID: 36814930 PMCID: PMC9939765 DOI: 10.3389/fimmu.2023.1050594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 01/23/2023] [Indexed: 02/09/2023] Open
Abstract
The corneal epithelium is continuously subjected to external stimuli that results in varying degrees of cellular damage. The use of live-cell imaging approaches has facilitated understanding of the cellular and molecular mechanisms underlying the corneal epithelial wound healing process. Here, we describe a live, ex vivo, whole-eye approach using laser scanning confocal microscopy to simultaneously induce and visualize short-term cellular responses following microdamage to the corneal epithelium. Live-cell imaging of corneal cell layers was enabled using the lipophilic fluorescent dyes, SGC5 or FM4-64, which, when injected into the anterior chamber of enucleated eyes, readily penetrated and labelled cell membranes. Necrotic microdamage to a defined region (30 μm x 30 μm) through the central plane of the corneal basal epithelium was induced by continuously scanning for at least one minute using high laser power and was dependent on the presence of lipophilic fluorescent dye. This whole-mount live-cell imaging and microdamage approach was used to examine the behavior of Cx3cr1:GFP-expressing resident corneal stromal macrophages (RCSMs). In undamaged corneas, RCSMs remained stationary, but exhibited a constant extension and retraction of short (~5 μm) semicircular, pseudopodia-like processes reminiscent of what has previously been reported in corneal dendritic cells. Within minutes of microdamage, nearby anterior RCSMs became highly polarized and extended projections towards the damaged region. The extension of the processes plateaued after about 30 minutes and remained stable over the course of 2-3 hours of imaging. Retrospective immunolabeling showed that these responding RCSMs were MHC class II+. This study adds to existing knowledge of immune cell behavior in response to corneal damage and introduces a simple corneal epithelial microdamage and wound healing paradigm.
Collapse
Affiliation(s)
- Sebastian M. D. Gulka
- Department of Biology, University of Victoria, Victoria, BC, Canada
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Brent Gowen
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | | | - Kerry R. Delaney
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | - Robert L. Chow
- Department of Biology, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
24
|
Zhao L, Fan J, Zhang C, Zhang Z, Dong J. CircRANBP17 modulated KDM1A to regulate neuroblastoma progression by sponging miR-27b-3p. Open Med (Wars) 2023; 18:20230672. [PMID: 36941992 PMCID: PMC10024347 DOI: 10.1515/med-2023-0672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 03/14/2023] Open
Abstract
Neuroblastoma (NB) is a common childhood cancer. Circular RNA RAN binding protein 17 (circRANBP17) has been identified to participate in diverse tumor progression. This study aims to explore the function and mechanism of circRANBP17 in NB. The levels of circRANBP17, miR-27b-3p and KDM1A in NB tissues and cells were measured by qRT-PCR. Mouse model assay was performed to investigate the effect of circRANBP17 knockdown on tumor formation in vivo. The levels of circRANBP17 and KDM1A were significantly up-regulated, and the level of miR-27b-3p was strikingly down-regulated in NB tissues and cells (SK-N-SH and SK-N-AS). Functional studies indicated that miR-27b-3p inhibitor mitigated the inhibitory effects on cell proliferation, migration, invasion and the promoting effect on cell apoptosis in SK-N-SH and SK-N-AS cells induced by circRANBP17 knockdown. Also, miR-27b-3p regulated NB cell malignancy by targeting KDM1A. Further studies revealed that miR-27b-3p inhibitor reversed the low expression of KDM1A induced by circRANBP17 knockdown. In support, circRANBP17 knockdown led to inhibition of tumor formation in vivo. In conclusion, circRANBP17 modulated KDM1A to promote cell proliferation, migration, invasion and restrain cell apoptosis in NB by sponging miR-27b-3p, and the new regulatory network may provide a theoretical basis for the further study of NB.
Collapse
Affiliation(s)
- Lijun Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, 215000, China
| | - Junying Fan
- Department of Nephrology, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Chunyang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Zhenjun Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Jun Dong
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, No. 1055, Sanxiang
Road, Gusu District, Suzhou City, Jiangsu Province, 215000, China
| |
Collapse
|
25
|
Robichaux DJ, Harata M, Murphy E, Karch J. Mitochondrial permeability transition pore-dependent necrosis. J Mol Cell Cardiol 2023; 174:47-55. [PMID: 36410526 PMCID: PMC9868081 DOI: 10.1016/j.yjmcc.2022.11.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/17/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022]
Abstract
Mitochondrial permeability transition pore (mPTP)-dependent cell death is a form of necrotic cell death that is driven by mitochondrial dysfunction by the opening of the mPTP and is triggered by increases in matrix levels of Ca2+ and reactive oxygen species. This form of cell death has been implicated in ischemic injuries of the heart and brain as well as numerous degenerative diseases in the brain and skeletal muscle. This review focuses on the molecular triggers and regulators of mPTP-dependent necrosis in the context of myocardial ischemia reperfusion injury. Research over the past 50 years has led to the identity of regulators and putative pore-forming components of the mPTP. Finally, downstream consequences of activation of the mPTP as well as ongoing questions and areas of research are discussed. These questions pose a particular interest as targeting the mPTP could potentially represent an efficacious therapeutic strategy to reduce infarct size following an ischemic event.
Collapse
Affiliation(s)
- Dexter J Robichaux
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Mikako Harata
- Cardiovascular Branch, NHLBI, NIH, 10 Center Drive, Bethesda, MD, USA
| | - Elizabeth Murphy
- Cardiovascular Branch, NHLBI, NIH, 10 Center Drive, Bethesda, MD, USA
| | - Jason Karch
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
26
|
Zhang Y, Chen S, Fan F, Xu N, Meng XL, Zhang Y, Lin JM. Neurotoxicity mechanism of aconitine in HT22 cells studied by microfluidic chip-mass spectrometry. J Pharm Anal 2023; 13:88-98. [PMID: 36820076 PMCID: PMC9937797 DOI: 10.1016/j.jpha.2022.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Aconitine, a common and main toxic component of Aconitum, is toxic to the central nervous system. However, the mechanism of aconitine neurotoxicity is not yet clear. In this work, we had the hypothesis that excitatory amino acids can trigger excitotoxicity as a pointcut to explore the mechanism of neurotoxicity induced by aconitine. HT22 cells were simulated by aconitine and the changes of target cell metabolites were real-time online investigated based on a microfluidic chip-mass spectrometry system. Meanwhile, to confirm the metabolic mechanism of aconitine toxicity on HT22 cells, the levels of lactate dehydrogenase, intracellular Ca2+, reactive oxygen species, glutathione and superoxide dismutase, and ratio of Bax/Bcl-2 protein were detected by molecular biotechnology. Integration of the detected results revealed that neurotoxicity induced by aconitine was associated with the process of excitotoxicity caused by glutamic acid and aspartic acid, which was followed by the accumulation of lactic acid and reduction of glucose. The surge of extracellular glutamic acid could further lead to a series of cascade reactions including intracellular Ca2+ overload and oxidative stress, and eventually result in cell apoptosis. In general, we illustrated a new mechanism of aconitine neurotoxicity and presented a novel analysis strategy that real-time online monitoring of cell metabolites can provide a new approach to mechanism analysis.
Collapse
Affiliation(s)
- Yingrui Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China,Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Shiyu Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China,Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Fangfang Fan
- Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ning Xu
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Xian-Li Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yi Zhang
- Ethnic Medicine Academic Heritage Innovation Research Center, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China,Corresponding author.
| | - Jin-Ming Lin
- Beijing Key Laboratory of Microanalytical Methods and Instrumentation, Department of Chemistry, Tsinghua University, Beijing, 100084, China,Corresponding author.
| |
Collapse
|
27
|
M M, Gadre S, Chhatar S, Chakraborty G, Ahmed N, Patra C, Patra M. Potent Ruthenium-Ferrocene Bimetallic Antitumor Antiangiogenic Agent That Circumvents Platinum Resistance: From Synthesis and Mechanistic Studies to In Vivo Evaluation in Zebrafish. J Med Chem 2022; 65:16353-16371. [PMID: 36459415 PMCID: PMC7616001 DOI: 10.1021/acs.jmedchem.2c01174] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Emergence of resistance in cancer cells and dose-limiting side effects severely limit the widespread use of platinum (Pt) anticancer drugs. Multi-action hybrid anticancer agents that are constructed by merging two or more pharmacophores offer the prospect of circumventing issues of Pt drugs. Herein, we report the design, synthesis, and in-depth biological evaluation of a ruthenium-ferrocene (Ru-Fc) bimetallic agent [(η6-p-cymene)Ru(1,1,1-trifluoro-4-oxo-4-ferrocenyl-but-2-en-2-olate)Cl] and its five analogues. Along with aquation/anation chemistry, we evaluated the in vitro antitumor potency, Pt cross-resistance profile, and in vivo antiangiogenic properties. A structure activity analysis was performed to understand the impact of Fc, CF3, and p-cymene groups on the anticancer potency of the Ru-Fc hybrid. Finally, in addition to assessing cellular uptake and intracellular distribution, we demonstrated that the Ru-Fc hybrid binds to nucleophilic biomolecules and produces reactive oxygen species, which causes mitochondrial dysfunction and induces ER stress, leading to poly(ADP-ribose) polymerase-mediated necroptotic cell death.
Collapse
Affiliation(s)
- Manikandan M
- Medicinal Chemistry and Cell Biology Laboratory, Department of Chemical Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai, Maharashtra 400005, India
| | - Shubhankar Gadre
- Medicinal Chemistry and Cell Biology Laboratory, Department of Chemical Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai, Maharashtra 400005, India
| | - Sushanta Chhatar
- Medicinal Chemistry and Cell Biology Laboratory, Department of Chemical Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai, Maharashtra 400005, India
| | - Gourav Chakraborty
- Department of Developmental Biology, Agharkar Research Institute, G G Agarkar Road, Pune, Maharashtra 411004, India
| | - Naushad Ahmed
- Department of Chemistry, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana 502085, India
| | - Chinmoy Patra
- Department of Developmental Biology, Agharkar Research Institute, G G Agarkar Road, Pune, Maharashtra 411004, India
| | - Malay Patra
- Medicinal Chemistry and Cell Biology Laboratory, Department of Chemical Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Mumbai, Maharashtra 400005, India
| |
Collapse
|
28
|
Eidizade F, Soukhtanloo M, Zhiani R, Mehrzad J, Mirzavi F. Inhibition of glioblastoma proliferation, invasion, and migration by Urolithin B through inducing G0/G1 arrest and targeting MMP-2/-9 expression and activity. Biofactors 2022; 49:379-389. [PMID: 36310375 DOI: 10.1002/biof.1915] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
One kind of brain cancer with a dismal prognosis is called glioblastoma multiforme (GBM) due to its high growth rate and widespread tumor cell invasion into various areas of the brain. To improve therapeutic approaches, the objective of this research investigates the cytotoxic, anti-metastatic, and apoptotic effect of urolithin-B (UB) as a bioactive metabolite of ellagitannins (ETs) on GBM U87 cells. The malignant GBM cell line (U87) was examined for apoptosis rate, cell cycle analysis, cell viability, mRNA expressions of several apoptotic and metastasis-associated genes, production of reactive oxygen species (ROS), MMP-2, and MMP-9 activity and protein expression, and migration ability. The findings revealed that UB decreased U87 GBM viability in a dose-dependent manner and NIH/3T3 normal cells with the IC50 value of 30 and 55 μM after 24 h, respectively. UB also induces necrosis and G0/G1 cell cycle arrest in U87 cells. UB also increases ROS production and caused down-regulation of Bcl2 and up-regulation of Bax apoptotic genes. Additionally, treatment of UB reduced the migration of U87 cells. The protein levels, mRNA expression, and the MMP-2 and MMP-9 enzyme activities also decreased concentration-dependently. So, due to the non-toxic nature of UB and its ability to induce apoptosis and reduce the U87 GBM cell invasion and migration, after more research, it can be regarded as a promising new anti-GBM compound.
Collapse
Affiliation(s)
- Fateme Eidizade
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Mohammad Soukhtanloo
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rahele Zhiani
- Department of Chemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
- New Materials Technology and Processing Research Center, Department of Chemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Jamshid Mehrzad
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
29
|
Zhang D, Wang Z, Yamamoto N, Wang M, Yi X, Li P, Lin R, Nasimi Z, Okada K, Mochida K, Noutoshi Y, Zheng A. Secreted Glycosyltransferase RsIA_GT of Rhizoctonia solani AG-1 IA Inhibits Defense Responses in Nicotiana benthamiana. Pathogens 2022; 11:pathogens11091026. [PMID: 36145458 PMCID: PMC9501517 DOI: 10.3390/pathogens11091026] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/31/2022] [Accepted: 09/03/2022] [Indexed: 11/16/2022] Open
Abstract
Anastomosis group AG-1 IA of Rhizoctonia solani Khün has a wide host range and threatens crop production. Various glycosyltransferases secreted by phytopathogenic fungi play an essential role in pathogenicity. Previously, we identified a glycosyltransferase RsIA_GT (AG11A_09161) as a secreted protein-encoding gene of R. solani AG-1 IA, whose expression levels increased during infection in rice. In this study, we further characterized the virulence function of RsIA_GT. It is conserved not only in Basidiomycota, including multiple anastomosis groups of R. solani, but also in other primary fungal taxonomic categories. RsIA_GT possesses a signal peptide (SP) for protein secretion, and its functionality was proven using yeast and Nicotiana benthamiana. The SP-truncated form of RsIA_GT (RsIA_GT(ΔS)) expressed in Escherichia coli-induced lesion-like phenotype in rice leaves when applied to punched leaves. However, Agrobacterium-mediated transient expressions of both the full-length RsIA_GT and RsIA_GT(ΔS) did not induce cell death in N. benthamiana leaves. Instead, only RsIA_GT(ΔS) suppressed the cell death induced by two reference cell death factors BAX and INF1 in N.benthamiana. RsIA_GT(ΔS)R154A D168A D170A, a mutant RsIA_GT(ΔS) for the glycosyltransferase catalytic domain, still suppressed the BAX- or INF1-induced cell death, suggesting that the cell death suppression activity of RsIA_GT(ΔS) would be independent from its enzymatic activity. RsIA_GT(ΔS) also suppressed the H2O2 production and callose deposition and showed an effect on the induction of defense genes associated with the expression of BAX and INF1. The transient expression of RsIA_GT(ΔS) in N. benthamiana enhanced the lesion area caused by R. solani AG-1 IA. The secreted glycosyltransferase, RsIA_GT, of R. solani AG-1 IA is likely to have a dual role in virulence inside and outside of host cells.
Collapse
Affiliation(s)
- Danhua Zhang
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhaoyilin Wang
- Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Naoki Yamamoto
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingyue Wang
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaoqun Yi
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China
| | - Ping Li
- Rice Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Runmao Lin
- Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Zohreh Nasimi
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China
| | - Kazunori Okada
- Agro-Biotechnology Research Center, The University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Keiichi Mochida
- Bioproductivity Informatics Research Team, RIKEN Center for Sustainable Resource Science, Yokohama 2300045, Japan
- Microalgae Production Control Technology Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, Yokohama 2300045, Japan
- Kihara Institute for Biological Research, Yokohama City University, Yokohama 2440813, Japan
- School of Information and Data Sciences, Nagasaki University, Nagasaki 852-8521, Japan
| | - Yoshiteru Noutoshi
- Graduate School of Environmental and Life Science, Okayama University, Okayama 700-8530, Japan
| | - Aiping Zheng
- College of Agronomy, Sichuan Agricultural University, Chengdu 611130, China
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Chengdu 611130, China
- Correspondence:
| |
Collapse
|
30
|
The enhanced mitochondrial dysfunction by cantleyoside confines inflammatory response and promotes apoptosis of human HFLS-RA cell line via AMPK/Sirt 1/NF-κB pathway activation. Biomed Pharmacother 2022; 149:112847. [PMID: 35364376 DOI: 10.1016/j.biopha.2022.112847] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE Cantleyoside (CA) is a kind of iridoid glycosides in Pterocephalus hookeri (C. B. Clarke) Höeck. The purpose of this study was to investigate the effects of CA on human rheumatoid arthritis fibroblast synovial cells (HFLS-RA). METHODS Cell proliferation of HFLS-RA was assessed by CCK-8. ELISA was used to detect cytokines NO, TNF-α, IL-1β/6, MCP-1, MMP-1/3/9 and metabolism-related ATPase activities and ATP levels. JC-1, DCFH-DA, Fluo-3 AM and Calcein AM probes were used to detect mitochondrial membrane potential (MMP), reactive oxygen species (ROS), Ca2+ and mitochondrial permeability conversion pore (MPTP), respectively. Isolated mitochondria assay was used to detect mitochondrial swelling. Oxygen consumption rate (OCR), extracellular acidification rate (ECAR) and real-time ATP production were measured using a Seahorse analyzer. Apoptosis was detected by TUNEL and Hoechst staining. Western blot was used to detect the expressions of AMPK/p-AMPK, Sirt 1, IκBα, NF-κB p65/p-NF-κB p65, Bcl-2 and Bax. Cytoplasmic nuclear isolation was also performed to detect the translocation of NF-κB. RESULTS CA significantly suppressed cell proliferation and the levels of NO, TNF-α, IL-1β/6, MCP-1 and MMP-1/3/9 in HFLS-RA. In addition, CA promoted the apoptosis of HFLS-RA by increasing TUNEL and Hoechst positive cells and the ratio of Bax/Bcl-2. Inhibition of energy metabolism in HFLS-RA by CA reduced OCR, ECAR and real-time ATP generation rate. Importantly, CA promoted p-AMPK and Sirt 1 expression, inhibited IκBα degradation to reduce p-NF-κB and translocation. CONCLUSION The results suggest that CA activates the AMPK/Sirt 1/NF-κB pathway by promoting mitochondrial dysfunction, thereby exerting anti-inflammatory and pro-apoptotic effects.
Collapse
|
31
|
Erfani R, Carmichael C, Sofokleous T, Wang Q. Nanosecond-pulsed DBD plasma treatment on human leukaemia Jurkat cells and monoblastic U937 cells in vitro. Sci Rep 2022; 12:6270. [PMID: 35428374 PMCID: PMC9012873 DOI: 10.1038/s41598-022-10056-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 03/24/2022] [Indexed: 11/09/2022] Open
Abstract
Plasma therapy offers an exciting and novel way of cancer treatment. Specifically, it is shown that Jurkat death rates are closely governed by the plasma treatment time. However, apart from time, alterations to different parameters of treatment process may yield better results. Here, Dielectric barrier discharge (DBD) reactors excited by a nanosecond-pulse energy source are used to investigate cell viability for longer exposure times as well as the effects of polarity of reactor on treatment. Plasma discharge regimes are discussed and assessed using imaging and thermal imaging methods. We found that by changing the polarity of reactor i.e. changing the direction of plasma discharge, the plasma discharge regime changes influencing directly the effectiveness of treatment. Our results showed that ns-DBD- reactor could induce both apoptosis and necrosis of human Jurkat and U937 cells, and this cytotoxic effect of plasma was not completely antagonized by N-acetyl cysteine. It indicates that plasma could induce ROS-independent cell death. Gene expression analyses revealed that p53, BAD, BID and caspase 9 may play vital roles in plasma caused cell death. In addition, our findings demonstrate how different parameters can influence the effectiveness of our reactors. Our assay reveals the custom ability nature of plasma reactors for hematologic cancer therapy and our findings can be used for further development of such reactors using multi-objective optimisation techniques.
Collapse
Affiliation(s)
- Rasool Erfani
- Department of Engineering, Manchester Metropolitan University, Manchester, M1 5GD, UK. .,Department of Civil, Environmental and Geomatic Engineering, UCL, London, WC1E 6BT, UK.
| | - Cameron Carmichael
- Department of Engineering, Manchester Metropolitan University, Manchester, M1 5GD, UK
| | - Thea Sofokleous
- Department of Life Sciences, Manchester Metropolitan University, Manchester, M1 5GD, UK
| | - Qiuyu Wang
- Department of Life Sciences, Manchester Metropolitan University, Manchester, M1 5GD, UK
| |
Collapse
|
32
|
Huang L, Liu J, Bie C, Liu H, Ji Y, Chen D, Zhu M, Kuang W. Advances in cell death - related signaling pathways in acute-on-chronic liver failure. Clin Res Hepatol Gastroenterol 2022; 46:101783. [PMID: 34339873 DOI: 10.1016/j.clinre.2021.101783] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/14/2021] [Indexed: 02/04/2023]
Abstract
Acute-on-chronic liver failure (ACLF) has been a hot spot in the field of liver disease research in recent years, with high morbidity, rapid course change and high mortality. Currently, there is the absence of specific treatment in clinical practice. Liver transplantation has the best therapeutic effect, but it is prone to have internal environment disorder and liver cell death after transplantation, which leads to the failure of transplantation.In recent years, with the development of molecular biology, scholars have explored the treatment of ACLF at the molecular level, and more and more molecular signaling pathways related to the treatment of ACLF have been discovered. Modulating the relevant signaling pathways to reduce the mortality of liver cells after transplantation may effectively improve the success rate of transplantation. In this review, we introduce some signaling pathways related to cell death and their research progress in acute-on-chronic liver failure.
Collapse
Affiliation(s)
- Liqiao Huang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Jie Liu
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen 518104, China; The First Affiliated Hospital, Guangzhou TCM University, Guangzhou 510006, China
| | - Caiqun Bie
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen 518104, China
| | - Helu Liu
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen 518104, China
| | - Yichun Ji
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Dongfeng Chen
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Meiling Zhu
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen 518104, China.
| | - Weihong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Key Laboratory of Research and Development of New Medical Materials of Guangdong Medical University, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
33
|
Xie Y, Xing Z, Wei J, Sun X, Zhao B, Chen Y, Geng Y, Jia Z, Zou H. Levosimendan Postconditioning Attenuates Cardiomyocyte Apoptosis after Myocardial Infarction. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:2988756. [PMID: 35132355 PMCID: PMC8817859 DOI: 10.1155/2022/2988756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/08/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND Levosimendan preconditioning has been shown to attenuate myocardial apoptosis in animal models. However, protective effects of levosimendan postconditioning against myocardial apoptosis following myocardial infarction (MI) have not been evaluated. Therefore, we investigated the effects of levosimendan postconditioning on myocardial apoptosis in MI rat models. METHODS In an anoxia/reoxygenation (A/R) model, H9c2 cells were pretreated with or without levosimendan postconditioning after which their apoptosis rates were assessed by flow cytometry, RT-qPCR, and western blot analyses. Then, postconditioning was performed with or without levosimendan in MI rat models. Myocardiocyte apoptosis was evaluated by echocardiography, TTC staining, TUNEL staining, immunohistochemical staining, RT-qPCR, and western blot analysis. RESULTS Levosimendan postconditioning inhibited H9c2 cell apoptosis in A/R models by elevating Bcl-2 while suppressing Caspase-3 and Bax at both mRNA and protein levels. Moreover, it improved cardiac functions and reduced the left ventricle infarction area in MI rat models. Compared to the MI control group, cardiomyocyte apoptosis rates in the levosimendan postconditioning group were low. The reduced cardiomyocyte apoptosis rates were associated with downregulation of Bax and Caspase-3 as well as with upregulation of Bcl-2 at mRNA and protein levels. CONCLUSIONS Levosimendan postconditioning of MI rat models protected against cardiomyocyte apoptosis, implying that it is a potential strategy for preventing cardiomyocyte apoptosis in the treatment of cardiac dysfunction following MI.
Collapse
Affiliation(s)
- Ying Xie
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, No. 245,Renmin East Road, Kunming, Yunnan Province 650051, China
| | - Zhengjiang Xing
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, No. 245,Renmin East Road, Kunming, Yunnan Province 650051, China
| | - Jie Wei
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, No. 245,Renmin East Road, Kunming, Yunnan Province 650051, China
| | - Xiaolin Sun
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, No. 245,Renmin East Road, Kunming, Yunnan Province 650051, China
| | - Bin Zhao
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, No. 245,Renmin East Road, Kunming, Yunnan Province 650051, China
| | - Yan Chen
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, No. 245,Renmin East Road, Kunming, Yunnan Province 650051, China
| | - Yue Geng
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, No. 245,Renmin East Road, Kunming, Yunnan Province 650051, China
| | - Zheng Jia
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, No. 245,Renmin East Road, Kunming, Yunnan Province 650051, China
| | - Honglin Zou
- Department of Cardiovascular Surgery, Yan'an Hospital Affiliated to Kunming Medical University, No. 245,Renmin East Road, Kunming, Yunnan Province 650051, China
| |
Collapse
|
34
|
Patel P, Mendoza A, Robichaux DJ, Wang MC, Wehrens XHT, Karch J. Inhibition of the Anti-Apoptotic Bcl-2 Family by BH3 Mimetics Sensitize the Mitochondrial Permeability Transition Pore Through Bax and Bak. Front Cell Dev Biol 2021; 9:765973. [PMID: 34926454 PMCID: PMC8672142 DOI: 10.3389/fcell.2021.765973] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/12/2021] [Indexed: 12/24/2022] Open
Abstract
Mitochondrial permeability transition pore (MPTP)-dependent necrosis contributes to numerous pathologies in the heart, brain, and skeletal muscle. The MPTP is a non-selective pore in the inner mitochondrial membrane that is triggered by high levels of matrix Ca2+, and sustained opening leads to mitochondrial dysfunction. Although the MPTP is defined by an increase in inner mitochondrial membrane permeability, the expression of pro-apoptotic Bcl-2 family members, Bax and Bak localization to the outer mitochondrial membrane is required for MPTP-dependent mitochondrial dysfunction and subsequent necrotic cell death. Contrary to the role of Bax and Bak in apoptosis, which is dependent on their oligomerization, MPTP-dependent necrosis does not require oligomerization as monomeric/inactive forms of Bax and Bak can facilitate mitochondrial dysfunction. However, the relationship between Bax and Bak activation/oligomerization and MPTP sensitization remains to be explored. Here, we use a combination of in vitro and ex vivo approaches to determine the role of the anti-apoptotic Bcl-2 family members, which regulate Bax/Bak activity, in necrotic cell death and MPTP sensitivity. To study the role of each predominantly expressed anti-apoptotic Bcl-2 family member (i.e., Mcl-1, Bcl-2, and Bcl-xL) in MPTP regulation, we utilize various BH3 mimetics that specifically bind to and inhibit each. We determined that the inhibition of each anti-apoptotic Bcl-2 family member lowers mitochondrial calcium retention capacity and sensitizes MPTP opening. Furthermore, the inhibition of each Bcl-2 family member exacerbates both apoptotic and necrotic cell death in vitro in a Bax/Bak-dependent manner. Our findings suggests that mitochondrial Ca2+ retention capacity and MPTP sensitivity is influenced by Bax/Bak activation/oligomerization on the outer mitochondrial membrane, providing further evidence of the crosstalk between the apoptotic and necrotic cell death pathways.
Collapse
Affiliation(s)
- Pooja Patel
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| | - Arielys Mendoza
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| | - Dexter J Robichaux
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| | - Meng C Wang
- Huffington Center on Aging, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, United States
| | - Xander H T Wehrens
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| | - Jason Karch
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States.,Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
35
|
Shi S, Wang L, van der Laan LJW, Pan Q, Verstegen MMA. Mitochondrial Dysfunction and Oxidative Stress in Liver Transplantation and Underlying Diseases: New Insights and Therapeutics. Transplantation 2021; 105:2362-2373. [PMID: 33577251 PMCID: PMC9005104 DOI: 10.1097/tp.0000000000003691] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/09/2021] [Accepted: 01/16/2021] [Indexed: 12/06/2022]
Abstract
Mitochondria are essential organelles for cellular energy and metabolism. Like with any organ, the liver highly depends on the function of these cellular powerhouses. Hepatotoxic insults often lead to an impairment of mitochondrial activity and an increase in oxidative stress, thereby compromising the metabolic and synthetic functions. Mitochondria play a critical role in ATP synthesis and the production or scavenging of free radicals. Mitochondria orchestrate many cellular signaling pathways involved in the regulation of cell death, metabolism, cell division, and progenitor cell differentiation. Mitochondrial dysfunction and oxidative stress are closely associated with ischemia-reperfusion injury during organ transplantation and with different liver diseases, including cholestasis, steatosis, viral hepatitis, and drug-induced liver injury. To develop novel mitochondria-targeting therapies or interventions, a better understanding of mitochondrial dysfunction and oxidative stress in hepatic pathogenesis is very much needed. Therapies targeting mitochondria impairment and oxidative imbalance in liver diseases have been extensively studied in preclinical and clinical research. In this review, we provide an overview of how oxidative stress and mitochondrial dysfunction affect liver diseases and liver transplantation. Furthermore, we summarize recent developments of antioxidant and mitochondria-targeted interventions.
Collapse
Affiliation(s)
- Shaojun Shi
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Ling Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
36
|
Basiglio CL, Crocenzi FA, Sánchez Pozzi EJ, Roma MG. Oxidative Stress and Localization Status of Hepatocellular Transporters: Impact on Bile Secretion and Role of Signaling Pathways. Antioxid Redox Signal 2021; 35:808-831. [PMID: 34293961 DOI: 10.1089/ars.2021.0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Significance: Most hepatopathies are primarily or secondarily cholestatic in nature. Oxidative stress (OS) is a frequent trait among them, and impairs the machinery to generate bile by triggering endocytic internalization of hepatocellular transporters, thus causing cholestasis. This is critical, since it leads to accelerated transporter degradation, which could explain the common post-transcriptional downregulation of transporter expression in human cholestatic diseases. Recent Advances: The mechanisms involved in OS-induced hepatocellular transporter internalization are being revealed. Filamentous actin (F-actin) cytoskeleton disorganization and/or detachment of crosslinking actin proteins that afford transporter stability have been characterized as causal factors. Activation of redox-sensitive signaling pathways leading to changes in phosphorylation status of these structures is involved, including Ca2+-mediated activation of "classical" and "novel" protein kinase C (PKC) isoforms or redox-signaling cascades downstream of NADPH oxidase. Critical Issues: Despite the well-known occurrence of hepatocellular transporter internalization in human hepatopathies, the cholestatic implications of this phenomenon have been overlooked. Accordingly, no specific treatment has been established in the clinical practice for its prevention/reversion. Future Directions: We need to improve our knowledge on the pro-oxidant triggering factors and the multiple signaling pathways that mediate this oxidative injury in each cholestatic hepatopathy, so as to envisage tailor-made therapeutic strategies for each case. Meanwhile, administration of antioxidants or heme oxygenase-1 induction to elevate the hepatocellular levels of the endogenous scavenger bilirubin are promising alternatives that need to be re-evaluated and implemented. They may complement current treatments in cholestasis aimed to enhance transcriptional carrier expression, by providing membrane stability to the newly synthesized carriers. Antioxid. Redox Signal. 35, 808-831.
Collapse
Affiliation(s)
- Cecilia L Basiglio
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| | - Fernando A Crocenzi
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| | - Enrique J Sánchez Pozzi
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| | - Marcelo G Roma
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| |
Collapse
|
37
|
Pires AS, Varela CL, Marques IA, Abrantes AM, Gonçalves C, Rodrigues T, Matafome P, Botelho MF, Roleira FMF, Tavares-da-Silva E. Oxymestane, a cytostatic steroid derivative of exemestane with greater antitumor activity in non-estrogen-dependent cell lines. J Steroid Biochem Mol Biol 2021; 212:105950. [PMID: 34271024 DOI: 10.1016/j.jsbmb.2021.105950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/29/2021] [Accepted: 07/11/2021] [Indexed: 02/06/2023]
Abstract
A new promising steroid derivative of Exemestane (Exe), the drug used for the treatment of estrogen-dependent breast cancer, was synthesized and evaluated against a set of human cancer cell lines. The new compound (Oxymestane-D1, Oxy) was tested comparatively with Exe against colon (C2BBe1, WiDr), liver (HepG2, HuH-7), lung (A549, H1299) and prostate (LNCaP, PC3) human cancer cell lines. Likewise, its effect on human colon normal cells (CCD-841 CoN) and human normal fibroblast cells (HFF-1) was studied. The cytostatic activity of Oxy was also compared with that of the reference cytostatic drugs used in chemotherapy protocols, namely carboplatin, cisplatin, doxorubicin, epirubicin, etoposide, flutamide, 5-fluorouracil, irinotecan, oxaliplatin and sorafenib. In all cell lines tested, Oxy proved to be more powerful cytostatic than Exe. Additionally, the IC50 at 72 h showed a three-fold activity greater than 5-fluorouracil in the WiDr cell line, twice as high as cisplatin for cell line A549 and five times higher than cisplatin for cell line H1299. Also, Oxy surprisingly revealed to induce DNA damage and inhibit the DNA damage response (DDR) proteins ATM, ATR, CHK1 and CHK2. The results obtained allow concluding that Oxy can be a promising anticancer agent to be used in chemotherapy protocols. Furthermore, its ability to inhibit crucial components of DDR can also be useful for the monotherapy or for combination with chemo and/or radiotherapy of cancer.
Collapse
Affiliation(s)
- Ana S Pires
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal; Clinical Academic Center of Coimbra, Praceta Prof. Mota Pinto, Coimbra, 3004-561, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal.
| | - Carla L Varela
- University of Coimbra, CIEPQPF, FFUC, Laboratory of Pharmaceutical Chemistry, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal.
| | - Inês A Marques
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal; Clinical Academic Center of Coimbra, Praceta Prof. Mota Pinto, Coimbra, 3004-561, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal; University of Coimbra, Faculty of Pharmacy, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal.
| | - Ana M Abrantes
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal; Clinical Academic Center of Coimbra, Praceta Prof. Mota Pinto, Coimbra, 3004-561, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal.
| | - Cristina Gonçalves
- Clinical Academic Center of Coimbra, Praceta Prof. Mota Pinto, Coimbra, 3004-561, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal; University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Laboratory of Oncobiology and Hematology and University Clinic of Hematology of Faculty of Medicine, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal.
| | - Tiago Rodrigues
- Clinical Academic Center of Coimbra, Praceta Prof. Mota Pinto, Coimbra, 3004-561, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal; University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Laboratory of Physiology of Faculty of Medicine, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal.
| | - Paulo Matafome
- Clinical Academic Center of Coimbra, Praceta Prof. Mota Pinto, Coimbra, 3004-561, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal; University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Laboratory of Physiology of Faculty of Medicine, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal.
| | - Maria F Botelho
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal; Clinical Academic Center of Coimbra, Praceta Prof. Mota Pinto, Coimbra, 3004-561, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal.
| | - Fernanda M F Roleira
- University of Coimbra, CIEPQPF, FFUC, Laboratory of Pharmaceutical Chemistry, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal.
| | - Elisiário Tavares-da-Silva
- University of Coimbra, CIEPQPF, FFUC, Laboratory of Pharmaceutical Chemistry, Azinhaga de Santa Comba, Pólo III - Pólo das Ciências da Saúde, Coimbra, 3000-548, Portugal.
| |
Collapse
|
38
|
MicroRNA MiR-27a-5p Alleviates the Cerulein-Induced Cell Apoptosis and Inflammatory Injury of AR42J Cells by Targeting Traf3 in Acute Pancreatitis. Inflammation 2021; 43:1988-1998. [PMID: 32647955 DOI: 10.1007/s10753-020-01272-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Acute pancreatitis (AP), a sudden inflammatory process of pancreas, is painful and may contribute to death. The aberrant expression of miR-27a-5p has been reported in many types of cancers and diseases including AP. Thus, it is urgent to manifest the functions and mechanism of miR-27a-5p in AP. The levels of miR-27a-5p, tumor necrosis factor (TNF) receptor-associated factor 3 (Traf3) in serum of AP patient, or cerulein-treated AR42J cells were detected by qRT-PCR. Functionally, the apoptotic rate, the protein levels of Bcl-2 and Bax, the caspase-3 activity, and the levels of IL-1β, IL-6, and TNF-α in cerulein-treated AR42J cells were measured by flow cytometry, Western blot, caspase-3 activity assay, and qRT-PCR and ELISA assay, respectively. In addition, the putative target of miR-27a-5p was predicted by TargetScan online database, and the dual luciferase reporter assay and RNA immunoprecipitation (RIP) assay were conducted to verify this interaction. Cerulein-treated mouse AP model was established to explore the role of miR-27a-5p in AP in vivo. The level of miR-27a-5p was notably downregulated in AP patients and cerulein-treated AR42J cells. The functional experiments indicated that miR-27a-5p mimics attenuated the promotion effects on cell apoptosis and the inflammatory response in AR42J cells caused by cerulein. The interaction between miR-27a-5p and Traf3 was predicted by TargetScan online database and validated by dual luciferase reporter assay and RIP assay. Following qRT-PCR results exhibited that Traf3 was apparently enhanced in cerulein-treated AR42J cells. The further functional experiments disclosed that Traf3 overexpression relieved the inhibitory effects on cell apoptosis and the inflammatory response induced by miR-27a-5p mimics in cerulein-treated AR42J cells. Moreover, miR-27a-5p alleviated cerulein-induced injury in vivo. In this study, we established the cerulein-treated AR42J cells as AP model in vitro. We validated that miR-27a-5p was significantly downregulated, and Traf3 was strikingly upregulated in AP patient and/or cerulein-treated AR42J cells. The further mechanistical and functional experiments unraveled that miR-27a-5p regulated Traf3 to relieve the cerulein-induced cell apoptosis and inflammatory injury of AR42J cells. Therefore, this novel regulatory network may provide therapeutic target for AP patients.
Collapse
|
39
|
Santra P, Amack JD. Loss of vacuolar-type H+-ATPase induces caspase-independent necrosis-like death of hair cells in zebrafish neuromasts. Dis Model Mech 2021; 14:dmm048997. [PMID: 34296747 PMCID: PMC8319552 DOI: 10.1242/dmm.048997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/15/2021] [Indexed: 01/24/2023] Open
Abstract
The vacuolar-type H+-ATPase (V-ATPase) is a multi-subunit proton pump that regulates cellular pH. V-ATPase activity modulates several cellular processes, but cell-type-specific functions remain poorly understood. Patients with mutations in specific V-ATPase subunits can develop sensorineural deafness, but the underlying mechanisms are unclear. Here, we show that V-ATPase mutations disrupt the formation of zebrafish neuromasts, which serve as a model to investigate hearing loss. V-ATPase mutant neuromasts are small and contain pyknotic nuclei that denote dying cells. Molecular markers and live imaging show that loss of V-ATPase induces mechanosensory hair cells in neuromasts, but not neighboring support cells, to undergo caspase-independent necrosis-like cell death. This is the first demonstration that loss of V-ATPase can lead to necrosis-like cell death in a specific cell type in vivo. Mechanistically, loss of V-ATPase reduces mitochondrial membrane potential in hair cells. Modulating the mitochondrial permeability transition pore, which regulates mitochondrial membrane potential, improves hair cell survival. These results have implications for understanding the causes of sensorineural deafness, and more broadly, reveal functions for V-ATPase in promoting survival of a specific cell type in vivo.
Collapse
Affiliation(s)
- Peu Santra
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
| | - Jeffrey D. Amack
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210, USA
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse, NY 13244, USA
| |
Collapse
|
40
|
Ramos PM, Bell LC, Wohlgemuth SE, Scheffler TL. Mitochondrial Function in Oxidative and Glycolytic Bovine Skeletal Muscle Postmortem. MEAT AND MUSCLE BIOLOGY 2021. [DOI: 10.22175/mmb.11698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Meat quality is traditionally associated with anaerobic metabolism due to cessation of the oxygen supply post-mortem. However, mitochondrial (mt) function early postmortem may affect the development of meat quality characteristics, such as adenosine triphosphate levels and pH decline. Therefore, the objective of this study was to evaluate mt function ex vivo during the first 24 h postmortem in muscles with differences in mt content. Samples from longissimus lumborum (LL) and diaphragm (Dia) were taken from steers (n = 6) at 1, 3, and 24 h postmortem and frozen to determine citrate synthase (CS) activity and mt protein expression (immunodetection) or were fresh-preserved for high-resolution respirometry. Integrative oxygen consumption rate (picomoles per second per milligram of tissue) was measured and normalized to CS activity as a proxy for mt content (intrinsic mt function, picomoles per second per unit CS). CS activity (P < 0.001) and mt protein expression (P < 0.001) were greater in Dia, which was reflected in mt respiration. Muscle type affected (P < 0.001) integrative leak respiration and was greater in mt from Dia; oxidative phosphorylation (OXPHOS) was also greater in Dia and influenced by time postmortem (muscle × time: P = 0.01). Intrinsic leak and OXPHOS were affected by muscle and time (muscle × time: P = 0.05 and P = 0.01, respectively), with the most pronounced differences at 24 h postmortem. Stimulation of OXPHOS by cytochrome c as an indicator of outer mt membrane integrity was influenced by muscle and time postmortem (muscle × time: P = 0.03); it was greater in mt from LL. Despite intrinsic differences in respiratory function at 24 h, mt from both muscles were intact and coupled at 1 h postmortem. Reduced content and respiratory function in mt from LL are associated with early fragmentation, which could impact protease activation and subsequently meat quality.
Collapse
|
41
|
Zheng XX, Li YC, Yang KL, He ZX, Wang ZL, Wang X, Jing HL, Cao YJ. Icariin reduces Glu-induced excitatory neurotoxicity via antioxidative and antiapoptotic pathways in SH-SY5Y cells. Phytother Res 2021; 35:3377-3389. [PMID: 33891785 DOI: 10.1002/ptr.7057] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 01/31/2021] [Accepted: 02/05/2021] [Indexed: 01/06/2023]
Abstract
Excessive glutamate (Glu) can lead to significant effects on neural cells through the generation of neurotoxic or excitotoxic cascades. Icariin (ICA) is a main active ingredient of Chinese Medicine Berberidaceae epimedium L., and has many biological activities, such as antiinflammation, antioxidative stress, and anti-depression. This study aims to evaluate the effect of ICA on Glu-induced excitatory neurotoxicity of SH-SY5Y cells. The cell viability assay was evaluated by the CCK-8 assay. The apoptosis, reactive oxygen species (ROS), and mitochondrial membrane potential were assessed by flow cytometry. Intracellular Ca2+ concentration was determined by using the fluorescent probe Fluo-3. Protein expression was detected by western blotting analysis. ICA can significantly enhance the SH-SY5Y cell viability reduced by Glu. At the same time, ICA can significantly reduce apoptosis, ROS, nitric oxide (NO) levels, and intracellular Ca2+ concentration, and significantly inhibit the increase of mitochondrial membrane potential. In addition, ICA significantly increased the expression of P47phox and iNOS, decreased p-JNK/JNK, p-P38/P38, Bax/Bcl-2, active caspase-3, and active caspase-9. These results indicate that ICA may reduce the excitatory neurotoxicity of Glu-induced SH-SY5Y cells through suppression of oxidative stress and apoptotic pathways, suggesting that ICA could be a potential therapeutic candidate for neurological disorders propagated by Glu toxicity.
Collapse
Affiliation(s)
- Xing Xing Zheng
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, Xi'an, PR China.,Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, PR China
| | - Ying Chun Li
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, Xi'an, PR China.,Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, PR China
| | - Kai Lin Yang
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, Xi'an, PR China.,Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, PR China
| | - Zhou Xiao He
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, Xi'an, PR China.,Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, PR China
| | - Zhao Liang Wang
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, Xi'an, PR China.,Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, PR China
| | - Xing Wang
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, Xi'an, PR China.,Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, PR China
| | - Hui Ling Jing
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, Xi'an, PR China.,Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, PR China
| | - Yan Jun Cao
- Biomedicine Key Laboratory of Shaanxi Province, School of Pharmacy, Northwest University, Xi'an, PR China.,Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, Xi'an, PR China
| |
Collapse
|
42
|
Wang H, Huang M, Bei W, Yang Y, Song L, Zhang D, Zhan W, Zhang Y, Chen X, Wang W, Wang L, Guo J. FTZ attenuates liver steatosis and fibrosis in the minipigs with type 2 diabetes by regulating the AMPK signaling pathway. Biomed Pharmacother 2021; 138:111532. [PMID: 34311531 DOI: 10.1016/j.biopha.2021.111532] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/30/2022] Open
Abstract
Fufang Zhenzhu Tiaozhi formula (FTZ), a preparation of Chinese herbal medicine, has various pharmacological properties, such as hypoglycemic, hypolipidemic, anticoagulant, and anti-inflammatory activities. Hepatocyte apoptosis is a marker of nonalcoholic steatohepatitis (NASH) and contributes to liver injury, fibrosis, and inflammation. Given the multiple effects of FTZ, we investigated whether FTZ can be a therapeutic agent for NASH and its mechanism. In the present study, we observed that FTZ treatment had an obviously favorable influence on hepatic steatosis and fibrosis in the histopathologic features of type 2 diabetes mellitus (T2DM) and coronary heart disease (CHD) with NASH minipigs. In addition, immunohistochemical analysis showed increased expression of the fibrotic marker α-smooth muscle actin (α-SMA), and a TUNEL assay revealed increased apoptotic positive hepatic cells in the liver tissues of the model group. Furthermore, FTZ administration reduced the increased expression of α-SMA, and FTZ inhibited apoptosis by affecting Bcl-2/Bax and cleaved caspase-3 expression. Mechanistically, our data suggested that FTZ treatment attenuated hepatic steatosis and fibrosis via the adenosine monophosphate-activated protein kinase (AMPK) pathway. In vitro studies showed that FTZ also attenuated intracellular lipid accumulation in HepG2 cells exposed to palmitic acid (PA) and oleic acid (OA). FTZ upregulated the expression levels of P-AMPK and BCL-2 and downregulated BAX. The changes induced by FTZ were reversed by Compound C, an inhibitor of AMPK. In conclusion, FTZ attenuated NASH by ameliorating steatosis and hepatocyte apoptosis, which is attributable to the regulation of the AMPK pathway.
Collapse
Affiliation(s)
- Hong Wang
- Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Minyi Huang
- Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Weijian Bei
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Yiqi Yang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China; Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Lixia Song
- Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Dongxing Zhang
- Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Wenjing Zhan
- Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Yuzhen Zhang
- Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Xu Chen
- Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China
| | - Weixuan Wang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China; Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Lexun Wang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China; Guangdong TCM Key Laboratory against Metabolic Diseases, China
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Key Unit of Modulating Liver to Treat Hyperlipemia SATCM (State Administration of Traditional Chinese Medicine), China; Institute of Chinese Medicinal Sciences, Guangdong Pharmaceutical University, China; Guangdong TCM Key Laboratory against Metabolic Diseases, China.
| |
Collapse
|
43
|
Song Y, Wang X, Wang X, Wang J, Hao Q, Hao J, Hou X. Osthole-Loaded Nanoemulsion Enhances Brain Target in the Treatment of Alzheimer's Disease via Intranasal Administration. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8844455. [PMID: 33564364 PMCID: PMC7850840 DOI: 10.1155/2021/8844455] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 01/04/2021] [Accepted: 01/09/2021] [Indexed: 02/06/2023]
Abstract
Osthole (OST) is a natural coumarin compound that exerts multiple pharmacologic effects. However, the poor water solubility and the low oral absorption of OST limit its clinical application for the treatment of neurologic diseases. A suitable preparation needs to be tailored to evade these unfavourable properties of OST. In this study, an OST nanoemulsion (OST-NE) was fabricated according to the pseudoternary phase diagram method, which was generally used to optimize the prescription in light of the solubility of OST in surfactants and cosurfactants. The final composition of OST-NE was 3.6% of ethyl oleate as oil phase, 11.4% of the surfactant (polyethylene glycol ester of 15-hydroxystearic acid: polyoxyethylene 35 castor oil = 1 : 1), 3% of polyethylene glycol 400 as cosurfactant, and 82% of the aqueous phase. The pharmacokinetic study of OST-NE showed that the brain-targeting coefficient of OST was larger by the nasal route than that by the intravenous route. Moreover, OST-NE inhibited cell death, decreased the apoptosis-related proteins (Bax and caspase-3), and enhanced the activity of antioxidant enzymes (superoxide dismutase and glutathione) in L-glutamate-induced SH-SY5Y cells. OST-NE improved the spatial memory ability, increased the acetylcholine content in the cerebral cortex, and decreased the activity of acetylcholinesterase in the hippocampus of Alzheimer's disease model mice. In conclusion, this study indicates that the bioavailability of OST was improved by using the OST-NE via the nasal route. A low dose of OST-NE maintained the neuroprotective effects of OST, such as inhibiting apoptosis and oxidative stress and regulating the cholinergic system. Therefore, OST-NE can be used as a possible alternative to improve its bioavailability in the prevention and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Yilei Song
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Xiangyu Wang
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Xingrong Wang
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Jianze Wang
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Qiulian Hao
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Jifu Hao
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Xueqin Hou
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| |
Collapse
|
44
|
Yang P, Shao Z, Besley NA, Neal SE, Buehne KL, Park J, Karageozian H, Karageozian V, Ryde IT, Meyer JN, Jaffe GJ. Risuteganib Protects against Hydroquinone-induced Injury in Human RPE Cells. Invest Ophthalmol Vis Sci 2021; 61:35. [PMID: 32818234 PMCID: PMC7443126 DOI: 10.1167/iovs.61.10.35] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Cigarette smoking has been implicated in the pathogenesis of AMD. Integrin dysfunctions have been associated with AMD. Herein, we investigate the effect of risuteganib (RSG), an integrin regulator, on RPE cell injury induced by hydroquinone (HQ), an important oxidant in cigarette smoke. Methods Cultured human RPE cells were treated with HQ in the presence or absence of RSG. Cell death, mitochondrial respiration, reactive oxygen species production, and mitochondrial membrane potential were measured by flow cytometry, XFe24 analyzer, and fluorescence plate reader, respectively. Whole transcriptome analysis and gene expression were analyzed by Illumina RNA sequencing and quantitative PCR, respectively. F-actin aggregation was visualized with phalloidin. Levels of heme oxygenase-1, P38, and heat shock protein 27 proteins were measured by Western blot. Results HQ induced necrosis and apoptosis, decreased mitochondrial bioenergetics, increased reactive oxygen species levels, decreased mitochondrial membrane potential, increased F-actin aggregates, and induced phosphorylation of P38 and heat shock protein 27. HQ, but not RSG alone, induced substantial transcriptome changes that were regulated by RSG cotreatment. RSG cotreatment significantly protected against HQ-induced necrosis and apoptosis, prevented HQ-reduced mitochondrial bioenergetics, decreased HQ-induced reactive oxygen species production, improved HQ-disrupted mitochondrial membrane potential, reduced F-actin aggregates, decreased phosphorylation of P38 and heat shock protein 27, and further upregulated HQ-induced heme oxygenase-1 protein levels. Conclusions RSG has no detectable adverse effects on healthy RPE cells, whereas RSG cotreatment protects against HQ-induced injury, mitochondrial dysfunction, and actin reorganization, suggesting a potential role for RSG therapy to treat retinal diseases such as AMD.
Collapse
Affiliation(s)
- Ping Yang
- Department of Ophthalmology, Duke University Eye Center, Durham, North Carolina, United States
| | - Zixuan Shao
- Allegro Ophthalmics, LLC, San Juan Capistrano, California, United States
| | - Nicholas A Besley
- Department of Ophthalmology, Duke University Eye Center, Durham, North Carolina, United States
| | - Samantha E Neal
- Department of Ophthalmology, Duke University Eye Center, Durham, North Carolina, United States
| | - Kristen L Buehne
- Department of Ophthalmology, Duke University Eye Center, Durham, North Carolina, United States
| | - John Park
- Allegro Ophthalmics, LLC, San Juan Capistrano, California, United States
| | - Hampar Karageozian
- Allegro Ophthalmics, LLC, San Juan Capistrano, California, United States
| | - Vicken Karageozian
- Allegro Ophthalmics, LLC, San Juan Capistrano, California, United States
| | - Ian T Ryde
- Nicholas School of the Environment, Duke University, Durham, North Carolina, United States
| | - Joel N Meyer
- Nicholas School of the Environment, Duke University, Durham, North Carolina, United States
| | - Glenn J Jaffe
- Department of Ophthalmology, Duke University Eye Center, Durham, North Carolina, United States
| |
Collapse
|
45
|
Regulated Necrotic Cell Death in Alternative Tumor Therapeutic Strategies. Cells 2020; 9:cells9122709. [PMID: 33348858 PMCID: PMC7767016 DOI: 10.3390/cells9122709] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022] Open
Abstract
The treatment of tumors requires the induction of cell death. Radiotherapy, chemotherapy, and immunotherapy are administered to kill cancer cells; however, some cancer cells are resistant to these therapies. Therefore, effective treatments require various strategies for the induction of cell death. Regulated cell death (RCD) is systematically controlled by intracellular signaling proteins. Apoptosis and autophagy are types of RCD that are morphologically different from necrosis, while necroptosis, pyroptosis, and ferroptosis are morphologically similar to necrosis. Unlike necrosis, regulated necrotic cell death (RNCD) is caused by disruption of the plasma membrane under the control of specific proteins and induces tissue inflammation. Various types of RNCD, such as necroptosis, pyroptosis, and ferroptosis, have been used as therapeutic strategies against various tumor types. In this review, the mechanisms of necroptosis, pyroptosis, and ferroptosis are described in detail, and a potential effective treatment strategy to increase the anticancer effects on apoptosis- or autophagy-resistant tumor types through the induction of RNCD is suggested.
Collapse
|
46
|
Wang L, Gong J, Wang J, Dan J, Wang P. Long Non-coding RNA MALAT1 Alleviates the Elevated Intraocular Pressure (Eiop)-induced Glaucoma Progression via Sponging miR-149-5p. Curr Eye Res 2020; 46:903-911. [PMID: 33108931 DOI: 10.1080/02713683.2020.1843686] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Background: Glaucoma is an optic neuropathic disease and contributed to the irreversible blindness caused by the slow death of retinal ganglion cells (RGCs). Long non-coding RNA (lncRNA) metastasis associated lung adenocarcinoma transcript 1 (MALAT1) was reported to be aberrantly expressed in diverse diseases, including glaucoma. However, the mechanism of MALAT1 in glaucoma was still undefined.Methods: The levels of MALAT1, microRNA-149-5p (miR-149-5p) in RGCs cultured under elevated pressure were measured by quantitative real-time polymerase chain reaction (qRT-PCR). The putative target of MALAT1 was predicted by starBase v2.0 online database, and dual luciferase reporter assay, RNA immunoprecipitation (RIP) assay and RNA pull-down assay were performed to verify this interaction. The cell viability of RGCs was measured by Cell Counting Kit-8 (CCK-8) assay. The apoptotic rate was evaluated via flow cytometry. The protein levels of apoptosis-related proteins (Bax, B-cell lymphoma 2 (Bcl-2)) and Cleaved caspase 3 were assessed by Western blot.Results: The level of MALAT1 was significantly down-regulated, and the level of miR-149-5p was distinctly up-regulated in RGCs under pressure in a dose-dependent manner. Functionally, MALAT1 overexpression or miR-149-5p inhibitor alleviated the inhibitory effect on cell viability and the promoted effect on apoptotic rate of RGCs in EIOP. The interaction between MALAT1 and miR-149-5p was predicted by starBase v2.0 online database, and dual luciferase reporter assay, RIP assay and RNA pull-down assay validated the interaction. Combined with the loss and gain experiment results, miR-149-5p was negatively interacted with MALAT1. Furthermore, miR-149-5p mimics mitigated the promoted impact on cell viability and the suppressive impact on apoptotic rate by targeting MALAT1.Conclusion: MALAT1 promoted cell proliferation and inhibited cell apoptosis of RGCs via targeting miR-149-5p in glaucoma in vitro, which might shed light on the mechanism of glaucoma pathogenesis.
Collapse
Affiliation(s)
- Linling Wang
- The Affiliated Renhe Hospital of China Three Gorges University (The Second Clinical Medical College of China Three Gorges University), Yichang, Hubei, China
| | - Jin Gong
- The Affiliated Renhe Hospital of China Three Gorges University (The Second Clinical Medical College of China Three Gorges University), Yichang, Hubei, China
| | - Junling Wang
- The Affiliated Renhe Hospital of China Three Gorges University (The Second Clinical Medical College of China Three Gorges University), Yichang, Hubei, China
| | - Jing Dan
- The Affiliated Renhe Hospital of China Three Gorges University (The Second Clinical Medical College of China Three Gorges University), Yichang, Hubei, China
| | - Ping Wang
- The Affiliated Renhe Hospital of China Three Gorges University (The Second Clinical Medical College of China Three Gorges University), Yichang, Hubei, China
| |
Collapse
|
47
|
Zhang M, Cheng K, Chen H, Tu J, Shen Y, Pang L, Wu W. Galectin-3 knock down inhibits cardiac ischemia-reperfusion injury through interacting with bcl-2 and modulating cell apoptosis. Arch Biochem Biophys 2020; 694:108602. [PMID: 32980351 DOI: 10.1016/j.abb.2020.108602] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/16/2020] [Accepted: 09/23/2020] [Indexed: 01/12/2023]
Abstract
Acute myocardial infarction (AMI) is a fetal cardiovascular disease with high morbidity and mortality worldwide. In the present study, we elucidated the role of galectin-3 in preventing myocardial ischemic reperfusion injury. We found that galactin-3 was significantly up-regulated in the myocardium and cardiomyocyte subjected to ischemia/reperfusion (I/R) and hypoxia/reoxygenation (H/R) treatment, respectively. Galectin-3 knockdown significantly decreased the ischemic size of the left ventricular and the apoptosis of cardiomyocytes. Moreover, galectin-3 knockdown reversed the decrease of mitochondrial membrane potential and inhibited the inflammation response in myocardium and cultured cardiomyocyte induced by I/R and H/R, respectively. Further, this study revealed that galectin-3 interacted with bcl-2, instead of bax, in the cardiomyocyte, and regulated the phosphorylation of AKT, p70s6k, JNK, IκB and p65. Our findings demonstrated that galectin-3 could prevent myocardial I/R injury through interacting with bcl-2.
Collapse
Affiliation(s)
- Meiqi Zhang
- Department of Intensive Care Unit, Hangzhou Hospital of Traditional Chinese Medicine (Dingqiao District), Guangxing Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Kang Cheng
- Department of Intensive Care Unit, Hangzhou Hospital of Traditional Chinese Medicine (Dingqiao District), Guangxing Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Huan Chen
- Department of Emergency Medicine, Zhejiang Provincial People' s Hospital (People' s Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Jianfeng Tu
- Department of Emergency Medicine, Zhejiang Provincial People' s Hospital (People' s Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Ye Shen
- Department of Emergency Medicine, Zhejiang Provincial People' s Hospital (People' s Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Lingxiao Pang
- Department of Emergency Medicine, Zhejiang Provincial People' s Hospital (People' s Hospital of Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Weihua Wu
- Department of Intensive Care Unit, Hangzhou Hospital of Traditional Chinese Medicine (Dingqiao District), Guangxing Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
48
|
Abstract
Acetaminophen (APAP)-induced acute liver failure (ALF) is a life-threatening disease with only a few treatment options available. Though extensive research has been conducted for more than 40 years, the underlying pathomechanisms are not completely understood. Here, we studied as to whether APAP-induced ALF can be prevented in mice by silencing the BH3-interacting domain death agonist (Bid) as a potential key player in APAP pathology. For silencing Bid expression in mice, siRNABid was formulated with the liver-specific siRNA delivery system DBTC and administered 48 h prior to APAP exposure. Mice which were pre-treated with HEPES (vehicleHEPES) and siRNALuci served as siRNA controls. Hepatic pathology was assessed by in vivo fluorescence microscopy, molecular biology, histology and laboratory analysis 6 h after APAP or PBS exposure. Application of siRNABid caused a significant decrease of mRNA and protein expression of Bid in APAP-exposed mice. Off-targets, such as cytochrome P450 2E1 and glutathione, which are known to be consumed under APAP intoxication, were comparably reduced in all APAP-exposed mice, underlining the specificity of Bid silencing. In APAP-exposed mice non-sterile inflammation with leukocyte infiltration and perfusion failure remained almost unaffected by Bid silencing. However, the Bid silencing reduced hepatocellular damage, evident by a remarkable decrease of DNA fragmented cells in APAP-exposed mice. In these mice, the expression of the pro-apoptotic protein Bax, which recently gained importance in the cell death pathway of regulated necrosis, was also significantly reduced, in line with a decrease in both, necrotic liver tissue and plasma transaminase activities. In addition, plasma levels of HMGB1, a marker of sterile inflammation, were significantly diminished. In conclusion, the liver-specific silencing of Bid expression did not protect APAP-exposed mice from microcirculatory dysfunction, but markedly protected the liver from necrotic cell death and in consequence from sterile inflammation. The study contributes to the understanding of the molecular mechanism of the APAP-induced pathogenic pathway by strengthening the importance of Bid and Bid silencing associated effects.
Collapse
|
49
|
Teoh SQ, Yap MKK. Naja sumatrana venom cytotoxin, sumaCTX exhibits concentration-dependent cytotoxicity via caspase-activated mitochondrial-mediated apoptosis without transitioning to necrosis. TOXIN REV 2020. [DOI: 10.1080/15569543.2020.1799408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Shun Qi Teoh
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Michelle Khai Khun Yap
- School of Science, Monash University Malaysia, Bandar Sunway, Malaysia
- Tropical Medicine and Biology Platform, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
50
|
Ramachandran A, Jaeschke H. A mitochondrial journey through acetaminophen hepatotoxicity. Food Chem Toxicol 2020; 140:111282. [PMID: 32209353 PMCID: PMC7254872 DOI: 10.1016/j.fct.2020.111282] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 12/11/2022]
Abstract
Acetaminophen (APAP) overdose is the leading cause of acute liver failure in the United States and APAP-induced hepatotoxicity is initiated by formation of a reactive metabolite which depletes hepatic glutathione and forms protein adducts. Studies over the years have established the critical role of c-Jun N terminal kinase (JNK) and its mitochondrial translocation, as well as mitochondrial oxidant stress and subsequent induction of the mitochondrial permeability transition in APAP pathophysiology. However, it is now evident that mitochondrial responses to APAP overdose are more nuanced than appreciated earlier, with multiple levels of control, for example, to dose of APAP. In addition, mitochondrial dynamics, as well as the organelle's importance in recovery and regeneration after APAP-induced liver injury is also being recognized, which are exciting new areas with significant therapeutic potential. Thus, this review examines the temporal course of hepatocyte mitochondrial responses to an APAP overdose with an emphasis on mechanistic response to various trigger checkpoints such as NAPQI-mitochondrial protein adduct formation and activated JNK translocation. Mitochondrial dynamics, the organelle's role in recovery after APAP and emerging areas of research which promise to provide further insight into modulation of APAP pathophysiology by these fascinating organelles will also be discussed.
Collapse
Affiliation(s)
- Anup Ramachandran
- Department of Pharmacology, Toxicology, and Therapeutic, University of Kansas Medical Center, Kansas City, KS, USA.
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology, and Therapeutic, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|