1
|
Chia WT, Chen KY, Yang CY, Hsieh CC, Tsao CH, Lin CK, Peng B, Ho SL, Chen YL, Chang SC, Chen YW. Okanin Inhibits Cell Growth and Induces Apoptosis and Pyroptosis in Oral Cancer. Cancers (Basel) 2024; 16:3195. [PMID: 39335166 PMCID: PMC11429813 DOI: 10.3390/cancers16183195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Okanin, a flavonoid compound derived from Bidens pilosa L., has garnered attention for its anti-inflammatory properties. Although Bidens pilosa is commonly used in healthcare products and functional foods, the anticancer potential of okanin, particularly in oral cancer, remains underexplored. This study aims to investigate the effects of okanin on oral cancer cell lines and its potential as a therapeutic agent. METHODS The study involved assessing the cytotoxic effects of okanin on oral cancer cell lines SAS, SCC25, HSC3, and OEC-M1. The IC50 values were determined using methylene blue assays, and the clonogenic capacity was evaluated through colony formation assays. Flow cytometry was used to analyze cell cycle progression and apoptosis. Caspase-3/7 activity assays and annexin V/7-AAD staining confirmed the induction of apoptosis and pyroptosis. In vivo efficacy was assessed using a SAS xenograft model, and immunohistochemical analysis of xenograft tissue was performed to examine pyroptosis-related markers. RESULTS Okanin exhibited potent cytotoxic effects with IC50 values of 12.0 ± 0.8, 58.9 ± 18.7, 18.1 ± 5.3, and 43.2 ± 6.2 μM in SAS, SCC25, HSC3, and OEC-M1 cells, respectively. It caused dose- and time-dependent reductions in cell viability and significantly impaired clonogenic capacity. Flow cytometry revealed G2/M cell cycle arrest and increased sub-G1 population, indicating cell cycle disruption and death. Okanin induced both apoptosis and pyroptosis, as confirmed by caspase-3/7 activity and annexin V/7-AAD staining. In vivo, okanin reduced tumor growth and involved pyroptosis-related markers such as CASP1, GSDMC, GSDMD, and GSDME. CONCLUSIONS Okanin demonstrates significant anticancer potential, particularly in oral cancer, by inducing both apoptosis and pyroptosis. Its efficacy in reducing tumor growth in vivo further supports its potential as a novel therapeutic option. Further mechanistic studies are needed to elucidate the pathways involved in okanin-mediated cell death and to explore its clinical applications.
Collapse
Grants
- TSGH-C01-109017 Tri-Service General Hospital, Taiwan, Republic of China
- TSGH-C05-110035 Tri-Service General Hospital, Taiwan, Republic of China
- TSGH-C04-111037 Tri-Service General Hospital, Taiwan, Republic of China
- TSGH-D-109160 Tri-Service General Hospital, Taiwan, Republic of China
- TSGH-D-110148 Tri-Service General Hospital, Taiwan, Republic of China
- TSGH-D-110149 Tri-Service General Hospital, Taiwan, Republic of China
- TSGH-D-110151 Tri-Service General Hospital, Taiwan, Republic of China
- TSGH-D-110152 Tri-Service General Hospital, Taiwan, Republic of China
- TSGH-D-112148 Tri-Service General Hospital, Taiwan, Republic of China
- TSGH-D-113066 Tri-Service General Hospital, Taiwan, Republic of China
- TSGH-D-110154 Tri-Service General Hospital, Taiwan, Republic of China
- TSGH-C03-113040 Tri-Service General Hospital, Taiwan, Republic of China
- TSGH-C02-112032 Tri-Service General Hospital, Taiwan, Republic of China
- MAB-E-109003 Ministry of National Defense, Taiwan, Republic of China
- MAB-D-110003 Ministry of National Defense, Taiwan, Republic of China
- MND-MAB-110-043 Ministry of National Defense, Taiwan, Republic of China
- MND-MAB-110-076 Ministry of National Defense, Taiwan, Republic of China
- MND-MAB-C-111036 Ministry of National Defense, Taiwan, Republic of China
- MAB-E-111002 Ministry of National Defense, Taiwan, Republic of China
- MND-MAB-D-111149 Ministry of National Defense, Taiwan, Republic of China
- MND-MAB-D-112176 Ministry of National Defense, Taiwan, Republic of China
- MND-MAB-C08-112033 Ministry of National Defense, Taiwan, Republic of China
- MND-MAB-D-113117 Ministry of National Defense, Taiwan, Republic of China
- KAFGH_E_111047 Kaohsiung Armed Forces General Hospital, Taiwan, Republic of China
- KAFGH_E_112061 Kaohsiung Armed Forces General Hospital, Taiwan, Republic of China
- KAFGH_E_113058 Kaohsiung Armed Forces General Hospital, Taiwan, Republic of China
- HAFGH_E_112018 Hualien Armed Forces General Hospital, Taiwan, Republic of China
- KSVGH112-135 Kaohsiung Veterans General Hospital, Taiwan, Republic of China
Collapse
Affiliation(s)
- Wei-Tso Chia
- Department of Orthopedics, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 302, Taiwan
- Department of Nursing, Yuan Pie University of Medical Technology, Hsinchu 302, Taiwan
- Tri-Service General Hospital, Taipei 114, Taiwan
| | - Kuei-Yuan Chen
- School of Dentistry, National Defense Medical Center, Taipei 114, Taiwan
- Department of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei 114, Taiwan
| | - Cheng-Yu Yang
- School of Dentistry, National Defense Medical Center, Taipei 114, Taiwan
- Department of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei 114, Taiwan
| | - Cheng-Chih Hsieh
- Department of Pharmacy, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
- School of Pharmacy and Institute of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan
| | - Chang-Huei Tsao
- Department of Microbiology and Immunology, National Defense Medical Center, Taipei 114, Taiwan
- Department of Medical Research, Tri-Service General Hospital, Taipei 114, Taiwan
| | - Chih-Kung Lin
- Division of Anatomic Pathology, Taipei Tzu Chi Hospital, New Taipei City 231, Taiwan
| | - Bo Peng
- School of Dentistry, National Defense Medical Center, Taipei 114, Taiwan
- Department of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei 114, Taiwan
| | - Sien-Lin Ho
- School of Dentistry, National Defense Medical Center, Taipei 114, Taiwan
- Department of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei 114, Taiwan
| | - Yi-Ling Chen
- Department of Dentistry, Kaohsiung Armed Forces General Hospital, Kaohsiung 813, Taiwan
| | - Szu-Chien Chang
- Department of Dentistry, Kaohsiung Armed Forces General Hospital, Kaohsiung 813, Taiwan
| | - Yuan-Wu Chen
- School of Dentistry, National Defense Medical Center, Taipei 114, Taiwan
- Department of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei 114, Taiwan
| |
Collapse
|
2
|
Conejo-García A, Jiménez-Martínez Y, Cámara R, Franco-Montalbán F, Peña-Martín J, Boulaiz H, Carrión MD. New substituted benzoxazine derivatives as potent inducers of membrane permeability and cell death. Bioorg Med Chem 2024; 111:117849. [PMID: 39068873 DOI: 10.1016/j.bmc.2024.117849] [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: 06/20/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
The search for new agents targeting different forms of cell death is an important research focus for developing new and potent antitumor therapies. As a contribution to this endeavor, we have designed and synthesized a series of new substituted 3,4-dihydro-2H-1,4-benzoxazine derivatives. These compounds have been evaluated for their efficacy against MCF-7 breast cancer and HCT-116 colon cancer cell lines. Overall, substituting this heterocycle led to improved antiproliferative activity compared to the unsubstituted derivative 1. The most active compounds, 2b and 4b, showed IC50 values of 2.27 and 3.26 μM against MCF-7 cells and 4.44 and 7.63 μM against HCT-116 cells, respectively. To investigate the mechanism of action of the target compounds, the inhibition profile of 8 kinases involved in cell signaling was studied highlighting residual activity on HER2 and JNK1 kinases. 2b and 4b showed a consistent binding mode to both receptor kinases, establishing significant interactions with known and catalytically important domains and residues. Compounds 2b and 4b exhibit potent cytotoxic activity by disrupting cell membrane permeability, likely triggering both inflammatory and non-inflammatory cell death mechanisms. This dual capability increases their versatility in the treatment of different stages or types of tumors, providing greater flexibility in clinical applications.
Collapse
Affiliation(s)
- Ana Conejo-García
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, Faculty of Pharmacy, Campus Cartuja s/n, University of Granada, 18071 Granada, Spain; Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Avenida de Madrid, 15, 18012 Granada, Spain
| | - Yaiza Jiménez-Martínez
- Department of Anatomy and Human Embryology, Faculty of Medicine, Avenida de la Investigación 11, University of Granada, 18016 Granada, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Avenida del Conocimiento s/n, 18016 Granada, Spain
| | - Rubén Cámara
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, Faculty of Pharmacy, Campus Cartuja s/n, University of Granada, 18071 Granada, Spain
| | - Francisco Franco-Montalbán
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, Faculty of Pharmacy, Campus Cartuja s/n, University of Granada, 18071 Granada, Spain
| | - Jesús Peña-Martín
- Department of Anatomy and Human Embryology, Faculty of Medicine, Avenida de la Investigación 11, University of Granada, 18016 Granada, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Avenida del Conocimiento s/n, 18016 Granada, Spain
| | - Houria Boulaiz
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Avenida de Madrid, 15, 18012 Granada, Spain; Department of Anatomy and Human Embryology, Faculty of Medicine, Avenida de la Investigación 11, University of Granada, 18016 Granada, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Avenida del Conocimiento s/n, 18016 Granada, Spain.
| | - M Dora Carrión
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Biomedicine and the Environment, Faculty of Pharmacy, Campus Cartuja s/n, University of Granada, 18071 Granada, Spain; Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, Avenida de Madrid, 15, 18012 Granada, Spain.
| |
Collapse
|
3
|
Ali W, Kulsoom, Wang F. Molecular probes for monitoring pyroptosis: design, imaging and theranostic application. Apoptosis 2024; 29:1038-1050. [PMID: 38772991 DOI: 10.1007/s10495-024-01980-3] [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] [Accepted: 05/13/2024] [Indexed: 05/23/2024]
Abstract
Pyroptosis is a recently discovered process of programmed cell death that is linked with tumor progression and potential treatment strategies. Unlike other forms of programmed cell death, such as apoptosis or necrosis, pyroptosis is associated with pore-forming proteins gasdermin D (GSDMD), which are cleaved by caspase enzymes to form oligomers. These oligomers are then inserted into the cell surface membrane, causing pores to consequently result in rapid cell death. Pyroptosis, in conjunction with immunotherapy, represents a promising avenue for prognostication and antitumor therapy, providing a more precise direction for disease treatment. To gain deeper insight into the mechanisms underlying pyroptosis in real-time, non-invasive and live cell imaging techniques are urgently needed. Non-invasive imaging techniques can enhance future diagnostic and therapeutic approaches for inflammatory diseases, including different types of tumors. This review article discusses various non-invasive molecular probes for detecting pyroptosis, including genetic reporters and nanomaterials. These strategies can enhance scientists' understanding of pyroptosis and help discover personalized and effective ways to treat inflammatory diseases, particularly tumors.
Collapse
Affiliation(s)
- Wajahat Ali
- Department of Medical Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Kulsoom
- Department of Biochemistry, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Fu Wang
- Department of Medical Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China.
- Xianyang Key Laboratory of Molecular Imaging and Drug Synthesis, School of Pharmacy, Shaanxi University of International Trade & Commerce, Xianyang, 712046, Shaanxi, China.
| |
Collapse
|
4
|
Yang H, Xiao L, Wu D, Zhang T, Ge P. O-GlcNAcylation of NLRP3 Contributes to Lipopolysaccharide-Induced Pyroptosis of Human Gingival Fibroblasts. Mol Biotechnol 2024; 66:2023-2031. [PMID: 37566188 DOI: 10.1007/s12033-023-00846-4] [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: 04/08/2023] [Accepted: 07/29/2023] [Indexed: 08/12/2023]
Abstract
Periodontitis is a leading chronic oral disorder and poses a serious burden on public health. O-GlcNAc glycosylation (O-GlcNAcylation) is regulated only by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA) and participates in the regulation of human gingival fibroblasts (HGFs) function. Hence, the purpose of this study is to investigate whether HGFs cell function and periodontitis pathogenesis are regulated by O-GlcNAcylation. Herein, we first established cell model of periodontitis induced by lipopolysaccharide (LPS). The cell viability was measured with CCK-8 assay. Pyroptosis was measured by flow cytometry and western blot. The inflammatory factors levels were detected with ELISA kits. Afterward, our findings indicated that LPS elevated the O-GlcNAcylation level of HGFs and inhibition of O-GlcNAcylation improved LPS-induced pyroptosis of HGFs. Mechanistically, LPS heightened the expression of OGT to induce the O-GlcNAcylation of NLRP3. Subsequently, we certified that Thr542 was the O-GlcNAcylation site of NLRP3. More importantly, upregulation of NLRP3 reversed the effects of OGT knockdown on LPS-induced pyroptosis. In general, the current research demonstrated that LPS contributed to the pyroptosis of HGFs by enhancing the OGT expression to promote O-GlcNAcylation of NLRP3, which suggested that O-GlcNAcylation of NLRP3 was a driving factor for periodontitis and offered a novel insight into the treatment of this disease.
Collapse
Affiliation(s)
- Hao Yang
- Department of Stomatology, First Branch Hospital of First Affilliated Hospital, Chongqing Medical University, No. 24, Shiyou Road, Yuzhong District, Chongqing, 400011, China
| | - Li Xiao
- Department of Stomatology, First Branch Hospital of First Affilliated Hospital, Chongqing Medical University, No. 24, Shiyou Road, Yuzhong District, Chongqing, 400011, China
| | - Dongxue Wu
- Department of Stomatology, First Branch Hospital of First Affilliated Hospital, Chongqing Medical University, No. 24, Shiyou Road, Yuzhong District, Chongqing, 400011, China
| | - Tingting Zhang
- Department of Stomatology, First Branch Hospital of First Affilliated Hospital, Chongqing Medical University, No. 24, Shiyou Road, Yuzhong District, Chongqing, 400011, China
| | - Ping Ge
- Department of Internal Medicine-Cardiovascular, First Branch Hospital of First Affilliated Hospital, Chongqing Medical University, No. 24, Shiyou Road, Yuzhong District, Chongqing, 400011, China.
| |
Collapse
|
5
|
Vinţeler N, Feurdean CN, Petkes R, Barabas R, Boşca BA, Muntean A, Feștilă D, Ilea A. Biomaterials Functionalized with Inflammasome Inhibitors-Premises and Perspectives. J Funct Biomater 2024; 15:32. [PMID: 38391885 PMCID: PMC10889089 DOI: 10.3390/jfb15020032] [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: 12/30/2023] [Revised: 01/21/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
This review aimed at searching literature for data regarding the inflammasomes' involvement in the pathogenesis of oral diseases (mainly periodontitis) and general pathologies, including approaches to control inflammasome-related pathogenic mechanisms. The inflammasomes are part of the innate immune response that activates inflammatory caspases by canonical and noncanonical pathways, to control the activity of Gasdermin D. Once an inflammasome is activated, pro-inflammatory cytokines, such as interleukins, are released. Thus, inflammasomes are involved in inflammatory, autoimmune and autoinflammatory diseases. The review also investigated novel therapies based on the use of phytochemicals and pharmaceutical substances for inhibiting inflammasome activity. Pharmaceutical substances can control the inflammasomes by three mechanisms: inhibiting the intracellular signaling pathways (Allopurinol and SS-31), blocking inflammasome components (VX-765, Emricasan and VX-740), and inhibiting cytokines mediated by the inflammasomes (Canakinumab, Anakinra and Rilonacept). Moreover, phytochemicals inhibit the inflammasomes by neutralizing reactive oxygen species. Biomaterials functionalized by the adsorption of therapeutic agents onto different nanomaterials could represent future research directions to facilitate multimodal and sequential treatment in oral pathologies.
Collapse
Affiliation(s)
- Norina Vinţeler
- Department of Oral Rehabilitation, Faculty of Dentistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Claudia Nicoleta Feurdean
- Department of Oral Rehabilitation, Faculty of Dentistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Regina Petkes
- Department of Chemistry and Chemical Engineering of Hungarian Line of Study, Faculty of Chemistry and Chemical Engineering, Babeș-Bolyai University, 400028 Cluj-Napoca, Romania
| | - Reka Barabas
- Department of Chemistry and Chemical Engineering of Hungarian Line of Study, Faculty of Chemistry and Chemical Engineering, Babeș-Bolyai University, 400028 Cluj-Napoca, Romania
| | - Bianca Adina Boşca
- Department of Histology, Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Alexandrina Muntean
- Department of Paediatric, Faculty of Dentistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca 400012, Romania
| | - Dana Feștilă
- Department of Orthodontics, Faculty of Dentistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca 400012, Romania
| | - Aranka Ilea
- Department of Oral Rehabilitation, Faculty of Dentistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
6
|
Wang Y, Jin M, Cheng CK, Li Q. Tubular injury in diabetic kidney disease: molecular mechanisms and potential therapeutic perspectives. Front Endocrinol (Lausanne) 2023; 14:1238927. [PMID: 37600689 PMCID: PMC10433744 DOI: 10.3389/fendo.2023.1238927] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Diabetic kidney disease (DKD) is a chronic complication of diabetes and the leading cause of end-stage renal disease (ESRD) worldwide. Currently, there are limited therapeutic drugs available for DKD. While previous research has primarily focused on glomerular injury, recent studies have increasingly emphasized the role of renal tubular injury in the pathogenesis of DKD. Various factors, including hyperglycemia, lipid accumulation, oxidative stress, hypoxia, RAAS, ER stress, inflammation, EMT and programmed cell death, have been shown to induce renal tubular injury and contribute to the progression of DKD. Additionally, traditional hypoglycemic drugs, anti-inflammation therapies, anti-senescence therapies, mineralocorticoid receptor antagonists, and stem cell therapies have demonstrated their potential to alleviate renal tubular injury in DKD. This review will provide insights into the latest research on the mechanisms and treatments of renal tubular injury in DKD.
Collapse
Affiliation(s)
- Yu Wang
- Department of Endocrinology and Metabolism, Shenzhen University General Hospital, Shenzhen, Guangdong, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Mingyue Jin
- Department of Endocrinology and Metabolism, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Chak Kwong Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qiang Li
- Department of Endocrinology and Metabolism, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Dubyak GR, Miller BA, Pearlman E. Pyroptosis in neutrophils: Multimodal integration of inflammasome and regulated cell death signaling pathways. Immunol Rev 2023; 314:229-249. [PMID: 36656082 PMCID: PMC10407921 DOI: 10.1111/imr.13186] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Pyroptosis is a proinflammatory mode of lytic cell death mediated by accumulation of plasma membrane (PM) macropores composed of gasdermin-family (GSDM) proteins. It facilitates two major functions in innate immunity: (i) elimination of intracellular replicative niches for pathogenic bacteria; and (ii) non-classical secretion of IL-1 family cytokines that amplify host-beneficial inflammatory responses to microbial infection or tissue damage. Physiological roles for gasdermin D (GSDMD) in pyroptosis and IL-1β release during inflammasome signaling have been extensively characterized in macrophages. This involves cleavage of GSDMD by caspase-1 to generate GSDMD macropores that mediate IL-1β efflux and progression to pyroptotic lysis. Neutrophils, which rapidly accumulate in large numbers at sites of tissue infection or damage, become the predominant local source of IL-1β in coordination with their potent microbiocidal capacity. Similar to macrophages, neutrophils express GSDMD and utilize the same spectrum of diverse inflammasome platforms for caspase-1-mediated cleavage of GSDMD. Distinct from macrophages, neutrophils possess a remarkable capacity to resist progression to GSDMD-dependent pyroptotic lysis to preserve their viability for efficient microbial killing while maintaining GSDMD-dependent mechanisms for export of bioactive IL-1β. Rather, neutrophils employ cell-specific mechanisms to conditionally engage GSDMD-mediated pyroptosis in response to bacterial pathogens that use neutrophils as replicative niches. GSDMD and pyroptosis have also been mechanistically linked to induction of NETosis, a signature neutrophil pathway that expels decondensed nuclear DNA into extracellular compartments for immobilization and killing of microbial pathogens. This review summarizes a rapidly growing number of recent studies that have produced new insights, unexpected mechanistic nuances, and some controversies regarding the regulation of, and roles for, neutrophil inflammasomes, pyroptosis, and GSDMs in diverse innate immune responses.
Collapse
Affiliation(s)
- George R. Dubyak
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Brandon A. Miller
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Eric Pearlman
- Department of Ophthalmology, University of California, Irvine, California, USA
- Department of Physiology and Biophysics, University of California, Irvine, California, USA
| |
Collapse
|
8
|
Cai Y, Zhou Y, Li Z, Xia P, ChenFu X, Shi A, Zhang J, Yu P. Non-coding RNAs in necroptosis, pyroptosis, and ferroptosis in cardiovascular diseases. Front Cardiovasc Med 2022; 9:909716. [PMID: 35990979 PMCID: PMC9386081 DOI: 10.3389/fcvm.2022.909716] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/04/2022] [Indexed: 11/18/2022] Open
Abstract
Accumulating evidence has proved that non-coding RNAs (ncRNAs) play a critical role in the genetic programming and gene regulation of cardiovascular diseases (CVDs). Cardiovascular disease morbidity and mortality are rising and have become a primary public health issue that requires immediate resolution through effective intervention. Numerous studies have revealed that new types of cell death, such as pyroptosis, necroptosis, and ferroptosis, play critical cellular roles in CVD progression. It is worth noting that ncRNAs are critical novel regulators of cardiovascular risk factors and cell functions by mediating pyroptosis, necroptosis, and ferroptosis. Thus, ncRNAs can be regarded as promising therapeutic targets for treating and diagnosing cardiovascular diseases. Recently, there has been a surge of interest in the mediation of ncRNAs on three types of cell death in regulating tissue homeostasis and pathophysiological conditions in CVDs. Although our understanding of ncRNAs remains in its infancy, the studies reviewed here may provide important new insights into how ncRNAs interact with CVDs. This review summarizes what is known about the functions of ncRNAs in modulating cell death-associated CVDs and their role in CVDs, as well as their current limitations and future prospects.
Collapse
Affiliation(s)
- Yuxi Cai
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yiwen Zhou
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhangwang Li
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Panpan Xia
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Metabolism and Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of National Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Xinxi ChenFu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Metabolism and Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- Branch of National Clinical Research Center for Metabolic Diseases, Nanchang, China
| | - Ao Shi
- School of Medicine, University of Nicosia, Nicosia, Cyprus
- School of Medicine, St. George University of London, London, United Kingdom
| | - Jing Zhang
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jing Zhang
| | - Peng Yu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Metabolism and Endocrinology, the Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, China
- *Correspondence: Peng Yu
| |
Collapse
|
9
|
Kari S, Subramanian K, Altomonte IA, Murugesan A, Yli-Harja O, Kandhavelu M. Programmed cell death detection methods: a systematic review and a categorical comparison. Apoptosis 2022; 27:482-508. [PMID: 35713779 PMCID: PMC9308588 DOI: 10.1007/s10495-022-01735-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2022] [Indexed: 01/15/2023]
Abstract
Programmed cell death is considered a key player in a variety of cellular processes that helps to regulate tissue growth, embryogenesis, cell turnover, immune response, and other biological processes. Among different types of cell death, apoptosis has been studied widely, especially in the field of cancer research to understand and analyse cellular mechanisms, and signaling pathways that control cell cycle arrest. Hallmarks of different types of cell death have been identified by following the patterns and events through microscopy. Identified biomarkers have also supported drug development to induce cell death in cancerous cells. There are various serological and microscopic techniques with advantages and limitations, that are available and are being utilized to detect and study the mechanism of cell death. The complexity of the mechanism and difficulties in distinguishing among different types of programmed cell death make it challenging to carry out the interventions and delay its progression. In this review, mechanisms of different forms of programmed cell death along with their conventional and unconventional methods of detection of have been critically reviewed systematically and categorized on the basis of morphological hallmarks and biomarkers to understand the principle, mechanism, application, advantages and disadvantages of each method. Furthermore, a very comprehensive comparative analysis has been drawn to highlight the most efficient and effective methods of detection of programmed cell death, helping researchers to make a reliable and prudent selection among the available methods of cell death assay. Conclusively, how programmed cell death detection methods can be improved and can provide information about distinctive stages of cell death detection have been discussed.
Collapse
Affiliation(s)
- Sana Kari
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101, Tampere, Finland
| | - Kumar Subramanian
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101, Tampere, Finland
| | - Ilenia Agata Altomonte
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101, Tampere, Finland
| | - Akshaya Murugesan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101, Tampere, Finland.,Department of Biotechnology, Lady Doak College, Thallakulam, Madurai, 625002, India
| | - Olli Yli-Harja
- Institute for Systems Biology, 1441N 34th Street, Seattle, WA, USA.,Computational Systems Biology Group, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101, Tampere, Finland
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101, Tampere, Finland. .,Department of Biotechnology, Lady Doak College, Thallakulam, Madurai, 625002, India.
| |
Collapse
|
10
|
Pyroptosis-Mediated Periodontal Disease. Int J Mol Sci 2021; 23:ijms23010372. [PMID: 35008798 PMCID: PMC8745163 DOI: 10.3390/ijms23010372] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 12/17/2022] Open
Abstract
Pyroptosis is a caspase-dependent process relevant to the understanding of beneficial host responses and medical conditions for which inflammation is central to the pathophysiology of the disease. Pyroptosis has been recently suggested as one of the pathways of exacerbated inflammation of periodontal tissues. Hence, this focused review aims to discuss pyroptosis as a pathological mechanism in the cause of periodontitis. The included articles presented similarities regarding methods, type of cells applied, and cell stimulation, as the outcomes also point to the same direction considering the cellular events. The collected data indicate that virulence factors present in the diseased periodontal tissues initiate the inflammasome route of tissue destruction with caspase activation, cleavage of gasdermin D, and secretion of interleukins IL-1β and IL-18. Consequently, removing periopathogens’ virulence factors that trigger pyroptosis is a potential strategy to combat periodontal disease and regain tissue homeostasis.
Collapse
|