1
|
Jung YY, Hong Y, Nam D, Deivasigamani A, Narula AS, Chinnathambi A, Namjoshi OA, Blough BE, Alharbi SA, Hui KM, Sethi G, Ahn KS. TMP: A dual modulator of apoptosis and autophagy via SHP-1 regulation in hepatocellular carcinoma. Life Sci 2025; 361:123316. [PMID: 39675549 DOI: 10.1016/j.lfs.2024.123316] [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: 09/03/2024] [Revised: 12/01/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) poses a significant health burden due to its high incidence, and current treatment effectiveness is hindered by drug resistance. Thus, investigation of novel therapeutic approaches derived from natural sources is crucial for improving patient outcomes. AIMS This study aimed to explore the potential of Tetramethylpyrazine (TMP), bioactive alkaloid (ligustrazine) isolated from Chuanxiong (Ligusticum Wallichii), in targeting HCC by inducing apoptosis and enhancing autophagy. The study focused on elucidating the molecular mechanisms underlying anti-cancer effects of TMP. MAIN METHODS To determine the influence of TMP on apoptosis and autophagy, Western blot analysis, annexin V assay, cell cycle analysis, acridine orange staining, and immunocytochemistry were performed. Next, the activation of the STAT3 signaling pathway and the anti-cancer effects of TMP in vivo were examined in an orthotopic HCCLM3-Lu mouse model. KEY FINDINGS TMP treatment induced apoptosis in HCCLM3 and Hep3B cells by activating key apoptotic factors while inhibiting proteins associated with cell survival and angiogenesis. Additionally, TMP enhanced autophagy by promoting the formation of autophagosomes and stimulating autophagy-related proteins. Furthermore, TMP suppressed the activation of the STAT3 signaling pathway by upregulating SHP-1, thereby inhibiting tumorigenesis and activating cell death pathways. Additionally, our in vivo research demonstrated that TMP significantly inhibited tumor growth and triggered the activation of both apoptosis and autophagy in tumor tissues. SIGNIFICANCE Our findings of this study demonstrate that TMP exerts a dual-action mechanism by modulating both apoptosis and autophagy, thus offering a promising strategy to overcome drug resistance in HCC.
Collapse
Affiliation(s)
- Young Yun Jung
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdae-mun-gu, Seoul 02447, Republic of Korea
| | - Yejin Hong
- Department of Acupuncture and Moxibustion, Kyung Hee University Korean Medicine Hospital, Seoul, Republic of Korea
| | - Dongwoo Nam
- Department of Acupuncture and Moxibustion, Kyung Hee University Korean Medicine Hospital, Seoul, Republic of Korea; Department of Acupuncture and Moxibustion, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Amudha Deivasigamani
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore
| | | | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Ojas A Namjoshi
- Engine Biosciences, 733 Industrial Rd, San Carlos, CA 94070, USA
| | - Bruce E Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, NC 27616, USA
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Kam Man Hui
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdae-mun-gu, Seoul 02447, Republic of Korea.
| |
Collapse
|
2
|
Cao Z, Tian K, Ran Y, Zhou H, Zhou L, Ding Y, Tang X. Beclin-1: a therapeutic target at the intersection of autophagy, immunotherapy, and cancer treatment. Front Immunol 2024; 15:1506426. [PMID: 39650649 PMCID: PMC11621085 DOI: 10.3389/fimmu.2024.1506426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/01/2024] [Indexed: 12/11/2024] Open
Abstract
The significant identification of Beclin-1's function in regulating autophagy flow signified a significant progression in our understanding of cellular operations. Beclin-1 acts as a scaffold for forming the PI3KC3 complex, controlling autophagy and cellular trafficking processes in a complicated way. This intricate protein has garnered considerable attention due to its substantial impact on the development of tumors. Strong evidence indicates Beclin-1 plays a critical role in controlling autophagy in various human cancer types and its intricate connection with apoptosis and ferroptosis. The potential of Beclin-1 as a viable target for cancer therapy is highlighted by its associations with key autophagy regulators such as AMPK, mTOR, and ATGs. Beclin-1 controls the growth and dissemination of tumors by autophagy. It also affects how tumors react to therapies such as chemotherapy and radiation therapy. The role of Beclin-1 in autophagy can influence apoptosis, depending on whether it supports cell survival or leads to cell death. Beclin-1 plays a crucial role in ferroptosis by increasing ATG5 levels, which in turn promotes autophagy-triggered ferroptosis. Finally, we analyzed the possible function of Beclin-1 in tumor immunology and drug sensitivity in cancers. In general, Beclin-1 has a significant impact on regulating autophagy, offering various potentials for medical intervention and altering our understanding of cancer biology.
Collapse
Affiliation(s)
- Zhumin Cao
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Ke Tian
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Yincheng Ran
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Haonan Zhou
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Lei Zhou
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Yana Ding
- Department of Hepatobiliary Surgery, District Traditional Chinese Medicine Hospital, Chongqing, China
| | - Xiaowei Tang
- Department of Hepatobiliary Surgery, District Traditional Chinese Medicine Hospital, Chongqing, China
| |
Collapse
|
3
|
Wang X, Li K, Song T, Xing S, Wang W, Fang Y. Advances in ferroptosis in head and neck cancer (Review). Biomed Rep 2024; 21:151. [PMID: 39247426 PMCID: PMC11375624 DOI: 10.3892/br.2024.1839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/06/2024] [Indexed: 09/10/2024] Open
Abstract
Ferroptosis is an iron-dependent form of cell death that was discovered in 2012. It encompasses the coordinated orchestration of three fundamental biological pathways: Iron homeostasis, glutathione regulation and lipid metabolism. Head and neck cancer (HNC) is a heterogeneous group of cancers occurring on the mucosal surfaces of the upper respiratory and digestive tracts. Head and neck squamous cell carcinoma is the most common type of HNC, accounting for >90% of HNC cases, and has high morbidity and mortality rates. Despite improvements in diagnosis and treatment, the 5-year survival rate hovers at a dismal 50-60%, with recurrence afflicting nearly 30% of patients, highlighting the inadequacies of currently available treatments. Of note, research exploring the nexus between ferroptosis and HNC remains scarce; however, the present review endeavors to synthesize current knowledge surrounding ferroptosis. The present review elaborated on the normal physiological role of ferroptosis and discussed its potential involvement in HNC pathogenesis. Therapeutic strategies and prognostic paradigms for HNC that target ferroptosis were also reviewed. This review aims to provide direction to catalyze future investigations into ferroptosis in HNC.
Collapse
Affiliation(s)
- Xinyi Wang
- Department of Dermatology, Fuyang People's Hospital Affiliated to Bengbu Medical University, Fuyang, Anhui 236000, P.R. China
| | - Kunpeng Li
- Department of Dermatology, Fuyang People's Hospital Affiliated to Bengbu Medical University, Fuyang, Anhui 236000, P.R. China
| | - Teng Song
- Department of Dermatology, Fuyang People's Hospital Affiliated to Bengbu Medical University, Fuyang, Anhui 236000, P.R. China
| | - Suliang Xing
- Department of Dermatology, Fuyang People's Hospital Affiliated to Bengbu Medical University, Fuyang, Anhui 236000, P.R. China
| | - Wei Wang
- Department of Dermatology, Fuyang People's Hospital Affiliated to Bengbu Medical University, Fuyang, Anhui 236000, P.R. China
| | - Yuhui Fang
- Department of Dermatology, Fuyang People's Hospital Affiliated to Bengbu Medical University, Fuyang, Anhui 236000, P.R. China
| |
Collapse
|
4
|
Resta SC, Guerra F, Talà A, Bucci C, Alifano P. Beyond Inflammation: Role of Pyroptosis Pathway Activation by Gram-Negative Bacteria and Their Outer Membrane Vesicles (OMVs) in the Interaction with the Host Cell. Cells 2024; 13:1758. [PMID: 39513865 PMCID: PMC11545737 DOI: 10.3390/cells13211758] [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: 09/27/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Pyroptosis is a gasdermin-mediated pro-inflammatory programmed cell death that, during microbial infections, aims to restrict the spreading of bacteria. Nevertheless, excessive pyroptosis activation leads to inflammation levels that are detrimental to the host. Pathogen-associated molecular patterns (PAMPs) present in bacteria and outer membrane vesicles (OMVs) can trigger pyroptosis pathways in different cell types with different outcomes. Moreover, some pathogens have evolved virulence factors that directly interfere with pyroptosis pathways, like Yersinia pestis YopM and Shigella flexneri IpaH7.8. Other virulence factors, such as those of Neisseria meningitidis, Neisseria gonorrhoeae, Salmonella enterica, and Helicobacter pylori affect pyroptosis pathways indirectly with important differences between pathogenic and commensal species of the same family. These pathogens deserve special attention because of the increasing antimicrobial resistance of S. flexneri and N. gonorrhoeae, the high prevalence of S. enterica and H. pylori, and the life-threatening diseases caused by N. meningitidis and Y. pestis. While inflammation due to macrophage pyroptosis has been extensively addressed, the effects of activation of pyroptosis pathways on modulation of cell cytoskeleton and cell-cell junctions in epithelia and endothelia and on the bacterial crossing of epithelial and endothelial barriers have only been partly investigated. Another important point is the diverse consequences of pyroptosis pathways on calcium influx, like activation of calcium-dependent enzymes and mitochondria dysregulation. This review will discuss the pyroptotic pathways activated by Gram-negative bacteria and their OMVs, analyzing the differences between pathogens and commensal bacteria. Particular attention will also be paid to the experimental models adopted and the main results obtained in the different models. Finally, strategies adopted by pathogens to modulate these pathways will be discussed with a perspective on the use of pyroptosis inhibitors as adjuvants in the treatment of infections.
Collapse
Affiliation(s)
- Silvia Caterina Resta
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy; (S.C.R.); (F.G.); (A.T.)
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy; (S.C.R.); (F.G.); (A.T.)
| | - Adelfia Talà
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy; (S.C.R.); (F.G.); (A.T.)
| | - Cecilia Bucci
- Department of Experimental Medicine (DiMeS), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy;
| | - Pietro Alifano
- Department of Experimental Medicine (DiMeS), University of Salento, Via Provinciale Lecce-Monteroni 165, 73100 Lecce, Italy;
| |
Collapse
|
5
|
Mu Y, Zhang Z, Zhou H, Ma L, Wang DA. Applications of nanotechnology in remodeling the tumour microenvironment for glioblastoma treatment. Biomater Sci 2024; 12:4045-4064. [PMID: 38993162 DOI: 10.1039/d4bm00665h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
With the increasing research and deepening understanding of the glioblastoma (GBM) tumour microenvironment (TME), novel and more effective therapeutic strategies have been proposed. The GBM TME involves intricate interactions between tumour and non-tumour cells, promoting tumour progression. Key therapeutic goals for GBM treatment include improving the immunosuppressive microenvironment, enhancing the cytotoxicity of immune cells against tumours, and inhibiting tumour growth and proliferation. Consequently, remodeling the GBM TME using nanotechnology has emerged as a promising approach. Nanoparticle-based drug delivery enables targeted delivery, thereby improving treatment specificity, facilitating combination therapies, and optimizing drug metabolism. This review provides an overview of the GBM TME and discusses the methods of remodeling the GBM TME using nanotechnology. Specifically, it explores the application of nanotechnology in ameliorating immune cell immunosuppression, inducing immunogenic cell death, stimulating, and recruiting immune cells, regulating tumour metabolism, and modulating the crosstalk between tumours and other cells.
Collapse
Affiliation(s)
- Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
| | - Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
| | - Liang Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China.
- Karolinska Institutet Ming Wai Lau Centre for Reparative Medicine, HKSTP, Sha Tin, Hong Kong SAR
- Centre for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, Hong Kong SAR 999077, China
| |
Collapse
|
6
|
Hernández-García D, García-Meléndrez C, Hernández-Martínez R, Collazo-Navarrete O, Covarrubias L. Macrophages allocate before apoptosis initiation and produce reactive oxygen species during interdigital phagocytosis. Biol Open 2024; 13:bio060492. [PMID: 39052046 PMCID: PMC11445842 DOI: 10.1242/bio.060492] [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: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 07/27/2024] Open
Abstract
During programmed cell death (PCD), it is commonly accepted that macrophages are recruited by apoptotic cells to complete cell degradation. Interdigital cell death, a classical model of PCD, contributes to digit individualization in limbs of mammals and other vertebrates. Here, we show that macrophages are present in interdigits before significant cell death occurs and remain after apoptosis inhibition. The typical interdigital phagocytic activity was not observed after a partial depletion of macrophages and was markedly reduced by engulfment/phagosome maturation inhibition, as detected by its association with high lysosomal activity. β-galactosidase activity in this region was also coupled with phagocytosis, against its relationship with cellular senescence. Interdigital phagocytosis correlated with high levels of reactive oxygen species (ROS), common in embryo regions carrying abundant cell death, suggesting that macrophages are the major source of ROS. ROS generation was dependent on NADPH oxidases and blood vessel integrity, but not directly associated with lysosomal activity. Therefore, macrophages prepattern regions where abundant cell death is going to occur, and high lysosomal activity and the generation of ROS by an oxidative burst-like phenomenon are activities of phagocytosis.
Collapse
Affiliation(s)
- David Hernández-García
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., CP 62210, México
| | - Celina García-Meléndrez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., CP 62210, México
| | - Rocío Hernández-Martínez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., CP 62210, México
| | - Omar Collazo-Navarrete
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., CP 62210, México
| | - Luis Covarrubias
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mor., CP 62210, México
| |
Collapse
|
7
|
Hai E, Li B, Zhang J, Zhang J. Sperm freezing damage: the role of regulated cell death. Cell Death Discov 2024; 10:239. [PMID: 38762505 PMCID: PMC11102515 DOI: 10.1038/s41420-024-02013-3] [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: 01/16/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024] Open
Abstract
Substantial progress in research on sperm cryopreservation has occurred since the twentieth century, especially focusing on improving sperm freezing procedures and optimizing semen extenders. However, the cellular biological mechanisms of sperm freezing damage are still unclear, which greatly restricts the promotion and development of sperm cryopreservation. An essential component of sperm freezing damage is the occurrence of cell death. Considering the existence of multiple types of cell death pathways, this review discusses connections between characteristics of regulated cell death (e.g., apoptosis and ferroptosis), and accidental cell death (e.g., intracellular ice crystals) with sperm freezing damage and explores possible future research directions in this field.
Collapse
Affiliation(s)
- Erhan Hai
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, Inner Mongolia, China
| | - Boyuan Li
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, Inner Mongolia, China
| | - Jian Zhang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, Inner Mongolia, China
| | - Jiaxin Zhang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, Inner Mongolia, China.
| |
Collapse
|
8
|
Ai L, Yi N, Qiu C, Huang W, Zhang K, Hou Q, Jia L, Li H, Liu L. Revolutionizing breast cancer treatment: Harnessing the related mechanisms and drugs for regulated cell death (Review). Int J Oncol 2024; 64:46. [PMID: 38456493 PMCID: PMC11000534 DOI: 10.3892/ijo.2024.5634] [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: 09/30/2023] [Accepted: 02/22/2024] [Indexed: 03/09/2024] Open
Abstract
Breast cancer arises from the malignant transformation of mammary epithelial cells under the influence of various carcinogenic factors, leading to a gradual increase in its prevalence. This disease has become the leading cause of mortality among female malignancies, posing a significant threat to the health of women. The timely identification of breast cancer remains challenging, often resulting in diagnosis at the advanced stages of the disease. Conventional therapeutic approaches, such as surgical excision, chemotherapy and radiotherapy, exhibit limited efficacy in controlling the progression and metastasis of the disease. Regulated cell death (RCD), a process essential for physiological tissue cell renewal, occurs within the body independently of external influences. In the context of cancer, research on RCD primarily focuses on cuproptosis, ferroptosis and pyroptosis. Mounting evidence suggests a marked association between these specific forms of RCD, and the onset and progression of breast cancer. For example, a cuproptosis vector can effectively bind copper ions to induce cuproptosis in breast cancer cells, thereby hindering their proliferation. Additionally, the expression of ferroptosis‑related genes can enhance the sensitivity of breast cancer cells to chemotherapy. Likewise, pyroptosis‑related proteins not only participate in pyroptosis, but also regulate the tumor microenvironment, ultimately leading to the death of breast cancer cells. The present review discusses the unique regulatory mechanisms of cuproptosis, ferroptosis and pyroptosis in breast cancer, and the mechanisms through which they are affected by conventional cancer drugs. Furthermore, it provides a comprehensive overview of the significance of these forms of RCD in modulating the efficacy of chemotherapy and highlights their shared characteristics. This knowledge may provide novel avenues for both clinical interventions and fundamental research in the context of breast cancer.
Collapse
Affiliation(s)
- Leyu Ai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Department of Clinical Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Na Yi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Chunhan Qiu
- Department of Clinical Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Wanyi Huang
- Medical College, Yan'an University, Yan'an, Shaanxi 716000, P.R. China
| | - Keke Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Qiulian Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Long Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Hui Li
- Central Laboratory of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| | - Ling Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Urumqi, Xinjiang Uygur Autonomous Region 830017, P.R. China
| |
Collapse
|
9
|
Guo J, Li R, Ouyang Z, Tang J, Zhang W, Chen H, Zhu Q, Zhang J, Zhu G. Insights into the mechanism of transcription factors in Pb 2+-induced apoptosis. Toxicology 2024; 503:153760. [PMID: 38387706 DOI: 10.1016/j.tox.2024.153760] [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: 10/19/2023] [Revised: 02/12/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
The health risks associated with exposure to heavy metals, such as Pb2+, are increasingly concerning the public. Pb2+ can cause significant harm to the human body through oxidative stress, autophagy, inflammation, and DNA damage, disrupting cellular homeostasis and ultimately leading to cell death. Among these mechanisms, apoptosis is considered crucial. It has been confirmed that transcription factors play a central role as mediators during the apoptosis process. Interestingly, these transcription factors have different effects on apoptosis depending on the concentration and duration of Pb2+ exposure. In this article, we systematically summarize the significant roles of several transcription factors in Pb2+-induced apoptosis. This information provides insights into therapeutic strategies and prognostic biomarkers for diseases related to Pb2+ exposure.
Collapse
Affiliation(s)
- Jingchong Guo
- The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Ruikang Li
- The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Zhuqing Ouyang
- The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Jiawen Tang
- The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Wei Zhang
- Department of Anatomy, Medical College of Nanchang University, Nanchang 330006, China
| | - Hui Chen
- Department of Anatomy, Medical College of Nanchang University, Nanchang 330006, China
| | - Qian Zhu
- Department of Anatomy, Medical College of Nanchang University, Nanchang 330006, China
| | - Jing Zhang
- Department of Anatomy, Medical College of Nanchang University, Nanchang 330006, China.
| | - Gaochun Zhu
- Department of Anatomy, Medical College of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
10
|
Yarychkivska O, Sharmin R, Elkhalil A, Ghose P. Apoptosis and beyond: A new era for programmed cell death in Caenorhabditis elegans. Semin Cell Dev Biol 2024; 154:14-22. [PMID: 36792437 DOI: 10.1016/j.semcdb.2023.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 01/27/2023] [Accepted: 02/02/2023] [Indexed: 02/16/2023]
Abstract
Programmed cell death (PCD) is crucial for normal development and homeostasis. Our first insights into the genetic regulation of apoptotic cell death came from in vivo studies in the powerful genetic model system of C. elegans. More recently, novel developmental cell death programs occurring both embryonically and post-embryonically, and sex-specifically, have been elucidated. Recent studies in the apoptotic setting have also shed new light on the intricacies of phagocytosis in particular. This review provides a brief historical perspective of the origins of PCD studies in C. elegans, followed by a more detailed description of non-canonical apoptotic and non-apoptotic death programs. We conclude by posing open questions and commenting on our outlook on the future of PCD studies in C. elegans, highlighting the importance of advanced imaging tools and the continued leveraging of C. elegans genetics both with classical and modern cutting-edge approaches.
Collapse
Affiliation(s)
| | | | | | - Piya Ghose
- The University of Texas at Arlington, USA.
| |
Collapse
|
11
|
Fernández-Lázaro D, Sanz B, Seco-Calvo J. The Mechanisms of Regulated Cell Death: Structural and Functional Proteomic Pathways Induced or Inhibited by a Specific Protein-A Narrative Review. Proteomes 2024; 12:3. [PMID: 38250814 PMCID: PMC10801515 DOI: 10.3390/proteomes12010003] [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: 12/04/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
Billions of cells die in us every hour, and our tissues do not shrink because there is a natural regulation where Cell Death (CD) is balanced with cell division. The process in which cells eliminate themselves in a controlled manner is called Programmed Cell Death (PCD). The PCD plays an important role during embryonic development, in maintaining homeostasis of the body's tissues, and in the elimination of damaged cells, under a wide range of physiological and developmental stimuli. A multitude of protein mediators of PCD have been identified and signals have been found to utilize common pathways elucidating the proteins involved. This narrative review focuses on caspase-dependent and caspase-independent PCD pathways. Included are studies of caspase-dependent PCD such as Anoikis, Catastrophe Mitotic, Pyroptosis, Emperitosis, Parthanatos and Cornification, and Caspase-Independent PCD as Wallerian Degeneration, Ferroptosis, Paraptosis, Entosis, Methuosis, and Extracellular Trap Abnormal Condition (ETosis), as well as neutrophil extracellular trap abnormal condition (NETosis) and Eosinophil Extracellular Trap Abnormal Condition (EETosis). Understanding PCD from those reported in this review could shed substantial light on the processes of biological homeostasis. In addition, identifying specific proteins involved in these processes is mandatory to identify molecular biomarkers, as well as therapeutic targets. This knowledge could provide the ability to modulate the PCD response and could lead to new therapeutic interventions in a wide range of diseases.
Collapse
Affiliation(s)
- Diego Fernández-Lázaro
- Department of Cellular Biology, Genetics, Histology and Pharmacology, Faculty of Health Sciences, University of Valladolid, Campus of Soria, 42004 Soria, Spain
- Neurobiology Research Group, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain
- SARCELLOMICS Research Group, 27071 León, Spain; (B.S.); (J.S.-C.)
| | - Begoña Sanz
- SARCELLOMICS Research Group, 27071 León, Spain; (B.S.); (J.S.-C.)
- Department of Physiology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Jesús Seco-Calvo
- SARCELLOMICS Research Group, 27071 León, Spain; (B.S.); (J.S.-C.)
- Department of Physiology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Institute of Biomedicine (IBIOMED), Universidad de León, 27071 León, Spain
| |
Collapse
|
12
|
Lv X, Wang B, Dong M, Wang W, Tang W, Qin J, Gao Y, Wei Y. The crosstalk between ferroptosis and autophagy in cancer. Autoimmunity 2023; 56:2289362. [PMID: 38069487 DOI: 10.1080/08916934.2023.2289362] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND In order to better understand the interplay between ferroptosis and autophagy, enhance the interpretation of the crosstalk between these two forms of regulated cell death, develop the effective pharmacological mechanisms for cancer treatment, discover novel biomarkers for better diagnostic, and envisage the future hotspots of the research on ferroptosis and autophagy, we harnessed bibliometric tools to study the articles published from 2012 to 2022 on the relationship between ferroptosis and autophagy. METHODS Web of Science Core Collection (WOSCC) database was used to conduct a comprehensive search and analysis of articles in this field from January 1, 2012, to September 1, 2022. The Citespace 6.1.R2 software and VOS viewer 6.1.8 software were utilized to analyze the overall structure of the network, network clusters, links between clusters, key nodes or pivot points, and pathways. RESULTS A total of 756 articles associated with the crosstalk between ferroptosis and autophagy were published in 512 journals by 4183 authors in 980 organizations from 55 countries or regions. The distribution of countries and organizations was demonstrated using CiteSpace and VOS viewer. The top three countries with the most articles were China (n = 511), United States (n = 166), and Germany (n = 37). The most productive institutions were Guangzhou Medical University and Central South University (n = 42), but their centralities were relatively low, which values were respective 0.04 and 0.03. Kang and Tang published the most articles related to ferroptosis and autophagy (n = 49), followed by Jiao Liu (n = 22), Guido Kroemer (n = 20), and Daniel Klionsky (n = 12). Published studies on ferroptosis and asthma have the most cited counts. The top three keywords with the highest frequencies were autophagy (n = 283), cell death (n = 243), and oxidative stress (n = 165). CONCLUSION Our results provide insights into the development of recognition related to the crosstalk between ferroptosis and autophagy, and the current molecular crosslinked mechanisms in the context of common signal transduction pathways or affecting cellular environment to induce the adaptive stress response and to activate the particular form of regulated cell death (RCD), and the development of cancer treatment based on novel targets and signaling regulatory networks provided by ferroptosis and autophagy.
Collapse
Affiliation(s)
- Xiaodi Lv
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Bin Wang
- Medicine School of Hexi College, Zhangye, Gansu, China
| | - Ming Dong
- Gumei community Health center of Minhang district of Shanghai, Shanghai, China
| | - Wenqian Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jingjing Qin
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Yanglai Gao
- Medicine School of Hexi College, Zhangye, Gansu, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Li C, Zhu X, Sun X, Guo X, Li W, Chen P, Shidlovskii YV, Zhou Q, Xue L. Slik maintains tissue homeostasis by preventing JNK-mediated apoptosis. Cell Div 2023; 18:16. [PMID: 37794497 PMCID: PMC10552427 DOI: 10.1186/s13008-023-00097-4] [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: 01/29/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND The c-Jun N-terminal kinase (JNK) pathway is an evolutionarily conserved regulator of cell death, which is essential for coordinating tissue homeostasis. In this study, we have characterized the Drosophila Ste20-like kinase Slik as a novel modulator of JNK pathway-mediated apoptotic cell death. RESULTS First, ectopic JNK signaling-triggered cell death is enhanced by slik depletion whereas suppressed by Slik overexpression. Second, loss of slik activates JNK signaling, which results in enhanced apoptosis and impaired tissue homeostasis. In addition, genetic epistasis analysis suggests that Slik acts upstream of or in parallel to Hep to regulate JNK-mediated apoptotic cell death. Moreover, Slik is necessary and sufficient for preventing physiologic JNK signaling-mediated cell death in development. Furthermore, introduction of STK10, the human ortholog of Slik, into Drosophila restores slik depletion-induced cell death and compromised tissue homeostasis. Lastly, knockdown of STK10 in human cancer cells also leads to JNK activation, which is cancelled by expression of Slik. CONCLUSIONS This study has uncovered an evolutionarily conserved role of Slik/STK10 in blocking JNK signaling, which is required for cell death inhibition and tissue homeostasis maintenance in development.
Collapse
Affiliation(s)
- Chenglin Li
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Xiaojie Zhu
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Xinyue Sun
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Xiaowei Guo
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Wenzhe Li
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Ping Chen
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Yulii V Shidlovskii
- Department of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Biology and General Genetics, Sechenov University, 8, bldg. 2 Trubetskaya St, Moscow, 119048, Russia
| | - Qian Zhou
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China.
| | - Lei Xue
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China.
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, Guangdong, China.
| |
Collapse
|
14
|
Kowalski S, Karska J, Łapińska Z, Hetnał B, Saczko J, Kulbacka J. An overview of programmed cell death: Apoptosis and pyroptosis-Mechanisms, differences, and significance in organism physiology and pathophysiology. J Cell Biochem 2023. [PMID: 37269535 DOI: 10.1002/jcb.30413] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 06/05/2023]
Abstract
Regulated cell death is an essential and heterogeneous process occurring in the life cycle of organisms, from embryonic development and aging to the regulation of homeostasis and organ maintenance. Under this term, we can distinguish many distinct pathways, including apoptosis and pyroptosis. Recently, there has been an increasing comprehension of the mechanisms governing these phenomena and their characteristic features. The coexistence of different types of cell death and the differences and similarities between them has been the subject of many studies. This review aims to present the latest literature in the field of pyroptosis and apoptosis and compare their molecular pathway's elements and significance in the physiology and pathophysiology of the organism.
Collapse
Affiliation(s)
- Szymon Kowalski
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Julia Karska
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Zofia Łapińska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Bartosz Hetnał
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
- Department of Immunology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| |
Collapse
|
15
|
Oubella A, Bimoussa A, Byadi S, Laamari Y, Fawzi M, N'ait Ousidi A, Oblak D, Auhmani A, Riahi A, Morjani H, Ait Itto MY. Cytotoxic and apoptotic effects of some (R)-carvone-isoxazoline derivatives on human fibrosarcoma and carcinoma cells: experimental evaluation for cytotoxicity, molecular docking and molecular dynamics studies. J Biomol Struct Dyn 2023; 41:1930-1943. [PMID: 35014592 DOI: 10.1080/07391102.2022.2025903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
This study aimed to analyze the cytotoxic and apoptotic effects of isoxazoline derivatives with monoterpene scaffold 9a-e in HT-1080 fibrosarcoma, MCF-7, and MDA-MB-231 breast carcinoma, and A-549 lung carcinoma. The cytotoxic effects data revealed that compounds 9a-e generally induced significant cell growth inhibition in all cell lines, with IC50 ranging from 10 to 30 µM. However, for compounds 9c and 9e, the IC50 reached a value of 100 µM in HT-1080 cells. Compounds 9a, 9b, and 9d could induce apoptosis in HT-1080 cells as demonstrated by Annexin-V labeling and Caspase-3/7 activity. The apoptotic effect was accompanied by cell cycle arrest in the S phase. Molecular docking and molecular dynamics confirmed the empirical assay results and confirmed the stability of the complex with the inhibition of the anti-apoptotic protein, leading to cancer cell death. Overall, these data suggest that the proposed isoxazoline derivatives may be potential candidates for further investigation to evaluate their efficacy and optimal use in cancer treatment.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ali Oubella
- Department of Chemistry, Faculty of Sciences Semlalia, Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Marrakech, Morocco
| | - Abdoullah Bimoussa
- Department of Chemistry, Faculty of Sciences Semlalia, Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Marrakech, Morocco
| | - Said Byadi
- Equipe de spectroscopie d'extraction et de valorisation, Synthèse organique, Laboratoire d'extraction et de valorisation, Faculté des sciences d'Ain Chock, Université Hassan II, Casablanca, Morocco
| | - Yassine Laamari
- Department of Chemistry, Faculty of Sciences Semlalia, Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Marrakech, Morocco
| | - Mourad Fawzi
- Department of Chemistry, Faculty of Sciences Semlalia, Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Marrakech, Morocco
| | - Abdellah N'ait Ousidi
- Department of Chemistry, Faculty of Sciences Semlalia, Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Marrakech, Morocco
| | - Domen Oblak
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Aziz Auhmani
- Department of Chemistry, Faculty of Sciences Semlalia, Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Marrakech, Morocco
| | - Abdelkhalek Riahi
- Equipe MSO, CNRS UMR 7312 Institut de Chimie Moléculaire Université de Reims Champagne-Ardenne, REIMS Cédex 2, France
| | - Hamid Morjani
- BioSpectroscopie Translationnelle, BioSpecT-EA7506, UFR de Pharmacie, Université de Reims Champagne-Ardenne, Reims Cedex, France
| | - My Youssef Ait Itto
- Department of Chemistry, Faculty of Sciences Semlalia, Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Marrakech, Morocco
| |
Collapse
|
16
|
Nössing C, Ryan KM. 50 years on and still very much alive: 'Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics'. Br J Cancer 2023; 128:426-431. [PMID: 36369364 PMCID: PMC9938139 DOI: 10.1038/s41416-022-02020-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/13/2022] Open
Abstract
Cell death is part of the lifecycle of every multicellular organism. Nineteenth-century pathologists already recognised that organised forms of cell death must exist to explain the demise and turnover of cells during metamorphosis (of insects), embryogenesis and normal tissue homoeostasis [1]. Nevertheless, Kerr, Wyllie and Currie in their seminal paper of 1972, were the first to collate and define the distinct morphological features of controlled cell death in different contexts [2]. To describe the processes of cell deletion observed under both physiological and pathological conditions, they coined the term 'Apoptosis' (derived from the Greek word 'ἀπόπτωσις', meaning 'dropping off or falling off' of petals from flowers). Kerr, Wyllie and Currie defined apoptosis as a mechanism 'complementary to mitosis in the regulation of animal cell populations'. In addition, they already recognised the potential to use this programmed form of cell death for cancer therapy, but they also emphasised the occurrence of apoptosis during cancer development. In this article, some 50 years after its initial publication in The British Journal of Cancer, we revaluate and put the authors initial assumptions and general concepts about apoptosis into the context of modern-day biology.
Collapse
Affiliation(s)
- Christoph Nössing
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Kevin M Ryan
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK.
- School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK.
| |
Collapse
|
17
|
Aghaei Z, Karbalaei N, Namavar MR, Haghani M, Razmkhah M, Ghaffari MK, Nemati M. Neuroprotective Effect of Wharton's Jelly-Derived Mesenchymal Stem Cell-Conditioned Medium (WJMSC-CM) on Diabetes-Associated Cognitive Impairment by Improving Oxidative Stress, Neuroinflammation, and Apoptosis. Stem Cells Int 2023; 2023:7852394. [PMID: 37081849 PMCID: PMC10113062 DOI: 10.1155/2023/7852394] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 04/22/2023] Open
Abstract
According to strong evidence, diabetes mellitus increases the risk of cognitive impairment. Mesenchymal stem cells have been shown to be potential therapeutic agents for neurological disorders. In the current study, we aimed to examine the effects of Wharton's jelly-derived mesenchymal stem cell-conditioned medium (WJMSC-CM) on learning and memory, oxidative stress, apoptosis, and histological changes in the hippocampus of diabetic rats. Randomly, 35 male Sprague Dawley rats weighing 260-300 g were allocated into five groups: control, diabetes, and three diabetic groups treated with insulin, WJMSC-CM, and DMEM. The injections of insulin (3 U/day, S.C.) and WJMSC-CM (10 mg/week, I.P.) were done for 60 days. The Morris water maze and open field were used to measure cognition and anxiety-like behaviors. Colorimetric assays were used to determine hippocampus glutathione (GSH), malondialdehyde (MDA) levels, and antioxidant enzyme activity. The histopathological evaluation of the hippocampus was performed by Nissl staining. The expression levels of Bax, Bcl-2, BDNF, and TNF-α were detected by real-time polymerase chain reaction (RT-PCR). According to our findings, WJMSC-CM significantly reduced and increased blood glucose and insulin levels, respectively. Enhanced cognition and improved anxiety-like behavior were also found in WJMSC-CM-treated diabetic rats. In addition, WJMSC-CM treatment reduced oxidative stress by lowering MDA and elevating GSH and antioxidant enzyme activity. Reduced TNF-α and enhanced Bcl-2 gene expression levels and elevated neuronal and nonneuronal (astrocytes and oligodendrocytes) cells were detected in the hippocampus of WJMSC-CM-treated diabetic rats. In conclusion, WJMSC-CM alleviated diabetes-related cognitive impairment by reducing oxidative stress, neuroinflammation, and apoptosis in diabetic rats.
Collapse
Affiliation(s)
- Zohre Aghaei
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Narges Karbalaei
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Namavar
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoud Haghani
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboobeh Razmkhah
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahdi Khorsand Ghaffari
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Nemati
- Department of Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
18
|
Li M, Yao L, He M, Huang H, Zheng H, Ma S, Zhong Z, Yu S, Sun M, Wang H. "Adjust Zang and arouse spirit" electroacupuncture ameliorates cognitive impairment by reducing endoplasmic reticulum stress in db/db mice. Front Endocrinol (Lausanne) 2023; 14:1185022. [PMID: 37152933 PMCID: PMC10154981 DOI: 10.3389/fendo.2023.1185022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Diabetic cognitive impairment (DCI) is a chronic complication of the central nervous system (CNS) caused by diabetes that affects learning and memory capacities over time. Recently, acupuncture has been shown to improve cognitive impairment in streptozotocin-induced diabetic rats. However, the effects of electroacupuncture on DCI and its underlying mechanism have not yet been elucidated in detail. Methods In this study, we used db/db mice as DCI animal models which showed low cognitive, learning and memory functions. Electroacupuncture significantly ameliorated DCI, which is reflected by better spatial learning and memory function using behavioral tests. The db/db mice with cognitive impairment were randomly divided into a model group (Mod) and an electroacupuncture treatment group (Acup), while db/m mice were used as a normal control group (Con). First, the mice were subjected to behavioural tests using the Morris water maze (MWM), and body weight, blood glucose, insulin, triglycerides (TG) and total cholesterol (TC) were observed; HE, Nissl, and TUNEL staining were used to observe the morphological changes and neuronal apoptosis in the mice hippocampus; Finally, Western blot and rt-PCR were applied to detect the essential proteins and mRNA of ERS and insulin signalling pathway, as well as the expression levels of Tau and Aβ. Results Electroacupuncture significantly ameliorated DCI, which is reflected by better spatial learning and memory function using behavioral tests. Moreover, electroacupuncture attenuated diabetes-induced morphological structure change, neuronal apoptosis in the hippocampus of db/db mice. Our results revealed that electroacupuncture could regulate the expression levels of Tau and Aβ by improving hippocampal ERS levels in db/db mice, inhibiting JNK activation, attenuating IRS1 serine phosphorylation, and restoring normal transduction of the insulin signaling pathway. Discussion In summary, ERS and insulin signaling pathway paly causal roles in DCI development. Electroacupuncture can significantly alleviate the pathogenesis of DCI, improve mice's learning and memory ability, and improve cognitive dysfunction. This study adds to our understanding of the effect of acupuncture on DCI and opens the door to further research on DCI.
Collapse
Affiliation(s)
- Mengyuan Li
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun Jilin, China
| | - Lin Yao
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun Jilin, China
| | - Min He
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun Jilin, China
| | - Haipeng Huang
- Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Haizhu Zheng
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun Jilin, China
| | - Shiqi Ma
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun Jilin, China
| | - Zhen Zhong
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun Jilin, China
| | - Shuo Yu
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun Jilin, China
| | - Mengmeng Sun
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun Jilin, China
- *Correspondence: Hongfeng Wang, ; Mengmeng Sun,
| | - Hongfeng Wang
- Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Hongfeng Wang, ; Mengmeng Sun,
| |
Collapse
|
19
|
Life-Saver or Undertaker: The Relationship between Primary Cilia and Cell Death in Vertebrate Embryonic Development. J Dev Biol 2022; 10:jdb10040052. [PMID: 36547474 PMCID: PMC9783631 DOI: 10.3390/jdb10040052] [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: 10/29/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The development of multicellular organisms requires a tightly coordinated network of cellular processes and intercellular signalling. For more than 20 years, it has been known that primary cilia are deeply involved in the mediation of intercellular signalling and that ciliary dysfunction results in severe developmental defects. Cilia-mediated signalling regulates cellular processes such as proliferation, differentiation, migration, etc. Another cellular process ensuring proper embryonic development is cell death. While the effect of cilia-mediated signalling on many cellular processes has been extensively studied, the relationship between primary cilia and cell death remains largely unknown. This article provides a short review on the current knowledge about this relationship.
Collapse
|
20
|
Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B, Liu J, Yu X, Shi S. Targeting cell death pathways for cancer therapy: recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol 2022; 15:174. [PMID: 36482419 PMCID: PMC9733270 DOI: 10.1186/s13045-022-01392-3] [Citation(s) in RCA: 316] [Impact Index Per Article: 105.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
Many types of human cells self-destruct to maintain biological homeostasis and defend the body against pathogenic substances. This process, called regulated cell death (RCD), is important for various biological activities, including the clearance of aberrant cells. Thus, RCD pathways represented by apoptosis have increased in importance as a target for the development of cancer medications in recent years. However, because tumor cells show avoidance to apoptosis, which causes treatment resistance and recurrence, numerous studies have been devoted to alternative cancer cell mortality processes, namely necroptosis, pyroptosis, ferroptosis, and cuproptosis; these RCD modalities have been extensively studied and shown to be crucial to cancer therapy effectiveness. Furthermore, evidence suggests that tumor cells undergoing regulated death may alter the immunogenicity of the tumor microenvironment (TME) to some extent, rendering it more suitable for inhibiting cancer progression and metastasis. In addition, other types of cells and components in the TME undergo the abovementioned forms of death and induce immune attacks on tumor cells, resulting in enhanced antitumor responses. Hence, this review discusses the molecular processes and features of necroptosis, pyroptosis, ferroptosis, and cuproptosis and the effects of these novel RCD modalities on tumor cell proliferation and cancer metastasis. Importantly, it introduces the complex effects of novel forms of tumor cell death on the TME and the regulated death of other cells in the TME that affect tumor biology. It also summarizes the potential agents and nanoparticles that induce or inhibit novel RCD pathways and their therapeutic effects on cancer based on evidence from in vivo and in vitro studies and reports clinical trials in which RCD inducers have been evaluated as treatments for cancer patients. Lastly, we also summarized the impact of modulating the RCD processes on cancer drug resistance and the advantages of adding RCD modulators to cancer treatment over conventional treatments.
Collapse
Affiliation(s)
- Xuhui Tong
- grid.452404.30000 0004 1808 0942Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rong Tang
- grid.452404.30000 0004 1808 0942Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Mingming Xiao
- grid.452404.30000 0004 1808 0942Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin Xu
- grid.452404.30000 0004 1808 0942Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Wang
- grid.452404.30000 0004 1808 0942Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bo Zhang
- grid.452404.30000 0004 1808 0942Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiang Liu
- grid.452404.30000 0004 1808 0942Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai, 200032 China ,grid.8547.e0000 0001 0125 2443Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China. .,Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
21
|
Fang W, Bell CM, Sapirstein A, Asami S, Leeper K, Zack DJ, Ji H, Kalhor R. Quantitative fate mapping: A general framework for analyzing progenitor state dynamics via retrospective lineage barcoding. Cell 2022; 185:4604-4620.e32. [PMID: 36423582 PMCID: PMC9708097 DOI: 10.1016/j.cell.2022.10.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 08/23/2022] [Accepted: 10/26/2022] [Indexed: 11/24/2022]
Abstract
Natural and induced somatic mutations that accumulate in the genome during development record the phylogenetic relationships of cells; whether these lineage barcodes capture the complex dynamics of progenitor states remains unclear. We introduce quantitative fate mapping, an approach to reconstruct the hierarchy, commitment times, population sizes, and commitment biases of intermediate progenitor states during development based on a time-scaled phylogeny of their descendants. To reconstruct time-scaled phylogenies from lineage barcodes, we introduce Phylotime, a scalable maximum likelihood clustering approach based on a general barcoding mutagenesis model. We validate these approaches using realistic in silico and in vitro barcoding experiments. We further establish criteria for the number of cells that must be analyzed for robust quantitative fate mapping and a progenitor state coverage statistic to assess the robustness. This work demonstrates how lineage barcodes, natural or synthetic, enable analyzing progenitor fate and dynamics long after embryonic development in any organism.
Collapse
Affiliation(s)
- Weixiang Fang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Claire M Bell
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Abel Sapirstein
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Soichiro Asami
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kathleen Leeper
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Donald J Zack
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| | - Reza Kalhor
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
22
|
Auhmani A, Fawzi M, Oubella A, El Mansouri AE, Bimoussa A, Ketatni EM, Saadi M, El Ammari L, Itto MYA, Morjani H. Novel hydrazono-2-iminothiazolidin-4-ones based on a monoterpenic skelton as potential antitumor agents: Synthesis, DFT studies, in vitro cytotoxicity, apoptosis inducing properties and molecular docking. Chem Biodivers 2022; 19:e202100836. [PMID: 35665594 DOI: 10.1002/cbdv.202100836] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 06/02/2022] [Indexed: 12/24/2022]
Abstract
A new series of analogs-2-imino-4-thiazolidinone hybrid derivatives were synthesized by treatment of limonaketone-thiosemicarbazone derivatives with ethyl bromoacetate and dimethyl acetylene dicarboxylate (DMAD) as the cyclizing reagents. The new derivatives were characterized by HRMS, 1 H NMR and 13 C NMR spectroscopy techniques and one of them is characterized by single crystal X-ray diffraction. The newly synthesized products were biologically evaluated in vitro for their cytotoxic activity against three cancer cell lines including fibrosarcoma (HT-1080), lung carcinoma (A549), and breast carcinoma (MCF-7). Data showed that compounds 9 and 10 were the most active against HT-1080 cancer cell lines with the IC 50 values of 15.85±1.75 and16.13±1.55µM, respectively. The possible mechanism of apoptosis induction by the derivatives was investigated using Annexin V staining, caspase-3/7 activity and cell cycle analysis. Compound 10 was highly induced apoptosis through caspase-3/7 activation and S-phase arrest in HT-1080, while compound 9 showed a moderate induction of apoptosis through G0/G1-phase arrest in the same cell line. The molecular docking showed that compounds 9 and 10 activated the caspase-3 by forming a stable protein-ligand complex. Besides, the presence of phenyl moiety in ligand 10 is responsible for the enhancement of the caspase-3 activation by the apparition of two additional hydrogen bonds with Cys163 and Gln161amino acids.
Collapse
Affiliation(s)
- Aziz Auhmani
- Universite Cadi Ayyad Faculte des Sciences Semlalia, CHEMISTRY, boulevard prince moulay abdellah, 40000, marrakech, MOROCCO
| | - Mourad Fawzi
- Universite Cadi Ayyad Faculte des Sciences Semlalia, chemistry, Boulevard prince moulay Abdellah, Marrakech, MOROCCO
| | - Ali Oubella
- Universite Cadi Ayyad Faculte des Sciences Semlalia, chemistry, Boulevard prince moulay Abdellah, Marrakech, MOROCCO
| | - Az-Eddine El Mansouri
- Universite Hassan II de Casablanca, chemistry, PO Box. 146 Casablanca, Casablanca, MOROCCO
| | - Abdoullah Bimoussa
- Universite Cadi Ayyad Faculte des Sciences Semlalia, chemistry, Boulevard prince moulay Abdellah, Marrakech, MOROCCO
| | - El Mostafa Ketatni
- Université sultan moulay slimane faculté des sciences et techniques, chemistry, PO Box 523, 23000 Beni-Mellal, Morocco, Béni-Mellal, MOROCCO
| | - Mohamed Saadi
- université mohamed V faculté des sciences, chemistry, Avenue Ibn Batouta, P. B 1014, Rabat, Morocco, Rabat, MOROCCO
| | - Lahcen El Ammari
- Mohammed V university faculté des sciences, chemistry, Avenue Ibn Batouta, P. B 1014, Rabat, Morocco, Rabat, MOROCCO
| | - Moulay Youssef Ait Itto
- université cadi ayyad faculté des sciences semlalia, chemistry, Boulevard prince moulay Abdellah, Marrakech, MOROCCO
| | - Hamid Morjani
- Université de reims champagne -ardenne, pharmacie, 51 rue Cognacq-Jay 51096 REIMS Cedex, Lille, FRANCE
| |
Collapse
|
23
|
Oubella A, Byadi S, Bimoussa A, Fawzi M, Auhmani A, Podlipnik C, Morjani H, Riahi A, Robert A, Itto MYA. Novel isoxazoline-linked 1,3,4-thiadiazole hybrids as anticancer agents: Design, synthesis, biological evaluation, molecular docking, and molecular dynamics simulation. Arch Pharm (Weinheim) 2022; 355:e2200066. [PMID: 35594031 DOI: 10.1002/ardp.202200066] [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: 02/08/2022] [Revised: 04/27/2022] [Accepted: 05/02/2022] [Indexed: 11/09/2022]
Abstract
In the current study, natural (R)-carvone was utilized as a starting material for the efficient synthesis of two series of isoxazoline derivatives bearing the 1,3,4-thiadiazole moiety. The new compounds were obtained in good yields and were characterized by 1 H and 13 C NMR and HRMS analysis. The newly synthesized monoterpenic isoxazoline 1,3,4-thiadiazole and their thiosemicarbazone intermediate derivatives were evaluated for their anticancer activity in four cancer cell lines (HT-1080, A-549, MCF-7, and MDA-MB-231). Most of the synthesized compounds exhibited moderate to high anticancer effects. Compound 13c showed the highest anticancer activity with IC50 values ranging from 19.33 ± 1.81 to 34.81 ± 3.03 µM. Further investigation revealed that compounds 12e and 13c could inhibit the cell growth of HT-1080 and MCF-7 cells by inducing apoptosis through caspase-3/7 activation. The apoptotic effect was accompanied by an S phase and G2/M cell cycle arrest for 13c and 12e, respectively. Compounds 12e and 13c were assessed in silico using molecular docking and molecular dynamics. We found that compound 13c is moderately active against the caspase-3 protein, which triggers apoptosis via intrinsic and extrinsic routes, making compound 13c a promising candidate to activate the proapoptotic protein (caspase-3).
Collapse
Affiliation(s)
- Ali Oubella
- Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Marrakesh, Morocco
| | - Said Byadi
- Equipe de Spectroscopie D'extraction et de Valorisation, Synthèse Organique, Laboratoire D'extraction et de Valorisation, Facultés des Sciences Ain Chock, Université Hassan II, Casablanca, Morocco
| | - Abdoullah Bimoussa
- Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Marrakesh, Morocco
| | - Mourad Fawzi
- Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Marrakesh, Morocco
| | - Aziz Auhmani
- Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Marrakesh, Morocco
| | - Crtomir Podlipnik
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Hamid Morjani
- BioSpectroscopie Translationnelle, BioSpecT-EA7506, UFR de Pharmacie, Université de Reims Champagne-Ardenne, Reims, France
| | - Abdelkhalek Riahi
- Equipe MSO, CNRS UMR 7312 Institut de Chimie Moléculaire (ICMR), Université de Reims Champagne-Ardenne, Reims, France
| | - Anthony Robert
- Equipe MSO, CNRS UMR 7312 Institut de Chimie Moléculaire (ICMR), Université de Reims Champagne-Ardenne, Reims, France
| | - My Youssef A Itto
- Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Marrakesh, Morocco
| |
Collapse
|
24
|
Mercau ME, Patwa S, Bhat KPL, Ghosh S, Rothlin CV. Cell death in development, maintenance, and diseases of the nervous system. Semin Immunopathol 2022; 44:725-738. [PMID: 35508671 DOI: 10.1007/s00281-022-00938-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/12/2022] [Indexed: 02/07/2023]
Abstract
Cell death, be it of neurons or glial cells, marks the development of the nervous system. Albeit relatively less so than in tissues such as the gut, cell death is also a feature of nervous system homeostasis-especially in context of adult neurogenesis. Finally, cell death is commonplace in acute brain injuries, chronic neurodegenerative diseases, and in some central nervous system tumors such as glioblastoma. Recent studies are enumerating the various molecular modalities involved in the execution of cells. Intimately linked with cell death are mechanisms of disposal that remove the dead cell and bring about a tissue-level response. Heretofore, the association between these methods of dying and physiological or pathological responses has remained nebulous. It is envisioned that careful cartography of death and disposal may reveal novel understandings of disease states and chart new therapeutic strategies in the near future.
Collapse
Affiliation(s)
- Maria E Mercau
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Siraj Patwa
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Krishna P L Bhat
- Department of Translational Molecular Pathology, Division of Pathology-Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sourav Ghosh
- Department of Neurology, School of Medicine, Yale University, New Haven, CT, USA.,Department of Pharmacology, School of Medicine, Yale University, New Haven, CT, USA
| | - Carla V Rothlin
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA. .,Department of Pharmacology, School of Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
25
|
Oubella A, Taia A, Byadi S, Ait Lahcen M, Bimoussa A, Essaber M, Podlipnik C, Morjani H, Ait Itto MY, Aatif A. Chemical profiling, cytotoxic activities through apoptosis induction in human fibrosarcoma and carcinoma cells, and molecular docking of some 1,2,3-triazole-isoxazoline hybrids using the eugenol as a precursors. J Biomol Struct Dyn 2022; 41:2759-2771. [PMID: 35174765 DOI: 10.1080/07391102.2022.2037466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In this research paper, we report the cytotoxic and apoptotic effects of 1,2,3-triazole derivatives in a unique 7a-g or hybrid form with isoxazoline 8a-g using the eugenol as a precursor in HT-1080 fibrosarcoma, MCF-7, and MDA-MB-231 breast carcinoma, and A-549 lung carcinoma. Data obtained on the cytotoxic effects have shown that hybrid compounds 8a-e induced a significant anticancer activity and are more important than the ones of 1,2,3-triazole derivatives 7a-g with IC50 ranging from 18 to 43 μM for the hybrids 8a-e and from 15 to 29 μM for mono-adducts 7a-g in all cell lines. Concerning the apoptotic study, compounds 7b and 8a can induce apoptosis in HT-1080 and A-549 cells as revealed by Annexin-V labeling and caspase-3/7 activity, also, the apoptotic effect was accompanied by cell cycle arrest at G2/M phase in the case of compounds 7b and 8a. Both compounds were evaluated in-silico through molecular docking and molecular dynamics and compound 8a is very active against Bcl-2 protein triggering apoptosis phenomenon by intrinsic pathway, therefore compound 8a is a potential candidate to inhibit the anti-apoptotic protein (Bcl-2).Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ali Oubella
- Department of Chemistry, Faculty of Sciences Semlalia, Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Marrakech, Morocco
| | - Abdelmaoujoud Taia
- Laboratory of Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, University of Cadi Ayyad, Marrakech, Morocco
| | - Said Byadi
- Equipe de spectroscopie d'extraction et de valorisation, Synthese organique, Laboratoire d'extraction et de valorisation, Faculté des sciences d'Ain Chock, Universite Hassan II, Casablanca, Morocco
| | - Marouane Ait Lahcen
- Laboratory of Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, University of Cadi Ayyad, Marrakech, Morocco
| | - Abdoullah Bimoussa
- Department of Chemistry, Faculty of Sciences Semlalia, Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Marrakech, Morocco
| | - Mohamed Essaber
- Laboratory of Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, University of Cadi Ayyad, Marrakech, Morocco
| | - Crtomir Podlipnik
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Hamid Morjani
- BioSpectroscopieTranslationnelle, BioSpecT-EA7506, UFR de Pharmacie, Université de Reims Champagne-Ardenne, Reims Cedex, France
| | - My Youssef Ait Itto
- Department of Chemistry, Faculty of Sciences Semlalia, Laboratory of Organic Synthesis and Physico-Molecular Chemistry, Marrakech, Morocco
| | - Abdeljalil Aatif
- Laboratory of Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, University of Cadi Ayyad, Marrakech, Morocco
| |
Collapse
|
26
|
Zhou X, Suo F, Haslinger K, Quax WJ. Artemisinin-Type Drugs in Tumor Cell Death: Mechanisms, Combination Treatment with Biologics and Nanoparticle Delivery. Pharmaceutics 2022; 14:395. [PMID: 35214127 PMCID: PMC8875250 DOI: 10.3390/pharmaceutics14020395] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Artemisinin, the most famous anti-malaria drug initially extracted from Artemisia annua L., also exhibits anti-tumor properties in vivo and in vitro. To improve its solubility and bioavailability, multiple derivatives have been synthesized. However, to reveal the anti-tumor mechanism and improve the efficacy of these artemisinin-type drugs, studies have been conducted in recent years. In this review, we first provide an overview of the effect of artemisinin-type drugs on the regulated cell death pathways, which may uncover novel therapeutic approaches. Then, to overcome the shortcomings of artemisinin-type drugs, we summarize the recent advances in two different therapeutic approaches, namely the combination therapy with biologics influencing regulated cell death, and the use of nanocarriers as drug delivery systems. For the former approach, we discuss the superiority of combination treatments compared to monotherapy in tumor cells based on their effects on regulated cell death. For the latter approach, we give a systematic overview of nanocarrier design principles used to deliver artemisinin-type drugs, including inorganic-based nanoparticles, liposomes, micelles, polymer-based nanoparticles, carbon-based nanoparticles, nanostructured lipid carriers and niosomes. Both approaches have yielded promising findings in vitro and in vivo, providing a strong scientific basis for further study and upcoming clinical trials.
Collapse
Affiliation(s)
| | | | - Kristina Haslinger
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (X.Z.); (F.S.)
| | - Wim J. Quax
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; (X.Z.); (F.S.)
| |
Collapse
|
27
|
Deep Conservation of Hid-Like RHG Gene Family Homologs in Winged Insects Revealed by "Taxon Hopping" BLAST. INSECTS 2021; 12:insects12110957. [PMID: 34821758 PMCID: PMC8620624 DOI: 10.3390/insects12110957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022]
Abstract
Together with sickle (skl), the Drosophila paralogs reaper (rpr), head involution defective (hid), and grim (RHG) control a critical switch in the induction of programmed cell death. RHG homologs have been identified in other dipteran and lepidopteran species but not beyond. Revisiting this issue with a "taxon hopping" BLAST search strategy in current genome and transcriptome resources, I detected high confidence RHG homologs in Coleoptera, Hymenoptera, Hemiptera, and Dictyoptera. Analyses of gene structure and protein sequence conservation revealed aconserved splicing pattern and highly conserved amino acid residues at both the N- and C-terminal ends that identify hid as the most ancestrally organized RHG gene family member in Drosophila. hid-like RHG homologs were also detected in mosquitoes, redefining their michelob_x (mx) genes as an expansion of derived RHG homologs. Only singleton homologs were detected in the large majority of other insect clades. Lepidopteran RHG homologs, however, stand out by producing an evolutionarily-derived splice isoform, identified in previous work, in addition to the newly detected hid-like isoform. Exceptional sequence diversification of select RHG homologs at the family- and genus-level explain their previous elusiveness in important insect genome model species like the red flour beetle Tribolium castaneum and the pea aphid Acyrthosiphon pisum. Combined, these findings expand the minimal age of the RHG gene family by about 100 million years and open new avenues for molecular cell death studies in insects.
Collapse
|
28
|
Oubella A, El Mansouri AE, Fawzi M, Bimoussa A, Laamari Y, Auhmani A, Morjani H, Robert A, Riahi A, Youssef Ait Itto M. Thiazolidinone-linked1,2,3-triazoles with monoterpenic skeleton as new potential anticancer agents: Design, synthesis and molecular docking studies. Bioorg Chem 2021; 115:105184. [PMID: 34333421 DOI: 10.1016/j.bioorg.2021.105184] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/15/2021] [Accepted: 07/13/2021] [Indexed: 01/07/2023]
Abstract
A novel series of 1,2,3-triazole-thiazolidinone-carvone hybrid compounds has been designed and synthesized using the copper-catalyzed Huisgen azide-alkyne 1,3-dipolar cycloaddition (CuAAC) process based on (R)-Carvone-O-propargylated 5-hydroxybenzylidene-thiazolidin-4-one derivative as starting material. All compounds were characterized and identified based on their NMR and HRMS spectroscopic data. HMBC correlations confirm that under the CuAAC reaction conditions, only the 1,4-disubstituted triazole regioisomers were formed. The targeted 1,2,3-triazole-thiazolidinone-carvone hybrids and their precursors were evaluated for their cytotoxic activity against four human cancer cell lines, including fibrosarcoma (HT-1080), lung carcinoma (A-549), and breast carcinoma (MCF-7 and MDA-MB-231). The obtained data showed that most of these compounds have moderate anti-proliferative activity with IC50 values between 15.04 ± 0.71 and 42.22 ± 1.20 µM. The mechanism of action of the most active compounds 14e and 14f suggested that they induce apoptosis through caspase-3/7 activation, and the compound 14e elicited S-phase arrest, while compound 14f evoked G2/M phase blockade. The molecular docking confirmed that compounds 14e and 14f were nicely bonded with caspace-3 leading up to stable protein-ligand complexes.
Collapse
Affiliation(s)
- Ali Oubella
- Laboratoire de Synthèse Organique et Physico-Chimie Moléculaire, Département de Chimie, Faculté des Sciences, Semlalia B.P 2390, Marrakech 40001, Morocco.
| | - Az-Eddine El Mansouri
- Laboratoire de Materiaux, Catalyse & Valorisation des Ressources Naturelles, URAC 24, Faculté des Sciences et Techniques, Universite Hassan II, Casablanca, Morocco; Laboratory of Biomolecular and Medicinal Chemistry, Department of Chemistry, Faculty of Science Semlalia, BP 2390, Marrakech 40001, Morocco
| | - Mourad Fawzi
- Laboratoire de Synthèse Organique et Physico-Chimie Moléculaire, Département de Chimie, Faculté des Sciences, Semlalia B.P 2390, Marrakech 40001, Morocco
| | - Abdoullah Bimoussa
- Laboratoire de Synthèse Organique et Physico-Chimie Moléculaire, Département de Chimie, Faculté des Sciences, Semlalia B.P 2390, Marrakech 40001, Morocco
| | - Yassine Laamari
- Laboratoire de Synthèse Organique et Physico-Chimie Moléculaire, Département de Chimie, Faculté des Sciences, Semlalia B.P 2390, Marrakech 40001, Morocco
| | - Aziz Auhmani
- Laboratoire de Synthèse Organique et Physico-Chimie Moléculaire, Département de Chimie, Faculté des Sciences, Semlalia B.P 2390, Marrakech 40001, Morocco
| | - Hamid Morjani
- BioSpectroscopie Translationnelle, BioSpecT-EA7506, UFR de Pharmacie, Université de Reims Champagne-Ardenne, 51 Rue Cognacq Jay, 51096, Reims Cedex, France
| | - Anthony Robert
- Equipe MSO, CNRS UMR 7312 Institut de Chimie Moléculaire Université de Reims Champagne-Ardenne, Bat. Europol'Agro - Moulin de La Housse UFR Sciences B.P., 1039, 51687 REIMS Cédex 2, France
| | - Abdelkhalek Riahi
- Equipe MSO, CNRS UMR 7312 Institut de Chimie Moléculaire Université de Reims Champagne-Ardenne, Bat. Europol'Agro - Moulin de La Housse UFR Sciences B.P., 1039, 51687 REIMS Cédex 2, France
| | - My Youssef Ait Itto
- Laboratoire de Synthèse Organique et Physico-Chimie Moléculaire, Département de Chimie, Faculté des Sciences, Semlalia B.P 2390, Marrakech 40001, Morocco.
| |
Collapse
|
29
|
Yawson EO, Akinola OB. Hippocampal cellular changes in androgen deprived insulin resistant rats. Metab Brain Dis 2021; 36:1037-1048. [PMID: 33666820 DOI: 10.1007/s11011-021-00678-8] [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: 08/31/2020] [Accepted: 01/26/2021] [Indexed: 10/22/2022]
Abstract
Androgen deprivation can be achieved through testosterone antagonists (chemical castration) with or without orchidectomy. We use a rat model to characterize hippocampal structural and functional changes that might be associated with a subset population of androgen deprived insulin-resistant patients. Adult male Wistar rats assigned into six (6) groups: control group (distilled water/sham), orchiectomy group (bilateral orchiectomy), flutamide group (oral flutamide; 11 mg/kg body weight), diabetes group (multiple low-dose of streptozotocin (STZ; 30 mg/kg body weight intraperitoneally), orchiectomy and diabetic group (bilateral orchiectomy with 30 mg/kg body weight of STZ), and orchiectomy/diabetic/flutamide group (bilateral orchiectomy with 30 mg/kg body weight of STZ with 11 mg/kg body weight of flutamide). Animals were sacrificed at 30 and 60 days respectively. Spatial learning and working memory behavior were assessed; while total plasma; testosterone, insulin levels, and fasting blood glucose were assayed; the Homeostasis model for insulin resistance was also calculated. Histological examinations by H&E and CFV, while immunohistochemical analysis of astrocytes, P53 protein, and NSE were performed. Androgen deprived insulin-resistant state caused altered learning and cognitive behavior through decreased percentage correct alternation to an increased escape latency period. Significant bidirectional correlates exist between the hormonal profiles relative to the control group (p < 0.05), especially in the 60 days post-orchiectomy. While histological and immunohistochemical data indicate microcellular derangement. That the summate effects of androgen deprivation and impaired insulin signaling exacerbate hippocampal neurodegenerative changes that merit further studies.
Collapse
Affiliation(s)
- Emmanuel O Yawson
- Neuroendocrinology division, Department of Anatomy, University of Ilorin, Ilorin, Nigeria
| | - Oluwole B Akinola
- Neuroendocrinology division, Department of Anatomy, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Nigeria.
| |
Collapse
|
30
|
The morphogenetic changes that lead to cell extrusion in development and cell competition. Dev Biol 2021; 477:1-10. [PMID: 33984304 DOI: 10.1016/j.ydbio.2021.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/28/2021] [Accepted: 05/05/2021] [Indexed: 12/30/2022]
Abstract
Cell extrusion is a morphogenetic process in which unfit or dying cells are eliminated from the tissue at the interface with healthy neighbours in homeostasis. This process is also highly associated with cell fate specification followed by differentiation in development. Spontaneous cell death occurs in development and inhibition of this process can result in abnormal development, suggesting that survival or death is part of cell fate specification during morphogenesis. Moreover, spontaneous somatic mutations in oncogenes or tumour suppressor genes can trigger new morphogenetic events at the interface with healthy cells. Cell competition is considered as the global quality control mechanism for causing unfit cells to be eliminated at the interface with healthy neighbours in proliferating tissues. In this review, I will discuss variations of cell extrusion that are coordinated by unfit cells and healthy neighbours in relation to the geometry and topology of the tissue in development and cell competition.
Collapse
|
31
|
Nicotinamide Supplementation Improves Oocyte Quality and Offspring Development by Modulating Mitochondrial Function in an Aged Caenorhabditis elegans Model. Antioxidants (Basel) 2021; 10:antiox10040519. [PMID: 33810497 PMCID: PMC8066965 DOI: 10.3390/antiox10040519] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022] Open
Abstract
Aging is associated with a decline in the quality of biological functions. Among the aging processes, reproductive aging is a critical process because of its intergenerational effects. However, the mechanisms underlying reproductive aging remain largely unknown. Female reproductive aging is the primary reason for limited fertility in mammals. Therefore, we attempted to investigate a modulator that can control female reproductive aging using a Caenorhabditis elegans model. In the present study, we examined the role of nicotinamide (NAM) in oocyte quality and offspring development. The levels of reactive oxygen species (ROS) and oxidative stress responses in aged oocytes, embryonic lethality, and developmental growth of the offspring were examined with maternal NAM supplementation. Supplementation with NAM improved oocyte quality, decreased embryonic lethality, and promoted germ cell apoptosis. Furthermore, NAM supplementation in aged mothers reduced ROS accumulation and improved mitochondrial function in oocytes. Consequently, the developmental growth and motility of offspring were improved. These findings suggest that NAM supplementation improves the health of the offspring produced by aged mothers through improved mitochondrial function. Taken together, our results imply that NAM supplementation in the aged mother improves oocyte quality and protects offspring by modulating mitochondrial function.
Collapse
|
32
|
Aguilera A, Klemenčič M, Sueldo DJ, Rzymski P, Giannuzzi L, Martin MV. Cell Death in Cyanobacteria: Current Understanding and Recommendations for a Consensus on Its Nomenclature. Front Microbiol 2021; 12:631654. [PMID: 33746925 PMCID: PMC7965980 DOI: 10.3389/fmicb.2021.631654] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/11/2021] [Indexed: 01/31/2023] Open
Abstract
Cyanobacteria are globally widespread photosynthetic prokaryotes and are major contributors to global biogeochemical cycles. One of the most critical processes determining cyanobacterial eco-physiology is cellular death. Evidence supports the existence of controlled cellular demise in cyanobacteria, and various forms of cell death have been described as a response to biotic and abiotic stresses. However, cell death research in this phylogenetic group is a relatively young field and understanding of the underlying mechanisms and molecular machinery underpinning this fundamental process remains largely elusive. Furthermore, no systematic classification of modes of cell death has yet been established for cyanobacteria. In this work, we analyzed the state of knowledge in the field of cyanobacterial cell death. Based on that, we propose unified criterion for the definition of accidental, regulated, and programmed forms of cell death in cyanobacteria based on molecular, biochemical, and morphologic aspects following the directions of the Nomenclature Committee on Cell Death (NCCD). With this, we aim to provide a guide to standardize the nomenclature related to this topic in a precise and consistent manner, which will facilitate further ecological, evolutionary, and applied research in the field of cyanobacterial cell death.
Collapse
Affiliation(s)
- Anabella Aguilera
- Centre for Ecology and Evolution in Microbial Model Systems (EEMiS), Linnaeus University, Kalmar, Sweden
| | - Marina Klemenčič
- Department of Chemistry and Biochemistry, Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Daniela J. Sueldo
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznan´, Poland
- Integrated Science Association (ISA), Universal Scientific Education and Research Network (USERN), Poznan´, Poland
| | - Leda Giannuzzi
- Centro de Investigación y Desarrollo en Criotecnología de Alimentos, Consejo Nacional de Investigaciones Científicas y Tecnológicas, Universidad Nacional de La Plata, La Plata, Argentina
- Área de Toxicología General, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - María Victoria Martin
- Instituto de Investigaciones en Biodiversidad y Biotecnología (INBIOTEC-CONICET), Fundación para Investigaciones Biológicas Aplicadas (CIB-FIBA), Mar del Plata, Argentina
| |
Collapse
|
33
|
Santagostino SF, Radaelli E. Special Focus on Regulated Cell Death: Emerging Mechanisms and Current Perspectives in Biology and Pathology. Vet Pathol 2021; 58:594-595. [PMID: 33576309 DOI: 10.1177/0300985820985326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
34
|
Gartner A, Engebrecht J. DNA repair, recombination, and damage signaling. Genetics 2021; 220:6522877. [PMID: 35137093 PMCID: PMC9097270 DOI: 10.1093/genetics/iyab178] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/10/2021] [Indexed: 01/09/2023] Open
Abstract
DNA must be accurately copied and propagated from one cell division to the next, and from one generation to the next. To ensure the faithful transmission of the genome, a plethora of distinct as well as overlapping DNA repair and recombination pathways have evolved. These pathways repair a large variety of lesions, including alterations to single nucleotides and DNA single and double-strand breaks, that are generated as a consequence of normal cellular function or by external DNA damaging agents. In addition to the proteins that mediate DNA repair, checkpoint pathways have also evolved to monitor the genome and coordinate the action of various repair pathways. Checkpoints facilitate repair by mediating a transient cell cycle arrest, or through initiation of cell suicide if DNA damage has overwhelmed repair capacity. In this chapter, we describe the attributes of Caenorhabditis elegans that facilitate analyses of DNA repair, recombination, and checkpoint signaling in the context of a whole animal. We review the current knowledge of C. elegans DNA repair, recombination, and DNA damage response pathways, and their role during development, growth, and in the germ line. We also discuss how the analysis of mutational signatures in C. elegans is helping to inform cancer mutational signatures in humans.
Collapse
Affiliation(s)
- Anton Gartner
- Department for Biological Sciences, IBS Center for Genomic Integrity, Ulsan National Institute of Science and Technology, Ulsan 689-798, Republic of Korea,Corresponding author: (A.G.); (J.E.)
| | - JoAnne Engebrecht
- Department of Molecular and Cellular Biology, University of California Davis, Davis, CA 95616, USA,Corresponding author: (A.G.); (J.E.)
| |
Collapse
|
35
|
Ramos-Ibeas P, Gimeno I, Cañón-Beltrán K, Gutiérrez-Adán A, Rizos D, Gómez E. Senescence and Apoptosis During in vitro Embryo Development in a Bovine Model. Front Cell Dev Biol 2020; 8:619902. [PMID: 33392207 PMCID: PMC7775420 DOI: 10.3389/fcell.2020.619902] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
According to the World Health Organization, infertility affects up to 14% of couples under reproductive age, leading to an exponential rise in the use of assisted reproduction as a route for conceiving a baby. In the same way, thousands of embryos are produced in cattle and other farm animals annually, leading to increased numbers of individuals born. All reproductive manipulations entail deviations of natural phenotypes and genotypes, with in vitro embryo technologies perhaps showing the biggest effects, although these alterations are still emerging. Most of these indications have been provided by animal models, in particular the bovine species, due to its similarities to human early embryo development. Oocytes and embryos are highly sensitive to environmental stress in vivo and in vitro. Thus, during in vitro culture, a number of stressful conditions affect embryonic quality and viability, inducing subfertility and/or long-term consequences that may reach the offspring. A high proportion of the embryos produced in vitro are arrested at a species-specific stage of development during the first cell divisions. These arrested embryos do not show signs of programmed cell death during early cleavage stages. Instead, defective in vitro produced embryos would enter a permanent cell cycle arrest compatible with cellular senescence, in which they show active metabolism and high reactive oxygen species levels. Later in development, mainly during the morula and blastocyst stages, apoptosis would mediate the elimination of certain cells, accomplishing both a physiological role in to balancing cell proliferation and death, and a pathological role preventing the transmission of damaged cells with an altered genome. The latter would acquire relevant importance in in vitro produced embryos that are submitted to stressful environmental stimuli. In this article, we review the mechanisms mediating apoptosis and senescence during early embryo development, with a focus on in vitro produced bovine embryos. Additionally, we shed light on the protective role of senescence and apoptosis to ensure that unhealthy cells and early embryos do not progress in development, avoiding long-term detrimental effects.
Collapse
Affiliation(s)
- Priscila Ramos-Ibeas
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain
| | - Isabel Gimeno
- Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), Gijón, Spain
| | - Karina Cañón-Beltrán
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain
| | - Alfonso Gutiérrez-Adán
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain
| | - Dimitrios Rizos
- Department of Animal Reproduction, National Institute for Agriculture and Food Research and Technology (INIA), Madrid, Spain
| | - Enrique Gómez
- Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), Gijón, Spain
| |
Collapse
|
36
|
Infection of Mammals and Mosquitoes by Alphaviruses: Involvement of Cell Death. Cells 2020; 9:cells9122612. [PMID: 33291372 PMCID: PMC7762023 DOI: 10.3390/cells9122612] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/27/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022] Open
Abstract
Alphaviruses, such as the chikungunya virus, are emerging and re-emerging viruses that pose a global public health threat. They are transmitted by blood-feeding arthropods, mainly mosquitoes, to humans and animals. Although alphaviruses cause debilitating diseases in mammalian hosts, it appears that they have no pathological effect on the mosquito vector. Alphavirus/host interactions are increasingly studied at cellular and molecular levels. While it seems clear that apoptosis plays a key role in some human pathologies, the role of cell death in determining the outcome of infections in mosquitoes remains to be fully understood. Here, we review the current knowledge on alphavirus-induced regulated cell death in hosts and vectors and the possible role they play in determining tolerance or resistance of mosquitoes.
Collapse
|
37
|
La Rosa P, Petrillo S, Fiorenza MT, Bertini ES, Piemonte F. Ferroptosis in Friedreich's Ataxia: A Metal-Induced Neurodegenerative Disease. Biomolecules 2020; 10:biom10111551. [PMID: 33202971 PMCID: PMC7696618 DOI: 10.3390/biom10111551] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023] Open
Abstract
Ferroptosis is an iron-dependent form of regulated cell death, arising from the accumulation of lipid-based reactive oxygen species when glutathione-dependent repair systems are compromised. Lipid peroxidation, mitochondrial impairment and iron dyshomeostasis are the hallmark of ferroptosis, which is emerging as a crucial player in neurodegeneration. This review provides an analysis of the most recent advances in ferroptosis, with a special focus on Friedreich's Ataxia (FA), the most common autosomal recessive neurodegenerative disease, caused by reduced levels of frataxin, a mitochondrial protein involved in iron-sulfur cluster synthesis and antioxidant defenses. The hypothesis is that the iron-induced oxidative damage accumulates over time in FA, lowering the ferroptosis threshold and leading to neuronal cell death and, at last, to cardiac failure. The use of anti-ferroptosis drugs combined with treatments able to activate the antioxidant response will be of paramount importance in FA therapy, such as in many other neurodegenerative diseases triggered by oxidative stress.
Collapse
Affiliation(s)
- Piergiorgio La Rosa
- Department of Psychology, Division of Neuroscience, Sapienza University of Rome, 00185 Rome, Italy; (P.L.R.); (M.T.F.)
| | - Sara Petrillo
- Unit of Muscular and Neurodegenerative Diseases, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (E.S.B.)
| | - Maria Teresa Fiorenza
- Department of Psychology, Division of Neuroscience, Sapienza University of Rome, 00185 Rome, Italy; (P.L.R.); (M.T.F.)
| | - Enrico Silvio Bertini
- Unit of Muscular and Neurodegenerative Diseases, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (E.S.B.)
| | - Fiorella Piemonte
- Unit of Muscular and Neurodegenerative Diseases, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (E.S.B.)
- Correspondence: ; Tel.: +39-06-6859-2102
| |
Collapse
|
38
|
Design, synthesis, biological evaluation and molecular docking of new uracil analogs-1,2,4-oxadiazole hybrids as potential anticancer agents. Bioorg Med Chem Lett 2020; 30:127438. [DOI: 10.1016/j.bmcl.2020.127438] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/03/2020] [Accepted: 07/21/2020] [Indexed: 01/12/2023]
|
39
|
Developmental Changes in Potassium Channels in Low Threshold Myelinated Ah-type Vagal Ganglion Neurons in Rats. Neuroscience 2020; 429:256-263. [PMID: 31962146 DOI: 10.1016/j.neuroscience.2020.01.004] [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: 08/03/2019] [Revised: 12/27/2019] [Accepted: 01/02/2020] [Indexed: 11/23/2022]
Abstract
Myelinated Ah-type vagal ganglion neurons (VGNs) were specific subpopulation in adult females, rather than neonate and key players in sexual dimorphism in baroreflex afferent function and closely associated with estrogen. However, the gender related development changes in Ah-type VGNs remains unknown. To quantify the developmental changes in ion channels overtime, the whole-cell patch-clamp technique was performed and three afferent fiber types of VGNs were identified upon electrophysiological/pharmacological validations. The K+ currents were recorded with or without specific blockers from postnatal day 4-32 and adult in both sexes. The electrophysiological data conjugated with analysis of action potential (AP) trajectory strongly indicated that in male rats, Ah-types were likely to disappear or transform during development. The percentage of myelinated A-, Ah-, and unmyelinated C-type afferents in females remained relatively steady during the 4-32-day period. Conversely, Ah-type afferents in males declined from levels comparable with those in females at birth to near absence in adulthood at 32 days. The coordinated changes in the current density of certain ion channels may be the underlying mechanism of developmental changes in AP waveform and neuroexcitability. As expected, the coordinated change between the down-regulation of iberiotoxin-sensitive and up-regulation of 4-aminopyridine-sensitive K+ currents played a key role in shaping AP and neuroexcitability in Ah-types during development. Our results demonstrated that the myelinated Ah-type VGNs in males almost disappear at 4 weeks old where closes to adult and the correlative ion channel changes contribute to the sexual dimorphism in visceral afferent function.
Collapse
|
40
|
Puñal VM, Paisley CE, Brecha FS, Lee MA, Perelli RM, Wang J, O’Koren EG, Ackley CR, Saban DR, Reese BE, Kay JN. Large-scale death of retinal astrocytes during normal development is non-apoptotic and implemented by microglia. PLoS Biol 2019; 17:e3000492. [PMID: 31626642 PMCID: PMC6821132 DOI: 10.1371/journal.pbio.3000492] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 10/30/2019] [Accepted: 09/26/2019] [Indexed: 12/28/2022] Open
Abstract
Naturally occurring cell death is a fundamental developmental mechanism for regulating cell numbers and sculpting developing organs. This is particularly true in the nervous system, where large numbers of neurons and oligodendrocytes are eliminated via apoptosis during normal development. Given the profound impact of death upon these two major cell populations, it is surprising that developmental death of another major cell type—the astrocyte—has rarely been studied. It is presently unclear whether astrocytes are subject to significant developmental death, and if so, how it occurs. Here, we address these questions using mouse retinal astrocytes as our model system. We show that the total number of retinal astrocytes declines by over 3-fold during a death period spanning postnatal days 5–14. Surprisingly, these astrocytes do not die by apoptosis, the canonical mechanism underlying the vast majority of developmental cell death. Instead, we find that microglia engulf astrocytes during the death period to promote their developmental removal. Genetic ablation of microglia inhibits astrocyte death, leading to a larger astrocyte population size at the end of the death period. However, astrocyte death is not completely blocked in the absence of microglia, apparently due to the ability of astrocytes to engulf each other. Nevertheless, mice lacking microglia showed significant anatomical changes to the retinal astrocyte network, with functional consequences for the astrocyte-associated vasculature leading to retinal hemorrhage. These results establish a novel modality for naturally occurring cell death and demonstrate its importance for the formation and integrity of the retinal gliovascular network. A study of the neonatal mouse retina shows that developmental cell death of retinal astrocytes does not occur by apoptosis but is instead mediated by microglia, which kill and engulf astrocytes to effect their developmental removal.
Collapse
Affiliation(s)
- Vanessa M. Puñal
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Caitlin E. Paisley
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Federica S. Brecha
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Monica A. Lee
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Robin M. Perelli
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Jingjing Wang
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Emily G. O’Koren
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Caroline R. Ackley
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States of America
- Department of Cellular, Molecular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, United States of America
| | - Daniel R. Saban
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Benjamin E. Reese
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, United States of America
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, California, United States of America
| | - Jeremy N. Kay
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
41
|
Claes M, De Groef L, Moons L. Target-Derived Neurotrophic Factor Deprivation Puts Retinal Ganglion Cells on Death Row: Cold Hard Evidence and Caveats. Int J Mol Sci 2019; 20:E4314. [PMID: 31484425 PMCID: PMC6747494 DOI: 10.3390/ijms20174314] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
Glaucoma and other optic neuropathies are characterized by axonal transport deficits. Axonal cargo travels back and forth between the soma and the axon terminus, a mechanism ensuring homeostasis and the viability of a neuron. An example of vital molecules in the axonal cargo are neurotrophic factors (NTFs). Hindered retrograde transport can cause a scarcity of those factors in the retina, which in turn can tilt the fate of retinal ganglion cells (RGCs) towards apoptosis. This postulation is one of the most widely recognized theories to explain RGC death in the disease progression of glaucoma and is known as the NTF deprivation theory. For several decades, research has been focused on the use of NTFs as a novel neuroprotective glaucoma treatment. Until now, results in animal models have been promising, but translation to the clinic has been highly disappointing. Are we lacking important knowledge to lever NTF therapies towards the therapeutic armamentarium? Or did we get the wrong end of the stick regarding the NTF deprivation theory? In this review, we will tackle the existing evidence and caveats advocating for and against the target-derived NTF deprivation theory in glaucoma, whilst digging into associated therapy efforts.
Collapse
Affiliation(s)
- Marie Claes
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lies De Groef
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lieve Moons
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
42
|
Kumar RS, Almansour AI, Arumugam N, Mohammad F, Kotresha D, Menéndez JC. Spirooxindole-pyrrolidine heterocyclic hybrids promotes apoptosis through activation of caspase-3. Bioorg Med Chem 2019; 27:2487-2498. [DOI: 10.1016/j.bmc.2019.03.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/28/2019] [Accepted: 03/04/2019] [Indexed: 12/27/2022]
|
43
|
Sun Y, Zhang D, Li C, Huang J, Li W, Qiu Y, Mao A, Zhou M, Xue L. Lic regulates JNK-mediated cell death in Drosophila. Cell Prolif 2019; 52:e12593. [PMID: 30847993 PMCID: PMC6536442 DOI: 10.1111/cpr.12593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 01/01/2023] Open
Abstract
Objectives The evolutionary conserved JNK pathway plays crucial role in cell death, yet factors that modulate this signalling have not been fully disclosed. In this study, we aim to identify additional factors that regulate JNK signalling in cell death, and characterize the underlying mechanisms. Materials and Methods Drosophila were raised on standard media, and cross was carried out at 25°C. The Gal4/UAS system was used to express proteins or RNAi in a specific temporal and spatial pattern. Gene expression was revealed by GFP fluorescence, X‐gal staining or immunostaining of 3rd instar larval eye and wing discs. Cell death was visualized by acridine orange (AO) staining. Images of fly eyes and wings were taken by OLYMPUS microscopes. Results We found that licorne (lic) encoding the Drosophila MKK3 is an essential regulator of JNK‐mediated cell death. Firstly, loss of lic suppressed ectopic Egr‐triggered JNK activation and cell death in eye and wing development. Secondary, lic is necessary for loss‐of‐cell polarity‐induced, physiological JNK‐dependent cell death in wing development. Thirdly, Lic overexpression is sufficient to initiate JNK‐mediated cell death in developing eyes and wings. Furthermore, ectopic Lic activates JNK signalling by promoting JNK phosphorylation. Finally, genetic epistatic analysis confirmed that Lic acts in parallel with Hep in the Egr‐JNK pathway. Conclusions This study not only identified Lic as a novel component of the JNK signalling, but also disclosed the crucial roles and mechanism of Lic in cell death.
Collapse
Affiliation(s)
- Yihao Sun
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Di Zhang
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Chenglin Li
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Jiuhong Huang
- International Academy of Targeted Therapeutics and Innovation, Chongqing University of Arts and Sciences, Chongqing, China
| | - Wenzhe Li
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Yu Qiu
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Aiwu Mao
- Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingcheng Zhou
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Lei Xue
- The First Rehabilitation Hospital of Shanghai, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| |
Collapse
|
44
|
Sijilmassi O, López-Alonso JM, Del Río Sevilla A, Murillo González J, Barrio Asensio MDC. Biometric Alterations of Mouse Embryonic Eye Structures Due to Short-Term Folic Acid Deficiency. Curr Eye Res 2018; 44:428-435. [PMID: 30403890 DOI: 10.1080/02713683.2018.1545911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Folic acid (FA) is an essential nutrient for normal embryonic development. FA deficiency (FAD) in maternal diet increases the risk of several defects among the progeny, especially, neural tube defects. The eye begins its development from the neural tube; however, the relationship between FAD and ocular development in the offspring has been little explored and it isn't known how the FAD affects the formation of the eye. Our objective was to analyze the effect of maternal FAD on mouse embryos ocular biometry. METHODS Female mice C57/BL/6J were distributed into three different groups, according to the assigned diet: control group fed a standard FA diet (2 mg FA/kg), FAD group for short term fed (0 mg FA/kg + 1% succinylsulfathiazole) from the day after mating until day 14.5 of gestation, and FAD group for long term fed the same FA-deficient diet for 6 weeks prior mating and continued with this diet during gestation. A total of 57 embryos (19 embryos of each dietary group) at 14.5 gestational days were evaluated. As indicators of changes in ocular biometry, we analyze two parameters: area and circularity of the lens and whole eye, and the area of the retina. The program used in the treatment and selection of the areas of interest was ImageJ. The statistical analysis was performed by IBM SPSS Statistics 19. RESULTS Regarding the measures of the area, FA-deficient lenses and eyes were smaller than that of controls. We have also observed increase in the size of the neural retina, spatially, in embryos from females fed FAD diet during long term. On the other hand, as regard to circularity measures, we have seen that eyes and lenses were more circular than control. CONCLUSION Maternal FAD diet for a very short term generates morphological changes in ocular structures to the offspring.
Collapse
Affiliation(s)
- Ouafa Sijilmassi
- a Faculty of Optics and Optometry, Anatomy and Human Embryology Department , Universidad Complutense De Madrid , Madrid , Spain.,b Faculty of Optics and Optometry, Optics Department , Universidad Complutense De Madrid , Madrid , Spain
| | - José Manuel López-Alonso
- b Faculty of Optics and Optometry, Optics Department , Universidad Complutense De Madrid , Madrid , Spain
| | - Aurora Del Río Sevilla
- a Faculty of Optics and Optometry, Anatomy and Human Embryology Department , Universidad Complutense De Madrid , Madrid , Spain
| | - Jorge Murillo González
- c Faculty of medicine, Anatomy and Human Embryology Department , Universidad Complutense De Madrid , Madrid , Spain
| | - María Del Carmen Barrio Asensio
- a Faculty of Optics and Optometry, Anatomy and Human Embryology Department , Universidad Complutense De Madrid , Madrid , Spain
| |
Collapse
|
45
|
Raiders SA, Eastwood MD, Bacher M, Priess JR. Binucleate germ cells in Caenorhabditis elegans are removed by physiological apoptosis. PLoS Genet 2018; 14:e1007417. [PMID: 30024879 PMCID: PMC6053125 DOI: 10.1371/journal.pgen.1007417] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/15/2018] [Indexed: 12/27/2022] Open
Abstract
Cell death plays a major role during C. elegans oogenesis, where over half of the oogenic germ cells die in a process termed physiological apoptosis. How germ cells are selected for physiological apoptosis, or instead become oocytes, is not understood. Most oocytes produce viable embryos when apoptosis is blocked, suggesting that physiological apoptosis does not function to cull defective germ cells. Instead, cells targeted for apoptosis may function as nurse cells; the germline is syncytial, and all germ cells appear to contribute cytoplasm to developing oocytes. C. elegans has been a leading model for the genetics and molecular biology of apoptosis and phagocytosis, but comparatively few studies have examined the cell biology of apoptotic cells. We used live imaging to identify and examine pre-apoptotic germ cells in the adult gonad. After initiating apoptosis, germ cells selectively export their mitochondria into the shared pool of syncytial cytoplasm; this transport appears to use the microtubule motor kinesin. The apoptotic cells then shrink as they expel most of their remaining cytoplasm, and close off from the syncytium. Shortly thereafter the apoptotic cells restructure their microtubule and actin cytoskeletons, possibly to maintain cell integrity; the microtubules form a novel, cortical array of stabilized microtubules, and actin and cofilin organize into giant cofilin-actin rods. We discovered that some apoptotic germ cells are binucleate; the binucleate germ cells can develop into binucleate oocytes in apoptosis-defective strains, and appear capable of producing triploid offspring. Our results suggest that the nuclear layer of the germline syncytium becomes folded during mitosis and growth, and that binucleate cells arise as the layer unfolds or everts; all of the binucleate cells are subsequently removed by apoptosis. These results show that physiological apoptosis targets at least two distinct populations of germ cells, and that the apoptosis machinery efficiently recognizes cells with two nuclei. Many germ cells die by apoptosis during the development of animal oocytes, including more than half of all germ cells in the model system C. elegans. How individual germ cells are selected for apoptosis, or survival, is not known. Here we study the cell biology of apoptosis. The C. elegans gonad is a syncytium, with nearly 1000 germ “cells” connected to a shared, core cytoplasm. Once apoptosis is initiated, germ cells selectively transport their mitochondria into the gonad core, apparently using the microtubule motor protein kinesin. The apoptotic cells next constrict, expelling most of their remaining cytoplasm into the core, and close off from the gonad core. The microtubule and actin cytoskeletons are remodeled and stabilized, presumably to maintain the integrity of the dying cell. The apoptotic cells form giant cofilin-actin rods, similar to rods described in stressed cultured cells and in human myopathies and neuropathies such as Alzheimer’s and Huntington’s disease. We show that some germ cells are binucleate; these cells appear to form during germline morphogenesis, and are removed by apoptosis. These results demonstrate heterogeneity between oogenic germ cells, and show that the apoptosis machinery efficiently recognizes and removes cells with two nuclei.
Collapse
Affiliation(s)
- Stephan A. Raiders
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Michael D. Eastwood
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Meghan Bacher
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - James R. Priess
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington, United States of America
- Department of Biology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
46
|
Vishal K, Bawa S, Brooks D, Bauman K, Geisbrecht ER. Thin is required for cell death in the Drosophila abdominal muscles by targeting DIAP1. Cell Death Dis 2018; 9:740. [PMID: 29970915 PMCID: PMC6030163 DOI: 10.1038/s41419-018-0756-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 05/28/2018] [Accepted: 05/30/2018] [Indexed: 12/23/2022]
Abstract
In holometabolous insects, developmentally controlled programmed cell death (PCD) is a conserved process that destroys a subset of larval tissues for the eventual creation of new adult structures. This process of histolysis is relatively well studied in salivary gland and midgut tissues, while knowledge concerning larval muscle destruction is limited. Here, we have examined the histolysis of a group of Drosophila larval abdominal muscles called the dorsal external oblique muscles (DEOMs). Previous studies have defined apoptosis as the primary mediator of DEOM breakdown, whose timing is controlled by ecdysone signaling. However, very little is known about other factors that contribute to DEOM destruction. In this paper, we examine the role of thin (tn), which encodes for the Drosophila homolog of mammalian TRIM32, in the regulation of DEOM histolysis. We find that loss of Tn blocks DEOM degradation independent of ecdysone signaling. Instead, tn genetically functions in a pathway with the death-associated inhibitor of apoptosis (DIAP1), Dronc, and death-associated APAF1-related killer (Dark) to regulate apoptosis. Importantly, blocking Tn results in the absence of active Caspase-3 immunostaining, upregulation of DIAP1 protein levels, and inhibition of Dronc activation. DIAP1 and Dronc mRNA levels are not altered in tn mutants, showing that Tn acts post-transcriptionally on DIAP1 to regulate apoptosis. Herein, we also find that the RING domain of Tn is required for DEOM histolysis as loss of this domain results in higher DIAP1 levels. Together, our results suggest that the direct control of DIAP1 levels, likely through the E3 ubiquitin ligase activity of Tn, provides a mechanism to regulate caspase activity and to facilitate muscle cell death.
Collapse
Affiliation(s)
- Kumar Vishal
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - Simranjot Bawa
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - David Brooks
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - Kenneth Bauman
- Department of Cell Biology and Biophysics, School of Biological Sciences, University of Missouri-Kansas, Kansas City, MO, 64110, USA
| | - Erika R Geisbrecht
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
47
|
Lee MK, Jung CS, Yoon JH, Lee N. Effects of resistance exercise on antioxidant enzyme activities and apoptosis-related protein expression of hippocampus in OLETF rats. Technol Health Care 2018; 26:457-467. [DOI: 10.3233/thc-181183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Min-Ki Lee
- Department of Physical Education, Kongju National University, Gongju-si, Chungnam 32588, Korea
| | - Chun-Seop Jung
- Department of Sports Science, Hannam University, Daejeon-si, Chungnam 34430, Korea
| | - Jin-Hwan Yoon
- Department of Sports Science, Hannam University, Daejeon-si, Chungnam 34430, Korea
| | - Namju Lee
- Department of Leisure Sports, School of Sports Science, Jungwon University, Goesan-gun, Chungbuk 28024, Korea
| |
Collapse
|
48
|
New 1,2,4-triazole-Chalcone hybrids induce Caspase-3 dependent apoptosis in A549 human lung adenocarcinoma cells. Eur J Med Chem 2018; 151:705-722. [DOI: 10.1016/j.ejmech.2018.03.073] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 03/23/2018] [Accepted: 03/24/2018] [Indexed: 11/30/2022]
|
49
|
Abstract
The proto-oncogene Myc is well known for its roles in promoting cell growth, proliferation and apoptosis. However, in this study, we found from a genetic screen that Myc inhibits, rather than promotes, cell death triggered by c-Jun N-terminal kinase (JNK) signaling in Drosophila. Firstly, expression of Drosophila Myc (dMyc) suppresses, whereas loss of dMyc enhances, ectopically activated JNK signaling-induced cell death. Secondly, dMyc impedes physiologically activated JNK pathway-mediated cell death. Thirdly, loss of dMyc triggers JNK pathway activation and JNK-dependent cell death. Finally, the mammalian cMyc gene, when expressed in Drosophila, impedes activated JNK signaling-induced cell death. Thus, besides its well-studied apoptosis promoting function, Myc also antagonizes JNK-mediated cell death in Drosophila, and this function is likely conserved from fly to human.
Collapse
Affiliation(s)
- Jiuhong Huang
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Yu Feng
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Xinhong Chen
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Wenzhe Li
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
| | - Lei Xue
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
| |
Collapse
|
50
|
Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, Annicchiarico-Petruzzelli M, Antonov AV, Arama E, Baehrecke EH, Barlev NA, Bazan NG, Bernassola F, Bertrand MJM, Bianchi K, Blagosklonny MV, Blomgren K, Borner C, Boya P, Brenner C, Campanella M, Candi E, Carmona-Gutierrez D, Cecconi F, Chan FKM, Chandel NS, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Cohen GM, Conrad M, Cubillos-Ruiz JR, Czabotar PE, D'Angiolella V, Dawson TM, Dawson VL, De Laurenzi V, De Maria R, Debatin KM, DeBerardinis RJ, Deshmukh M, Di Daniele N, Di Virgilio F, Dixit VM, Dixon SJ, Duckett CS, Dynlacht BD, El-Deiry WS, Elrod JW, Fimia GM, Fulda S, García-Sáez AJ, Garg AD, Garrido C, Gavathiotis E, Golstein P, Gottlieb E, Green DR, Greene LA, Gronemeyer H, Gross A, Hajnoczky G, Hardwick JM, Harris IS, Hengartner MO, Hetz C, Ichijo H, Jäättelä M, Joseph B, Jost PJ, Juin PP, Kaiser WJ, Karin M, Kaufmann T, Kepp O, Kimchi A, Kitsis RN, Klionsky DJ, Knight RA, Kumar S, Lee SW, Lemasters JJ, Levine B, Linkermann A, Lipton SA, Lockshin RA, López-Otín C, Lowe SW, Luedde T, Lugli E, MacFarlane M, Madeo F, Malewicz M, Malorni W, Manic G, Marine JC, Martin SJ, Martinou JC, Medema JP, Mehlen P, Meier P, Melino S, Miao EA, Molkentin JD, Moll UM, Muñoz-Pinedo C, Nagata S, Nuñez G, Oberst A, Oren M, Overholtzer M, Pagano M, Panaretakis T, Pasparakis M, Penninger JM, Pereira DM, Pervaiz S, Peter ME, Piacentini M, Pinton P, Prehn JHM, Puthalakath H, Rabinovich GA, Rehm M, Rizzuto R, Rodrigues CMP, Rubinsztein DC, Rudel T, Ryan KM, Sayan E, Scorrano L, Shao F, Shi Y, Silke J, Simon HU, Sistigu A, Stockwell BR, Strasser A, Szabadkai G, Tait SWG, Tang D, Tavernarakis N, Thorburn A, Tsujimoto Y, Turk B, Vanden Berghe T, Vandenabeele P, Vander Heiden MG, Villunger A, Virgin HW, Vousden KH, Vucic D, Wagner EF, Walczak H, Wallach D, Wang Y, Wells JA, Wood W, Yuan J, Zakeri Z, Zhivotovsky B, Zitvogel L, Melino G, Kroemer G. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ 2018; 25:486-541. [PMID: 29362479 PMCID: PMC5864239 DOI: 10.1038/s41418-017-0012-4] [Citation(s) in RCA: 4091] [Impact Index Per Article: 584.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 02/06/2023] Open
Abstract
Over the past decade, the Nomenclature Committee on Cell Death (NCCD) has formulated guidelines for the definition and interpretation of cell death from morphological, biochemical, and functional perspectives. Since the field continues to expand and novel mechanisms that orchestrate multiple cell death pathways are unveiled, we propose an updated classification of cell death subroutines focusing on mechanistic and essential (as opposed to correlative and dispensable) aspects of the process. As we provide molecularly oriented definitions of terms including intrinsic apoptosis, extrinsic apoptosis, mitochondrial permeability transition (MPT)-driven necrosis, necroptosis, ferroptosis, pyroptosis, parthanatos, entotic cell death, NETotic cell death, lysosome-dependent cell death, autophagy-dependent cell death, immunogenic cell death, cellular senescence, and mitotic catastrophe, we discuss the utility of neologisms that refer to highly specialized instances of these processes. The mission of the NCCD is to provide a widely accepted nomenclature on cell death in support of the continued development of the field.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Paris Descartes/Paris V University, Paris, France.
| | - Ilio Vitale
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Stuart A Aaronson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John M Abrams
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dieter Adam
- Institute of Immunology, Kiel University, Kiel, Germany
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Emad S Alnemri
- Department of Biochemistry and Molecular Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lucia Altucci
- Department of Biochemistry, Biophysics and General Pathology, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Ivano Amelio
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - David W Andrews
- Biological Sciences, Sunnybrook Research Institute, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | | | - Alexey V Antonov
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - Eli Arama
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Nickolai A Barlev
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - Nicolas G Bazan
- Neuroscience Center of Excellence, Louisiana State University School of Medicine, New Orleans, LA, USA
| | - Francesca Bernassola
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Mathieu J M Bertrand
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Katiuscia Bianchi
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | | | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, Albert Ludwigs University, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), Faculty of Medicine, Albert Ludwigs University, Freiburg, Germany
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Center for Biological Investigation (CIB), Spanish National Research Council (CSIC), Madrid, Spain
| | - Catherine Brenner
- INSERM U1180, Châtenay Malabry, France
- University of Paris Sud/Paris Saclay, Orsay, France
| | - Michelangelo Campanella
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, London, UK
- University College London Consortium for Mitochondrial Research, London, UK
| | - Eleonora Candi
- Biochemistry Laboratory, Dermopatic Institute of Immaculate (IDI) IRCCS, Rome, Italy
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | | | - Francesco Cecconi
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Unit of Cell Stress and Survival, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Francis K-M Chan
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Navdeep S Chandel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emily H Cheng
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jerry E Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John A Cidlowski
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC, USA
| | - Aaron Ciechanover
- Technion Integrated Cancer Center (TICC), The Ruth and Bruce Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | - Gerald M Cohen
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Marcus Conrad
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Juan R Cubillos-Ruiz
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Peter E Czabotar
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Vincenzo D'Angiolella
- Cancer Research UK and Medical Research Council Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vincenzo De Laurenzi
- Department of Medical, Oral and Biotechnological Sciences, CeSI-MetUniversity of Chieti-Pescara "G. d'Annunzio", Chieti, Italy
| | - Ruggero De Maria
- Institute of General Pathology, Catholic University "Sacro Cuore", Rome, Italy
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mohanish Deshmukh
- Department of Cell Biology and Physiology, Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - Nicola Di Daniele
- Hypertension and Nephrology Unit, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Vishva M Dixit
- Department of Physiological Chemistry, Genentech, South San Francisco, CA, USA
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Colin S Duckett
- Baylor Scott & White Research Institute, Baylor College of Medicine, Dallas, TX, USA
| | - Brian D Dynlacht
- Department of Pathology, New York University School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - John W Elrod
- Center for Translational Medicine, Department of Pharmacology, Lewis Katz School of Medicine at Temple University School of Medicine, Philadelphia, PA, USA
| | - Gian Maria Fimia
- National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani", Rome, Italy
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site, Frankfurt, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ana J García-Sáez
- Interfaculty Institute of Biochemistry, Tübingen University, Tübingen, Germany
| | - Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Lab, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Carmen Garrido
- INSERM U1231 "Lipides Nutrition Cancer", Dijon, France
- Faculty of Medicine, University of Burgundy France Comté, Dijon, France
- Cancer Centre Georges François Leclerc, Dijon, France
| | - Evripidis Gavathiotis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Pierre Golstein
- Immunology Center of Marseille-Luminy, Aix Marseille University, Marseille, France
| | - Eyal Gottlieb
- Technion Integrated Cancer Center (TICC), The Ruth and Bruce Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Douglas R Green
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Lloyd A Greene
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Hinrich Gronemeyer
- Team labeled "Ligue Contre le Cancer", Department of Functional Genomics and Cancer, Institute of Genetics and Molecular and Cellular Biology (IGBMC), Illkirch, France
- CNRS UMR 7104, Illkirch, France
- INSERM U964, Illkirch, France
- University of Strasbourg, Illkirch, France
| | - Atan Gross
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Gyorgy Hajnoczky
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - J Marie Hardwick
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Isaac S Harris
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Cellular and Molecular Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Hidenori Ichijo
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Marja Jäättelä
- Cell Death and Metabolism Unit, Center for Autophagy, Recycling and Disease, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Bertrand Joseph
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Philipp J Jost
- III Medical Department for Hematology and Oncology, Technical University Munich, Munich, Germany
| | - Philippe P Juin
- Team 8 "Stress adaptation and tumor escape", CRCINA-INSERM U1232, Nantes, France
- University of Nantes, Nantes, France
- University of Angers, Angers, France
- Institute of Cancer Research in Western France, Saint-Herblain, France
| | - William J Kaiser
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, TX, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, University of California San Diego, La Jolla, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Oliver Kepp
- Paris Descartes/Paris V University, Paris, France
- Faculty of Medicine, Paris Sud/Paris XI University, Kremlin-Bicêtre, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Campus, Villejuif, France
- Team 11 labeled "Ligue Nationale contre le Cancer", Cordeliers Research Center, Paris, France
- INSERM U1138, Paris, France
- Pierre et Marie Curie/Paris VI University, Paris, France
| | - Adi Kimchi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Richard N Kitsis
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Daniel J Klionsky
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Richard A Knight
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Sam W Lee
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - John J Lemasters
- Center for Cell Death, Injury and Regeneration, Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
- Center for Cell Death, Injury and Regeneration, Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Beth Levine
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andreas Linkermann
- Division of Nephrology, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Stuart A Lipton
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
- Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA, USA
| | - Richard A Lockshin
- Department of Biology, St. John's University, Queens, NY, USA
- Queens College of the City University of New York, Queens, NY, USA
| | - Carlos López-Otín
- Departament of Biochemistry and Molecular Biology, Faculty of Medicine, University Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Scott W Lowe
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tom Luedde
- Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Enrico Lugli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Humanitas Flow Cytometry Core, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Marion MacFarlane
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - Frank Madeo
- Department Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Michal Malewicz
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
| | - Walter Malorni
- National Centre for Gender Medicine, Italian National Institute of Health (ISS), Rome, Italy
| | - Gwenola Manic
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Seamus J Martin
- Departments of Genetics, Trinity College, University of Dublin, Dublin 2, Ireland
| | - Jean-Claude Martinou
- Department of Cell Biology, Faculty of Sciences, University of Geneva, Geneva, Switzerland
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
- Cancer Genomics Center, Amsterdam, The Netherlands
| | - Patrick Mehlen
- Apoptosis, Cancer and Development laboratory, CRCL, Lyon, France
- Team labeled "La Ligue contre le Cancer", Lyon, France
- LabEx DEVweCAN, Lyon, France
- INSERM U1052, Lyon, France
- CNRS UMR5286, Lyon, France
- Department of Translational Research and Innovation, Léon Bérard Cancer Center, Lyon, France
| | - Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Mary-Jean Mitchell Green Building, Chester Beatty Laboratories, London, UK
| | - Sonia Melino
- Department of Chemical Sciences and Technologies, University of Rome, Tor Vergata, Rome, Italy
| | - Edward A Miao
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC, USA
| | - Jeffery D Molkentin
- Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ute M Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Cristina Muñoz-Pinedo
- Cell Death Regulation Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Shigekazu Nagata
- Laboratory of Biochemistry and Immunology, World Premier International (WPI) Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| | - Gabriel Nuñez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
- Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Andrew Oberst
- Department of Immunology, University of Washington, Seattle, WA, USA
- Center for Innate Immunity and Immune Disease, Seattle, WA, USA
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute, Rehovot, Israel
| | - Michael Overholtzer
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michele Pagano
- Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York University School of Medicine, New York, NY, USA
| | - Theocharis Panaretakis
- Department of Genitourinary Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Manolis Pasparakis
- Institute for Genetics, Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Campus Vienna BioCentre, Vienna, Austria
| | - David M Pereira
- REQUIMTE/LAQV, Laboratory of Pharmacognosy, Department of Chemistry, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
- National University Cancer Institute, National University Health System (NUHS), Singapore, Singapore
| | - Marcus E Peter
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mauro Piacentini
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
- National Institute for Infectious Diseases IRCCS "Lazzaro Spallanzani", Rome, Italy
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
- LTTA center, University of Ferrara, Ferrara, Italy
- Maria Cecilia Hospital, GVM Care & Research, Health Science Foundation, Cotignola, Italy
| | - Jochen H M Prehn
- Department of Physiology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Hamsa Puthalakath
- Department of Biochemistry, La Trobe University, Victoria, Australia
| | - Gabriel A Rabinovich
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine (IBYME), National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
- Department of Biological Chemistry, Faculty of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, Stuttgart, Germany
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Cecilia M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research (CIMR), University of Cambridge, Cambridge, UK
| | - Thomas Rudel
- Department of Microbiology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Kevin M Ryan
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Emre Sayan
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Luca Scorrano
- Department of Biology, University of Padua, Padua, Italy
- Venetian Institute of Molecular Medicine, Padua, Italy
| | - Feng Shao
- National Institute of Biological Sciences, Beijing, China
| | - Yufang Shi
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Chinese Academy of Sciences, Shanghai, China
- Jiangsu Key Laboratory of Stem Cells and Medicinal Biomaterials, Institutes for Translational Medicine, Soochow University, Suzhou, China
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - John Silke
- Department of Medical Biology, The University of Melbourne, Melbourne, Victoria, Australia
- Division of Inflammation, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Antonella Sistigu
- Institute of General Pathology, Catholic University "Sacro Cuore", Rome, Italy
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Chemistry, Columbia University, New York, NY, USA
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Gyorgy Szabadkai
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Department of Cell and Developmental Biology, University College London Consortium for Mitochondrial Research, London, UK
- Francis Crick Institute, London, UK
| | | | - Daolin Tang
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
- Center for DAMP Biology, Guangzhou Medical University, Guangzhou, Guangdong, China
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou Medical University, Guangzhou, Guangdong, China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou, Guangdong, China
- Key Laboratory for Protein Modification and Degradation of Guangdong Province, Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas Medical School, University of Crete, Heraklion, Greece
| | - Andrew Thorburn
- Department of Pharmacology, University of Colorado, Aurora, CO, USA
| | | | - Boris Turk
- Department Biochemistry and Molecular Biology, "Jozef Stefan" Institute, Ljubljana, Slovenia
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Tom Vanden Berghe
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- VIB Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Andreas Villunger
- Division of Developmental Immunology, Innsbruck Medical University, Innsbruck, Austria
| | - Herbert W Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Domagoj Vucic
- Department of Early Discovery Biochemistry, Genentech, South San Francisco, CA, USA
| | - Erwin F Wagner
- Genes, Development and Disease Group, Cancer Cell Biology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, London, UK
| | - David Wallach
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Ying Wang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Will Wood
- School of Cellular and Molecular Medicine, Faculty of Biomedical Sciences, University of Bristol, Bristol, UK
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zahra Zakeri
- Department of Biology, Queens College of the City University of New York, Queens, NY, USA
| | - Boris Zhivotovsky
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Laurence Zitvogel
- Faculty of Medicine, Paris Sud/Paris XI University, Kremlin-Bicêtre, France
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
| | - Gerry Melino
- Medical Research Council (MRC) Toxicology Unit, Leicester University, Leicester, UK
- Department of Experimental Medicine and Surgery, University of Rome "Tor Vergata", Rome, Italy
| | - Guido Kroemer
- Paris Descartes/Paris V University, Paris, France.
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden.
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Campus, Villejuif, France.
- Team 11 labeled "Ligue Nationale contre le Cancer", Cordeliers Research Center, Paris, France.
- INSERM U1138, Paris, France.
- Pierre et Marie Curie/Paris VI University, Paris, France.
- Biology Pole, European Hospital George Pompidou, AP-HP, Paris, France.
| |
Collapse
|