1
|
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
|
2
|
Luobin L, Wanxin H, Yingxin G, Qinzhou Z, Zefeng L, Danyang W, Huaqin L. Nanomedicine-induced programmed cell death in cancer therapy: mechanisms and perspectives. Cell Death Discov 2024; 10:386. [PMID: 39209834 PMCID: PMC11362291 DOI: 10.1038/s41420-024-02121-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/20/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The balance of programmed cell death (PCD) mechanisms, including apoptosis, autophagy, necroptosis and others, is pivotal in cancer progression and treatment. Dysregulation of these pathways results in uncontrolled cell growth and resistance to conventional therapies. Nanomedicine offers a promising solution in oncology through targeted drug delivery enabling precise targeting of cancer cells while preserving healthy tissues. This approach reduces the side effects of traditional chemotherapy and enhances treatment efficacy by engaging PCD pathways. We details each PCD pathway, their mechanisms, and innovative nanomedicine strategies to activate these pathways, thereby enhancing therapeutic specificity and minimizing harm to healthy tissues. The precision of nanotechnology in targeting PCD pathways promises significant improvements in cancer treatment outcomes. This synergy between nanotechnology and targeted PCD activation could lead to more effective and less toxic cancer therapies, heralding a new era in cancer treatment.
Collapse
Affiliation(s)
- Lin Luobin
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - He Wanxin
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China
| | - Guo Yingxin
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China
| | - Zheng Qinzhou
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Liang Zefeng
- School of Life Sciences and Biopharmaceuticals, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Wu Danyang
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China
| | - Li Huaqin
- School of Health Sciences, Guangzhou Xinhua University, 19 Huamei Road, Tianhe District, Guangzhou, 510520, China.
| |
Collapse
|
3
|
Nagavath R, Thupurani MK, Badithapuram V, Manchal R, Vasam CS, Thirukovela NS. Organo NHC catalyzed aqueous synthesis of 4β-isoxazole-podophyllotoxins: in vitro anticancer, caspase activation, tubulin polymerization inhibition and molecular docking studies. RSC Adv 2024; 14:23574-23582. [PMID: 39070249 PMCID: PMC11276401 DOI: 10.1039/d4ra04297b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
We present, for the first time, the organo-N-heterocyclic carbene (NHC) catalyzed 1,3-dipolar cycloaddition of 4β-O-propargyl podophyllotoxin (1) with in situ aromatic nitrile oxides to afford regioselective 4β-isoxazolepodophyllotoxin hybrids (6a-n) in benign aqueous-organic media. Preliminary anticancer activity results showed that compound 6e displayed superior activity against MCF-7, HeLa and MIA PaCa2 human cell lines compared with podophyllotoxin. Compounds 6j and 6n showed greater activity against the MCF-7 cell line than the positive control. Caspase activation studies revealed that compound 6e at 20 μg ml-1 concentration had greater caspase 3/7 activation in MCF-7 and MIAPaCa2 cells than podophyllotoxin. Furthermore, in vitro tubulin polymerization inhibition studies revealed that compound 6e showed comparable activity with podophyllotoxin. Finally, in silico molecular docking studies of compounds 6e, 6j, 6n and podophyllotoxin on α,β-tubulin (pdb id 1SA0) revealed that compound 6n showed excellent binding energies and inhibition constants compared with podophyllotoxin.
Collapse
Affiliation(s)
- Rajkumar Nagavath
- Department of Chemistry, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| | - Murali Krishna Thupurani
- Department of Biotechnology, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| | - Vinitha Badithapuram
- Department of Chemistry, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| | - Ravinder Manchal
- Department of Chemistry, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| | | | - Narasimha Swamy Thirukovela
- Department of Chemistry, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| |
Collapse
|
4
|
Guerrache A, Micheau O. TNF-Related Apoptosis-Inducing Ligand: Non-Apoptotic Signalling. Cells 2024; 13:521. [PMID: 38534365 PMCID: PMC10968836 DOI: 10.3390/cells13060521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/01/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL or Apo2 or TNFSF10) belongs to the TNF superfamily. When bound to its agonistic receptors, TRAIL can induce apoptosis in tumour cells, while sparing healthy cells. Over the last three decades, this tumour selectivity has prompted many studies aiming at evaluating the anti-tumoral potential of TRAIL or its derivatives. Although most of these attempts have failed, so far, novel formulations are still being evaluated. However, emerging evidence indicates that TRAIL can also trigger a non-canonical signal transduction pathway that is likely to be detrimental for its use in oncology. Likewise, an increasing number of studies suggest that in some circumstances TRAIL can induce, via Death receptor 5 (DR5), tumour cell motility, potentially leading to and contributing to tumour metastasis. While the pro-apoptotic signal transduction machinery of TRAIL is well known from a mechanistic point of view, that of the non-canonical pathway is less understood. In this study, we the current state of knowledge of TRAIL non-canonical signalling.
Collapse
Affiliation(s)
- Abderrahmane Guerrache
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
| | - Olivier Micheau
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
- Laboratoire d’Excellence LipSTIC, 21000 Dijon, France
| |
Collapse
|
5
|
Kandouz M. Cell Death, by Any Other Name…. Cells 2024; 13:325. [PMID: 38391938 PMCID: PMC10886887 DOI: 10.3390/cells13040325] [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: 12/31/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Studies trying to understand cell death, this ultimate biological process, can be traced back to a century ago. Yet, unlike many other fashionable research interests, research on cell death is more alive than ever. New modes of cell death are discovered in specific contexts, as are new molecular pathways. But what is "cell death", really? This question has not found a definitive answer yet. Nevertheless, part of the answer is irreversibility, whereby cells can no longer recover from stress or injury. Here, we identify the most distinctive features of different modes of cell death, focusing on the executive final stages. In addition to the final stages, these modes can differ in their triggering stimulus, thus referring to the initial stages. Within this framework, we use a few illustrative examples to examine how intercellular communication factors in the demise of cells. First, we discuss the interplay between cell-cell communication and cell death during a few steps in the early development of multicellular organisms. Next, we will discuss this interplay in a fully developed and functional tissue, the gut, which is among the most rapidly renewing tissues in the body and, therefore, makes extensive use of cell death. Furthermore, we will discuss how the balance between cell death and communication is modified during a pathological condition, i.e., colon tumorigenesis, and how it could shed light on resistance to cancer therapy. Finally, we briefly review data on the role of cell-cell communication modes in the propagation of cell death signals and how this has been considered as a potential therapeutic approach. Far from vainly trying to provide a comprehensive review, we launch an invitation to ponder over the significance of cell death diversity and how it provides multiple opportunities for the contribution of various modes of intercellular communication.
Collapse
Affiliation(s)
- Mustapha Kandouz
- Department of Pathology, School of Medicine, Wayne State University, 540 East Canfield Avenue, Detroit, MI 48201, USA;
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
6
|
Bienvenu AL, Ballut L, Picot S. Specifically Targeting Metacaspases of Candida: A New Therapeutic Opportunity. J Fungi (Basel) 2024; 10:90. [PMID: 38392762 PMCID: PMC10889698 DOI: 10.3390/jof10020090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
The World Health Organization (WHO) recently published a list of fungal priority pathogens, including Candida albicans and C. auris. The increased level of resistance of Candida is raising concern, considering the availability of only four classes of medicine. The WHO is seeking novel agent classes with different targets and mechanisms of action. Targeting Candida metacaspases to control intrinsic cell death could provide new therapeutic opportunities for invasive candidiasis. In this review, we provide the available evidence for Candida cell death, describe Candida metacaspases, and discuss the potential of Candida metacaspases to offer a new specific target. Targeting Candida cell death has good scientific rationale given that the fungicidal activity of many marketed antifungals is mediated, among others, by cell death triggering. But none of the available antifungals are specifically activating Candida metacaspases, making this target a new therapeutic opportunity for non-susceptible isolates. It is expected that antifungals based on the activation of fungi metacaspases will have a broad spectrum of action, as metacaspases have been described in many fungi, including filamentous fungi. Considering this original mechanism of action, it could be of great interest to combine these new antifungal candidates with existing antifungals. This approach would help to avoid the development of antifungal resistance, which is especially increasing in Candida.
Collapse
Affiliation(s)
- Anne-Lise Bienvenu
- Service Pharmacie, Groupement Hospitalier Nord, Hospices Civils de Lyon, 69004 Lyon, France
- Malaria Research Unit, University Lyon, UMR 5246 CNRS-INSA-CPE-University Lyon1, 69100 Villeurbanne, France
| | - Lionel Ballut
- Molecular Microbiology and Structural Biochemistry, UMR 5086, CNRS-Université de Lyon, 69367 Lyon, France
| | - Stephane Picot
- Malaria Research Unit, University Lyon, UMR 5246 CNRS-INSA-CPE-University Lyon1, 69100 Villeurbanne, France
- Institute of Parasitology and Medical Mycology, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, 69004 Lyon, France
| |
Collapse
|
7
|
Kubickova A, De Sanctis JB, Hajduch M. Isoform-Directed Control of c-Myc Functions: Understanding the Balance from Proliferation to Growth Arrest. Int J Mol Sci 2023; 24:17524. [PMID: 38139353 PMCID: PMC10743581 DOI: 10.3390/ijms242417524] [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: 10/17/2023] [Revised: 12/07/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
The transcription factor c-Myc, a key regulator of cellular processes, has long been associated with roles in cell proliferation and apoptosis. This review analyses the multiple functions of c-Myc by examining the different c-Myc isoforms in detail. The impact of different c-Myc isoforms, in particular p64 and p67, on fundamental biological processes remains controversial. It is necessary to investigate the different isoforms in the context of proto-oncogenesis. The current knowledge base suggests that neoplastic lesions may possess the means for self-destruction via increased c-Myc activity. This review presents the most relevant information on the c-Myc locus and focuses on a number of isoforms, including p64 and p67. This compilation provides a basis for the development of therapeutic approaches that target the potent growth arresting and pro-apoptotic functions of c-Myc. This information can then be used to develop targeted interventions against specific isoforms with the aim of shifting the oncogenic effects of c-Myc from pro-proliferative to pro-apoptotic. The research summarised in this review can deepen our understanding of how c-Myc activity contributes to different cellular responses, which will be crucial in developing effective therapeutic strategies; for example, isoform-specific approaches may allow for precise modulation of c-Myc function.
Collapse
Affiliation(s)
- Agata Kubickova
- Institute of Molecular and Translational Medicine, Palacky University and University Hospital Olomouc, Hnevotinska 1333/5, 77900 Olomouc, Czech Republic; (A.K.); (J.B.D.S.)
- Institute of Molecular and Translational Medicine, Czech Advanced Technology and Research Institute, Palacky University in Olomouc, Hnevotinska 1333/5, 77900 Olomouc, Czech Republic
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Palacky University and University Hospital Olomouc, Hnevotinska 1333/5, 77900 Olomouc, Czech Republic; (A.K.); (J.B.D.S.)
- Institute of Molecular and Translational Medicine, Czech Advanced Technology and Research Institute, Palacky University in Olomouc, Hnevotinska 1333/5, 77900 Olomouc, Czech Republic
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Palacky University and University Hospital Olomouc, Hnevotinska 1333/5, 77900 Olomouc, Czech Republic; (A.K.); (J.B.D.S.)
- Institute of Molecular and Translational Medicine, Czech Advanced Technology and Research Institute, Palacky University in Olomouc, Hnevotinska 1333/5, 77900 Olomouc, Czech Republic
| |
Collapse
|
8
|
Nafchi NAM, Chilcott EM, Brown S, Fuller HR, Bowerman M, Yáñez-Muñoz RJ. Enhanced expression of the human Survival motor neuron 1 gene from a codon-optimised cDNA transgene in vitro and in vivo. Gene Ther 2023; 30:812-825. [PMID: 37322133 DOI: 10.1038/s41434-023-00406-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 04/14/2023] [Accepted: 05/04/2023] [Indexed: 06/17/2023]
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disease particularly characterised by degeneration of ventral motor neurons. Survival motor neuron (SMN) 1 gene mutations cause SMA, and gene addition strategies to replace the faulty SMN1 copy are a therapeutic option. We have developed a novel, codon-optimised hSMN1 transgene and produced integration-proficient and integration-deficient lentiviral vectors with cytomegalovirus (CMV), human synapsin (hSYN) or human phosphoglycerate kinase (hPGK) promoters to determine the optimal expression cassette configuration. Integrating, CMV-driven and codon-optimised hSMN1 lentiviral vectors resulted in the highest production of functional SMN protein in vitro. Integration-deficient lentiviral vectors also led to significant expression of the optimised transgene and are expected to be safer than integrating vectors. Lentiviral delivery in culture led to activation of the DNA damage response, in particular elevating levels of phosphorylated ataxia telangiectasia mutated (pATM) and γH2AX, but the optimised hSMN1 transgene showed some protective effects. Neonatal delivery of adeno-associated viral vector (AAV9) vector encoding the optimised transgene to the Smn2B/- mouse model of SMA resulted in a significant increase of SMN protein levels in liver and spinal cord. This work shows the potential of a novel codon-optimised hSMN1 transgene as a therapeutic strategy for SMA.
Collapse
Affiliation(s)
- Neda A M Nafchi
- AGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, TW20 0EX, UK
| | - Ellie M Chilcott
- AGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, TW20 0EX, UK
| | - Sharon Brown
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry, SY10 7AG, UK
| | - Heidi R Fuller
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry, SY10 7AG, UK
| | - Melissa Bowerman
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry, SY10 7AG, UK
- School of Medicine, Keele University, Staffordshire, ST5 5BG, UK
| | - Rafael J Yáñez-Muñoz
- AGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, TW20 0EX, UK.
| |
Collapse
|
9
|
Kim HY, Kang HG, Kim HM, Jeong HJ. Anti-tumor activity of trimethoprim-sulfamethoxazole against melanoma skin cancer through triggering allergic reaction and promoting immunity. Int Immunopharmacol 2023; 123:110742. [PMID: 37536185 DOI: 10.1016/j.intimp.2023.110742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/20/2023] [Accepted: 07/28/2023] [Indexed: 08/05/2023]
Abstract
The anti-cancer impact of an allergic reaction is strongly linked to immunity enhancement. Trimethoprim-sulfamethoxazole (TMP-SMX), an antibiotic, has potential immunomodulatory effects, but has side effects such as allergies. Thus far, the effects and underlying mechanisms of TMP-SMX in melanoma have not been clarified. This study examined the potential roles of TMP-SMX in melanoma skin cancer using an immunodeficient mouse model. TMP-SMX significantly improved the survival rate and reduced the tumor weight and growth and vascular endothelial growth factor levels in melanoma skin cancer of immunodeficient mice. In the forced swimming test, TMP-SMX significantly reduced immobility time compared to the melanoma skin cancer of immunodeficient mice, indicating improved immunity. TMP-SMX significantly increased infiltration of mast cells and release of allergy-related mediators (IgE, histamine, interleukin (IL)-4, IL-5, IL-13, and IL-33) and immune-enhancing mediators (tumor necrosis factor-α, IL-2, IL-6, and IL-12). In addition, the administration of TMP-SMX significantly increased the caspase-3, 8, and 9 activities. Furthermore, mice given TMP-SMX showed no adverse reactions according to the blood biochemical parameters. TMP-SMX significantly inhibits the growth of melanoma skin cancer by triggering an allergic reaction and promotingimmunity. Hence, we propose that TMP-SMX may be used as an immune booster in cancer chemotherapy.
Collapse
Affiliation(s)
- Hee-Yun Kim
- Biochip Research Center, Hoseo University, Asan, Chungnam 31499, Republic of Korea
| | - Ho-Geun Kang
- Department of Bio-Convergence System, Graduate School, Hoseo University, Asan 31499, Republic of Korea
| | - Hyung-Min Kim
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Hyun-Ja Jeong
- Biochip Research Center, Hoseo University, Asan, Chungnam 31499, Republic of Korea; Department of Bio-Convergence System, Graduate School, Hoseo University, Asan 31499, Republic of Korea; Department of Food Science & Technology, Hoseo University, 20, Hoseo-ro 79beon-gil, Baebang-eup, Asan, Chungnam 31499, Republic of Korea.
| |
Collapse
|
10
|
Antitumor and antimicrobial effect of syringic acid urea cocrystal: Structural and spectroscopic characterization, DFT calculation and biological evaluation. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
11
|
Sakthivel D, Brown-Suedel A, Bouchier-Hayes L. The role of the nucleolus in regulating the cell cycle and the DNA damage response. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 135:203-241. [PMID: 37061332 DOI: 10.1016/bs.apcsb.2023.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
The nucleolus has long been perceived as the site for ribosome biogenesis, but numerous studies suggest that the nucleolus carefully sequesters crucial proteins involved in multiple cellular functions. Among these, the role of nucleolus in cell cycle regulation is the most evident. The nucleolus is the first responder of growth-related signals to mediate normal cell cycle progression. The nucleolus also senses different cellular stress insults by activating diverse pathways that arrest the cell cycle, promote DNA repair, or initiate apoptosis. Here, we review the emerging concepts on how the ribosomal and nonribosomal nucleolar proteins mediate such cellular effects.
Collapse
|
12
|
Barteneva NS, Meirkhanova A, Malashenkov D, Vorobjev IA. To Die or Not to Die-Regulated Cell Death and Survival in Cyanobacteria. Microorganisms 2022; 10:1657. [PMID: 36014075 PMCID: PMC9415839 DOI: 10.3390/microorganisms10081657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/06/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
Regulated cell death (RCD) is central to the development, integrity, and functionality of multicellular organisms. In the last decade, evidence has accumulated that RCD is a universal phenomenon in all life domains. Cyanobacteria are of specific interest due to their importance in aquatic and terrestrial habitats and their role as primary producers in global nutrient cycling. Current knowledge on cyanobacterial RCD is based mainly on biochemical and morphological observations, often by methods directly transferred from vertebrate research and with limited understanding of the molecular genetic basis. However, the metabolism of different cyanobacteria groups relies on photosynthesis and nitrogen fixation, whereas mitochondria are the central executioner of cell death in vertebrates. Moreover, cyanobacteria chosen as biological models in RCD studies are mainly colonial or filamentous multicellular organisms. On the other hand, unicellular cyanobacteria have regulated programs of cellular survival (RCS) such as chlorosis and post-chlorosis resuscitation. The co-existence of different genetically regulated programs in cyanobacterial populations may have been a top engine in life diversification. Development of cyanobacteria-specific methods for identification and characterization of RCD and wider use of single-cell analysis combined with intelligent image-based cell sorting and metagenomics would shed more light on the underlying molecular mechanisms and help us to address the complex colonial interactions during these events. In this review, we focus on the functional implications of RCD in cyanobacterial communities.
Collapse
Affiliation(s)
- Natasha S. Barteneva
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 000010, Kazakhstan
| | | | | | | |
Collapse
|
13
|
Erfanparast L, Taghizadieh M, Shekarchi AA. Non-Coding RNAs and Oral Cancer: Small Molecules With Big Functions. Front Oncol 2022; 12:914593. [PMID: 35898889 PMCID: PMC9309727 DOI: 10.3389/fonc.2022.914593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/16/2022] [Indexed: 12/24/2022] Open
Abstract
Oral cancer remains a major public concern with considerable socioeconomic impact in the world. Despite substantial advancements have been made in treating oral cancer, the five-year survival rate for oral cancer remained undesirable, and the molecular mechanisms underlying OSCC carcinogenesis have not been fully understood. Noncoding RNAs (ncRNAs) include transfer RNAs (tRNAs), as well as small RNAs such as microRNAs, and the long ncRNAs such as HOTAIR are a large segment of the transcriptome that do not have apparent protein-coding roles, but they have been verified to play important roles in diverse biological processes, including cancer cell development. Cell death, such as apoptosis, necrosis, and autophagy, plays a vital role in the progression of cancer. A better understanding of the regulatory relationships between ncRNAs and these various types of cancer cell death is therefore urgently required. The occurrence and development of oral cancer can be controlled by increasing or decreasing the expression of ncRNAs, a method which confers broad prospects for oral cancer treatment. Therefore, it is urgent for us to understand the influence of ncRNAs on the development of different modes of oral tumor death, and to evaluate whether ncRNAs have the potential to be used as biological targets for inducing cell death and recurrence of chemotherapy. The purpose of this review is to describe the impact of ncRNAs on cell apoptosis and autophagy in oral cancer in order to explore potential targets for oral cancer therapy.
Collapse
Affiliation(s)
- Leila Erfanparast
- Department of Pediatric Dentistry, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Taghizadieh
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- *Correspondence: Mohammad Taghizadieh,
| | - Ali Akbar Shekarchi
- Department of Pathology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Effect of Fumonisin B1 on Proliferation and Apoptosis of Intestinal Porcine Epithelial Cells. Toxins (Basel) 2022; 14:toxins14070471. [PMID: 35878209 PMCID: PMC9323054 DOI: 10.3390/toxins14070471] [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: 06/17/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 11/30/2022] Open
Abstract
Fumonisin B1 (FB1), which is a mycotoxin produced by Fusarium moniliforme and Fusarium rotarum, has a number of toxic effects in animals. Moldy feed containing FB1 can damage the intestine. In this study, we used intestinal porcine epithelial cells (IPEC-J2) as an in vitro model to explore the effects of FB1 on cell cycle and apoptosis. The results showed that IPEC-J2 cells treated with 10, 20, and 40 μg/mL FB1 for 48 h experienced different degrees of damage manifested as decreases in cell number and viability, as well as cell shrinkage and floating. In addition, FB1 reduced cell proliferation and the mRNA and protein expression of proliferating cell nuclear antigen (PCNA), cyclin-dependent kinase 2 (CDK2), CDK4, cyclinD1, and cyclinE1. FB1 blocked the cell cycle in the G1 phase. FB1 also induced mitochondrial pathway apoptosis, reduced mitochondrial membrane potential, and promoted mRNA and protein expression of Caspase3, Caspase9, and Bax. The findings suggest that FB1 can induce IPEC-J2 cell damage, block the cell cycle, and promote cell apoptosis.
Collapse
|
15
|
Lopez KE, Bouchier-Hayes L. Lethal and Non-Lethal Functions of Caspases in the DNA Damage Response. Cells 2022; 11:cells11121887. [PMID: 35741016 PMCID: PMC9221191 DOI: 10.3390/cells11121887] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/12/2022] Open
Abstract
Members of the caspase family are well known for their roles in the initiation and execution of cell death. Due to their function in the removal of damaged cells that could otherwise become malignant, caspases are important players in the DNA damage response (DDR), a network of pathways that prevent genomic instability. However, emerging evidence of caspases positively or negatively impacting the accumulation of DNA damage in the absence of cell death demonstrates that caspases play a role in the DDR that is independent of their role in apoptosis. This review highlights the apoptotic and non-apoptotic roles of caspases in the DDR and how they can impact genomic stability and cancer treatment.
Collapse
Affiliation(s)
- Karla E. Lopez
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- William T. Shearer Center for Human Immunobiology, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Lisa Bouchier-Hayes
- Department of Pediatrics, Division of Hematology-Oncology, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- William T. Shearer Center for Human Immunobiology, Texas Children’s Hospital, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
16
|
Silva LJ, Silva CR, Sá LG, Barroso FD, Cândido TM, Queiroz HA, Almeida Moreira LE, Baccallini OV, Cavalcanti BC, Silva J, Marinho ES, Moraes MO, Neto JB, Júnior HV. Antifungal activity of dexamethasone against fluconazole-resistant Candida albicans and its activity against biofilms. Future Microbiol 2022; 17:607-620. [PMID: 35411812 DOI: 10.2217/fmb-2021-0146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Objective: The present study investigated the antifungal action of dexamethasone disodium phosphate (Dex). Methodology: Susceptibility testing was performed using the Clinical & Laboratory Standards Institute protocol; M27-A3, checkerboard test and biofilm were evaluated with two isolates of Candida albicans, hyphal production test, molecular docking analysis and flow cytometry analysis. Result: Dex and fluconazole (FLC) together had a synergistic effect. Mature biofilm was reduced when treated with Dex alone or in combination. Dex and FLC promoted a decrease in the production of hyphae and changes in the level of mitochondrial depolarization, increased generation of reactive oxygen species, loss of membrane integrity, increased phosphatidylserine externalization and molecular docking; there was interaction with ALS3 and SAP5 targets. Conclusion: Dex showed antifungal activity against FLC-resistant C. albicans strains.
Collapse
Affiliation(s)
- Lisandra J Silva
- School of Pharmacy, Laboratory for Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, Ceará, Brazil
| | - Cecília R Silva
- School of Pharmacy, Laboratory for Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, Ceará, Brazil
| | - Lívia Gav Sá
- School of Pharmacy, Laboratory for Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Department of Chemistry, Group for Theoretical Chemistry & Electrochemistry (GQTE), State University of Ceará, Limoeiro do Norte, Ceará, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, Ceará, Brazil
| | - Fatima Dd Barroso
- School of Pharmacy, Laboratory for Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, Ceará, Brazil
| | - Thiago M Cândido
- School of Pharmacy, Laboratory for Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, Ceará, Brazil
| | - Helaine A Queiroz
- School of Pharmacy, Laboratory for Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, Ceará, Brazil
| | - Lara E Almeida Moreira
- School of Pharmacy, Laboratory for Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, Ceará, Brazil
| | - Octavio V Baccallini
- School of Pharmacy, Laboratory for Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, Ceará, Brazil
| | | | - Jacilene Silva
- Christus University Center (UNICHRISTUS), Fortaleza, Ceará, Brazil
| | | | - Manoel O Moraes
- Christus University Center (UNICHRISTUS), Fortaleza, Ceará, Brazil
| | - João Ba Neto
- School of Pharmacy, Laboratory for Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Department of Chemistry, Group for Theoretical Chemistry & Electrochemistry (GQTE), State University of Ceará, Limoeiro do Norte, Ceará, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, Ceará, Brazil
| | - Hélio Vn Júnior
- School of Pharmacy, Laboratory for Bioprospection of Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, Brazil.,Christus University Center (UNICHRISTUS), Fortaleza, Ceará, Brazil
| |
Collapse
|
17
|
Qian L, Liu J, Ma W, Liu Y, Wang X, Liu D. Hydrogen-Rich Water Ameliorates Murine Chronic Graft-versus-Host Disease through Antioxidation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1165928. [PMID: 34691352 PMCID: PMC8531770 DOI: 10.1155/2021/1165928] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 09/25/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an important treatment option for various hematopoietic diseases and certain hereditary diseases. Chronic graft-versus-host disease (cGVHD) has become the main life-threatening complication and cause of death in later stage postallo-HSCT. Current treatment options for cGVHD are limited. Hydrogen gas (H2) has been demonstrated that has antioxidative, anti-inflammatory, and antifibrosis effects. The aim of this study was to confirm whether oral administration hydrogen-rich water exerted therapeutic effects on a scleroderma cGVHD mouse model and tried to explain the mechanism underly it. METHODS A mouse cGVHD model was established by haploidentical bone marrow transplantation. To evaluate therapeutic effects of H2 on cGVHD, survival rate, changes in clinical scores, and skin pathologic characteristics of cGVHD mice were observed. To evaluate its therapeutic mechanism, we detected the expression levels of antioxidative enzymes heme oxygenase-1(HO-1) and NAD (P)H: quinone acceptor oxidoreductase 1(NQO1) in skin homogenates. We also detected the expression level of the apoptotic protein caspase-3 in skin homogenates. RESULTS 1-month survival rate of cGVHD mice in the hydrogen group reached 93.3%, significantly higher than 66.7% in the nonhydrogen group (p < 0.05). Clinical score of cGVHD mice was improved by hydrogen-rich water at 96 days posttransplantation (2.2 versus 4.5, p < 0.05). The skin pathological condition of cGVHD mice was significantly improved by hydrogen-rich water. At 96 days posttransplantation, average skin pathological hematoxylin and eosin (HE) staining score in the hydrogen group was 1.05, which was significantly lower than 3.2 in the nonhydrogen group (p < 0.01). Average Masson staining score was 0.6 point in the hydrogen group, lower than 0.9 point in the nonhydrogen group (p < 0.05). Both the relative expression levels of HO-1 and NQO1 proteins in skin specimens of cGVHD mice in the hydrogen group were lower than that in the nonhydrogen group (2.47 versus 6.21 and 1.83 versus 3.59, p < 0.05). The relative expression level of caspase-3 protein in skin specimens of cGVHD mice increased to 7.17 on the 96th day after transplantation, significantly higher than 4.36 in the hydrogen group. CONCLUSION In this study, we found that oral hydrogen-rich water improved the survival rate and clinical symptoms of cGVHD mice by antioxidant and antiapoptosis. This study would pave the way for further clinical study, which may provide a new treatment option for cGVHD.
Collapse
Affiliation(s)
- Liren Qian
- Senior Department of Hematology, The Fifth Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Jiaxin Liu
- Senior Department of Hematology, The Fifth Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Weina Ma
- Senior Department of Hematology, The Fifth Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Yu Liu
- Senior Department of Hematology, The Fifth Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Xiaona Wang
- Senior Department of Hematology, The Fifth Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Daihong Liu
- Senior Department of Hematology, The Fifth Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| |
Collapse
|
18
|
Chang SN, Khan I, Kim CG, Park SM, Choi DK, Lee H, Hwang BS, Kang SC, Park JG. Decursinol Angelate Arrest Melanoma Cell Proliferation by Initiating Cell Death and Tumor Shrinkage via Induction of Apoptosis. Int J Mol Sci 2021; 22:4096. [PMID: 33921050 PMCID: PMC8071397 DOI: 10.3390/ijms22084096] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Melanoma is known to aggressively metastasize and is one of the prominent causes of skin cancer mortality. This study was designed to assess the molecular mechanism of decursinol angelate (DA) against murine melanoma cell line (B16F10 cells). Treatment of DA resulted in growth inhibition and cell cycle arrest at G0/G1 (p < 0.001) phase, evaluated through immunoblotting. Moreover, autophagy-related proteins such as ATG-5 (p < 0.0001), ATG-7 (p < 0.0001), beclin-1 (p < 0.0001) and transition of LC3-I to LC3-II (p < 0.0001) were markedly decreased, indicating autophagosome inhibition. Additionally, DA treatment triggered apoptotic events which were corroborated by the occurrence of distorted nuclei, elevated reactive oxygen species (ROS) levels and reduction in the mitochondrial membrane potential. Subsequently, there was an increase in the expression of pro-apoptotic protein Bax in a dose-dependent manner, with the corresponding downregulation of Bcl-2 expression and cytochrome C expression following 24 h DA treatment in A375.SM and B16F10 cells. We substantiated our results for apoptotic occurrence through flow cytometry in B16F10 cells. Furthermore, we treated B16F10 cells with N-acetyl-L-cysteine (NAC). NAC treatment upregulated ATG-5 (p < 0.0001), beclin-1 (p < 0.0001) and LC3-I to LC3-II (p < 0.0001) conversion, which was inhibited in the DA treatment group. We also noticed a systematic upregulation of important markers for progression of G1 cell phase such as CDK-2 (p < 0.029), CDK-4 (p < 0.036), cyclin D1 (p < 0.0003) and cyclin E (p < 0.020) upon NAC treatment. In addition, we also observed a significant fold reduction (p < 0.05) in ROS fluorescent intensity and the expression of Bax (p < 0.0001), cytochrome C (p < 0.0001), cleaved caspase-9 (p > 0.010) and cleaved caspase-3 (p < 0.0001). NAC treatment was able to ameliorate DA-induced apoptosis and cell cycle arrest to support our finding. Our in vivo xenograft model also revealed similar findings, such as downregulation of CDK-2 (p < 0.0001) and CDK-4 (p < 0.0142) and upregulation of Bax (p < 0.0001), cytochrome C (p < 0.0001), cleaved caspase 3 (p < 0.0001) and cleaved caspase 9 (p < 0.0001). In summary, our study revealed that DA is an effective treatment against B16F10 melanoma cells and xenograft mice model.
Collapse
Affiliation(s)
- Sukkum Ngullie Chang
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.N.C.); (C.G.K.); (S.M.P.)
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea;
| | - Imran Khan
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea;
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Chang Geon Kim
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.N.C.); (C.G.K.); (S.M.P.)
| | - Seon Min Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.N.C.); (C.G.K.); (S.M.P.)
| | - Dong Kyu Choi
- New Drug Development Center, DGMIF, 88 Dongnae-ro, Dong-gu, Daegu 41061, Korea; (D.K.C.); (H.L.)
| | - Heejin Lee
- New Drug Development Center, DGMIF, 88 Dongnae-ro, Dong-gu, Daegu 41061, Korea; (D.K.C.); (H.L.)
| | - Buyng Su Hwang
- Nakdonggang National Institute of Biological Resources, Sangju 37242, Korea;
| | - Sun Chul Kang
- Department of Biotechnology, Daegu University, Gyeongsan 38453, Korea;
| | - Jae Gyu Park
- Advanced Bio Convergence Center (ABCC), Pohang Technopark Foundation, Pohang 37668, Korea; (S.N.C.); (C.G.K.); (S.M.P.)
| |
Collapse
|
19
|
Griffiths K, Lee JJ, Frenneaux MP, Feelisch M, Madhani M. Nitrite and myocardial ischaemia reperfusion injury. Where are we now? Pharmacol Ther 2021; 223:107819. [PMID: 33600852 DOI: 10.1016/j.pharmthera.2021.107819] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/25/2021] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease remains the leading cause of death worldwide despite major advances in technology and treatment, with coronary heart disease (CHD) being a key contributor. Following an acute myocardial infarction (AMI), it is imperative that blood flow is rapidly restored to the ischaemic myocardium. However, this restoration is associated with an increased risk of additional complications and further cardiomyocyte death, termed myocardial ischaemia reperfusion injury (IRI). Endogenously produced nitric oxide (NO) plays an important role in protecting the myocardium from IRI. It is well established that NO mediates many of its downstream functions through the 'canonical' NO-sGC-cGMP pathway, which is vital for cardiovascular homeostasis; however, this pathway can become impaired in the face of inadequate delivery of necessary substrates, in particular L-arginine, oxygen and reducing equivalents. Recently, it has been shown that during conditions of ischaemia an alternative pathway for NO generation exists, which has become known as the 'nitrate-nitrite-NO pathway'. This pathway has been reported to improve endothelial dysfunction, protect against myocardial IRI and attenuate infarct size in various experimental models. Furthermore, emerging evidence suggests that nitrite itself provides multi-faceted protection, in an NO-independent fashion, against a myriad of pathophysiologies attributed to IRI. In this review, we explore the existing pre-clinical and clinical evidence for the role of nitrate and nitrite in cardioprotection and discuss the lessons learnt from the clinical trials for nitrite as a perconditioning agent. We also discuss the potential future for nitrite as a pre-conditioning intervention in man.
Collapse
Affiliation(s)
- Kayleigh Griffiths
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Jordan J Lee
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Michael P Frenneaux
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Melanie Madhani
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| |
Collapse
|
20
|
Cui Z, Shen X, Zhang X, Li F, Amevor FK, Zhu Q, Wang Y, Li D, Shu G, Tian Y, Zhao X. A functional polymorphism of inhibin alpha subunit at miR-181b-1-3p-binding site regulates proliferation and apoptosis of chicken ovarian granular cells. Cell Tissue Res 2021; 384:545-560. [PMID: 33439349 DOI: 10.1007/s00441-020-03356-w] [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: 08/01/2020] [Accepted: 11/18/2020] [Indexed: 10/22/2022]
Abstract
INHA, the gene encoding the inhibin alpha subunit, was involved in folliculogenesis in mammals, but no study was reported for its working pathway in birds. Here we hypothesize that gene polymorphism in INHA 3'UTR might influence miRNAs binding efficiency and further affect the function of this gene. Thus, we investigated the association between the 3'UTR single-nucleotide polymorphisms (SNPs) in INHA and the laying performance in chickens and further explore their possible molecular cascades in granulosa cells (GC). Five SNPs were detected in Tianfu green-shell layers and g. 22,178,975 G > A was significantly associated with total egg numbers at the age of 300 days (EN, n = 286). Birds carrying the AA genotype laid more EN than those with GG (P < 0.05). The allele transition from G to A in the 3'UTR of INHA gene destroyed a binding site which was targeted by miR-181b-1-3p. The expression abundances of INHA mRNA increased firstly and then decreased with follicle growing, and reached the top in the sixth largest pre-ovulation follicle, whereas miR-181b-1-3p levels in chicken pre-hierarchical follicles had the contrary tendency. Further studies indicated that high levels of miR-181b-1-3p increased apoptosis and reduced GC proliferation while miR-181b-1-3p inhibitors decreased apoptosis and promoted GC proliferation. Additionally, depression of INHA increased apoptosis and reduced GC proliferation via a caspase-3-dependent mitochondrial pathway. Generally, the mutation in INHA 3'UTR was tightly correlated with egg production in chickens, and blocked a binding site of miR-181b-1-3p. miR-181b-1-3p inhibited GC proliferation and promoted apoptosis by targeting INHA.
Collapse
Affiliation(s)
- Zhifu Cui
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiaoxu Shen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xianxian Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Fugui Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Felix Kwame Amevor
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Qing Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Diyan Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Gang Shu
- Department of Pharmacy, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yaofu Tian
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiaoling Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
21
|
Liu N, Lv B, Zeng Q. Hirsutenone selectively induces cytotoxic effects in human thyroid cancer cells by inhibiting cell migration and invasion, inducing apoptosis and targeting Wnt/beta-catenin signalling pathway. ALL LIFE 2021. [DOI: 10.1080/26895293.2021.1946432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Nan Liu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Bin Lv
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Qingdong Zeng
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| |
Collapse
|
22
|
Petricciuolo M, Davidescu M, Fettucciari K, Gatticchi L, Brancorsini S, Roberti R, Corazzi L, Macchioni L. The efficacy of the anticancer 3-bromopyruvate is potentiated by antimycin and menadione by unbalancing mitochondrial ROS production and disposal in U118 glioblastoma cells. Heliyon 2020; 6:e05741. [PMID: 33364504 PMCID: PMC7753915 DOI: 10.1016/j.heliyon.2020.e05741] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/06/2020] [Accepted: 12/11/2020] [Indexed: 12/14/2022] Open
Abstract
Metabolic reprogramming of tumour cells sustains cancer progression. Similar to other cancer cells, glioblastoma cells exhibit an increased glycolytic flow, which encourages the use of antiglycolytics as an effective complementary therapy. We used the antiglycolytic 3-bromopyruvate (3BP) as a metabolic modifier to treat U118 glioblastoma cells and investigated the toxic effects and the conditions to increase drug effectiveness at the lowest concentration. Cellular vitality was not affected by 3BP concentrations lower than 40 μM, although p-Akt dephosphorylation, p53 degradation, and ATP reduction occurred already at 30 μM 3BP. ROS generated in mitochondria were enhanced at 30 μM 3BP, possibly by unbalancing their generation and their disposal because of glutathione peroxidase inhibition. ROS triggered JNK and ERK phosphorylation, and cyt c release outside mitochondria, not accompanied by caspases-9 and -3 activation, probably due to 3BP-dependent alkylation of cysteine residues at caspase-9 catalytic site. To explore the possibility of sensitizing cells to 3BP treatment, we exploited 3BP effects on mitochondria by using 30 μM 3BP in association with antimycin A or menadione concentrations that in themselves exhibit poor toxicity. 3BP effect on cyt c release and cell vitality loss was potentiated due the greater oxidative stress induced by antimycin or menadione association with 3BP, supporting a preeminent role of mitochondrial ROS in 3BP toxicity. Indeed, the scavenger of mitochondrial superoxide MitoTEMPO counteracted 3BP-induced cyt c release and weakened the potentiating effect of 3BP/antimycin association. In conclusion, the biochemical mechanisms leading U118 glioblastoma cells to viability loss following 3BP treatment rely on mitochondrial ROS-dependent pathways. Their potentiation at low 3BP concentrations is consistent with the goal to minimize the toxic effect of the drug towards non-cancer cells.
Collapse
Affiliation(s)
- Maya Petricciuolo
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Magdalena Davidescu
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Katia Fettucciari
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Leonardo Gatticchi
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Stefano Brancorsini
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Rita Roberti
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Lanfranco Corazzi
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| | - Lara Macchioni
- Department of Experimental Medicine, University of Perugia, 06132, Perugia, Italy
| |
Collapse
|
23
|
Brown-Suedel AN, Bouchier-Hayes L. Caspase-2 Substrates: To Apoptosis, Cell Cycle Control, and Beyond. Front Cell Dev Biol 2020; 8:610022. [PMID: 33425918 PMCID: PMC7785872 DOI: 10.3389/fcell.2020.610022] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/03/2020] [Indexed: 01/12/2023] Open
Abstract
Caspase-2 belongs to the caspase family of proteins responsible for essential cellular functions including apoptosis and inflammation. Uniquely, caspase-2 has been identified as a tumor suppressor, but how it regulates this function is still unknown. For many years, caspase-2 has been considered an “orphan” caspase because, although it is able to induce apoptosis, there is an abundance of conflicting evidence that questions its necessity for apoptosis. Recent evidence supports that caspase-2 has non-apoptotic functions in the cell cycle and protection from genomic instability. It is unclear how caspase-2 regulates these opposing functions, which has made the mechanism of tumor suppression by caspase-2 difficult to determine. As a protease, caspase-2 likely exerts its functions by proteolytic cleavage of cellular substrates. This review highlights the known substrates of caspase-2 with a special focus on their functional relevance to caspase-2’s role as a tumor suppressor.
Collapse
Affiliation(s)
- Alexandra N Brown-Suedel
- Hematology-Oncology Section, Department of Pediatrics, Department of Molecular Cell Biology, Baylor College of Medicine, Houston, TX, United States.,William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX, United States
| | - Lisa Bouchier-Hayes
- Hematology-Oncology Section, Department of Pediatrics, Department of Molecular Cell Biology, Baylor College of Medicine, Houston, TX, United States.,William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
24
|
Saliev T, Baiskhanova D, Beznosko D, Begimbetova D, Umbayev B, Nurgozhin T, Fakhradiyev I, Tanabayev B, Pavalkis D. A New Insight on the Radioprotective Potential of Epsilon-Aminocaproic Acid. MEDICINA (KAUNAS, LITHUANIA) 2020; 56:E663. [PMID: 33266046 PMCID: PMC7760922 DOI: 10.3390/medicina56120663] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/13/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022]
Abstract
Background and objectives: The aim of the study was to scrutinize the ability of epsilon-aminocaproic acid (EACA) to prevent radiation-induced damage to human cells. Materials and Methods: Human peripheral blood mononuclear cells (PBMCs) were exposed to ionizing radiation at three low doses (22.62 mGy, 45.27 mGy, and 67.88 mGy) in the presence of EACA at the concentration of 50 ng/mL. Results: EACA was able to prevent cell death induced by low-dose X-ray radiation and suppress the formation of reactive oxygen species (ROS). EACA also demonstrated a capacity to protect DNA from radiation-induced damage. The data indicated that EACA is capable of suppression of radiation-induced apoptosis. Comparative tests of antioxidative activity of EACA and a range of free radical scavengers showed an ability of EACA to effectively inhibit the generation of ROS. Conclusions: This study showed that the pretreatment of PBMCs with EACA is able to protect the cells from radiation-elicited damage, including free radicals' formation, DNA damage, and apoptosis.
Collapse
Affiliation(s)
- Timur Saliev
- S.D. Asfendiyarov Kazakh National Medical University, Almaty 050000, Kazakhstan; (T.N.); (I.F.)
| | - Dinara Baiskhanova
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (D.B.); (D.B.); (B.U.)
| | | | - Dinara Begimbetova
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (D.B.); (D.B.); (B.U.)
| | - Bauyrzhan Umbayev
- National Laboratory Astana, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (D.B.); (D.B.); (B.U.)
| | - Talgat Nurgozhin
- S.D. Asfendiyarov Kazakh National Medical University, Almaty 050000, Kazakhstan; (T.N.); (I.F.)
| | - Ildar Fakhradiyev
- S.D. Asfendiyarov Kazakh National Medical University, Almaty 050000, Kazakhstan; (T.N.); (I.F.)
| | | | - Dainius Pavalkis
- NJSC “Astana Medical University”, Nur-sultan 010000, Kazakhstan;
| |
Collapse
|
25
|
The Evolutionary Origins of Programmed Cell Death Signaling. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a036442. [PMID: 31818855 DOI: 10.1101/cshperspect.a036442] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Programmed cell death (PCD) pathways are found in many phyla, ranging from developmentally programmed apoptosis in animals to cell-autonomous programmed necrosis pathways that limit the spread of biotrophic pathogens in multicellular assemblies. Prominent examples for the latter include animal necroptosis and pyroptosis, plant hypersensitive response (HR), and fungal heterokaryon incompatibility (HI) pathways. PCD pathways in the different kingdoms show fundamental differences in execution mechanism, morphology of the dying cells, and in the biological sequelae. Nevertheless, recent studies have revealed remarkable evolutionary parallels, including a striking sequence relationship between the "HeLo" domains found in the pore-forming components of necroptosis and some types of plant HR and fungal HI pathways. Other PCD execution components show cross-kingdom conservation as well, or are derived from prokaryotic ancestors. The currently available data suggest a model, wherein the primordial eukaryotic PCD pathway used proteins similar to present-day plant R-proteins and caused necrotic cell death by direct action of Toll and IL-1 receptor (TIR) and HeLo-like domains.
Collapse
|
26
|
Boice A, Bouchier-Hayes L. Targeting apoptotic caspases in cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118688. [PMID: 32087180 DOI: 10.1016/j.bbamcr.2020.118688] [Citation(s) in RCA: 217] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/20/2020] [Accepted: 02/15/2020] [Indexed: 12/30/2022]
Abstract
Members of the caspase family of proteases play essential roles in the initiation and execution of apoptosis. These caspases are divided into two groups: the initiator caspases (caspase-2, -8, -9 and -10), which are the first to be activated in response to a signal, and the executioner caspases (caspase-3, -6, and -7) that carry out the demolition phase of apoptosis. Many conventional cancer therapies induce apoptosis to remove the cancer cell by engaging these caspases indirectly. Newer therapeutic applications have been designed, including those that specifically activate individual caspases using gene therapy approaches and small molecules that repress natural inhibitors of caspases already present in the cell. For such approaches to have maximal clinical efficacy, emerging insights into non-apoptotic roles of these caspases need to be considered. This review will discuss the roles of caspases as safeguards against cancer in the context of the advantages and potential limitations of targeting apoptotic caspases for the treatment of cancer.
Collapse
Affiliation(s)
- Ashley Boice
- Department of Pediatrics, Division of Hematology-Oncology and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA
| | - Lisa Bouchier-Hayes
- Department of Pediatrics, Division of Hematology-Oncology and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|
27
|
Thabassum Akhtar Iqbal S, Tirupathi Pichiah PB, Raja S, Arunachalam S. Paeonol Reverses Adriamycin Induced Cardiac Pathological Remodeling through Notch1 Signaling Reactivation in H9c2 Cells and Adult Zebrafish Heart. Chem Res Toxicol 2020; 33:312-323. [PMID: 31307187 DOI: 10.1021/acs.chemrestox.9b00093] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adriamycin is a commonly prescribed chemotherapeutic drug for a wide range of cancers. Adriamycin causes cardiotoxicity as an adverse effect that limits its clinical application in cancer treatment. Several mechanisms have been proposed to explain the toxicity it causes in heart cells. Disruption of inherent cardiac repair mechanism is the least understood mechanism of Adriamycin-induced cardiotoxicity. Adriamycin induces pathological remodeling in cardiac cells by promoting apoptosis, hypertrophy, and fibrosis. We found that Adriamycin inhibited Notch1 in a time- and dose-dependent manner in H9c2 cells. We used Paeonol, a Notch1 activator, and analyzed the markers of apoptosis, hypertrophy, and fibrosis in H9c2 cells in vitro and in adult zebrafish heart in vivo as model systems to study Adriamycin-induced cardiotoxicity. Paeonol activated Notch1 signaling and expression of its downstream target genes effectively in the Adriamycin-treated condition in vitro and in vivo. Also we detected that Notch activation using Paeonol protected the cells from apoptosis, collagen deposition, and hypertrophy response using functional assays. We conclude that Adriamycin induced cardiotoxicity by promoting the pathological cardiac remodeling through inhibition of Notch1 signaling and that the Notch1 reactivation by Paeonol protected the cells and reversed the cardiotoxicity.
Collapse
Affiliation(s)
- Syeda Thabassum Akhtar Iqbal
- School of Bio-Sciences and Technology , Vellore Institute of Technology , Vellore , Tamilnadu PIN 632014 , India
| | | | - Sudhakaran Raja
- School of Bio-Sciences and Technology , Vellore Institute of Technology , Vellore , Tamilnadu PIN 632014 , India
| | - Sankarganesh Arunachalam
- Department of Biotechnology , Kalasalingam Academy of Research and Education , Krishnankoil , Virudhunagar, Tamilnadu PIN 626126 , India
| |
Collapse
|
28
|
Kazim N, Adhikari A, Oh TJ, Davie J. The transcription elongation factor TCEA3 induces apoptosis in rhabdomyosarcoma. Cell Death Dis 2020; 11:67. [PMID: 31988307 PMCID: PMC6985194 DOI: 10.1038/s41419-020-2258-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/13/2022]
Abstract
TCEA3 is one of three genes representing the transcription elongation factor TFIIS family in vertebrates. TCEA3 is upregulated during skeletal muscle differentiation and acts to promote muscle specific gene expression during myogenesis. Rhabdomyosarcoma (RMS) is a pediatric cancer derived from the muscle lineage, but the expression or function of TCEA3 in RMS was uncharacterized. We found that TCEA3 expression was strongly inhibited in RMS cell lines representing both ERMS and ARMS subtypes of RMS. TCEA3 expression correlates with DNA methylation and we show that TBX2 is also involved in the repression of TCEA3 in RMS cell lines. Ectopic expression of TCEA3 inhibited proliferation of RMS cell lines and initiated apoptosis through both the intrinsic and extrinsic pathways. We found that only pan-caspase inhibitors could block apoptosis in the presence of TCEA3. While expression of TCEA3 is highest in skeletal muscle, expression has been detected in other tissues as well, including breast, ovarian and prostate. We found that ectopic expression of TCEA3 also promotes apoptosis in HeLa, MCF7, MDA-231, and PC3 cell lines, representing cervical, breast, and prostate cancer, respectively. Restoration of TCEA3 expression in RMS cell lines enhanced sensitivity to chemotherapeutic drugs, including TRAIL. Thus, TCEA3 presents a novel target for therapeutic strategies to promote apoptosis and enhance sensitivity to current chemotherapeutic drugs.
Collapse
Affiliation(s)
- Noor Kazim
- Department of Biomedical Science, Cornell University, Ithaca, NY, 14850, USA
| | - Abhinav Adhikari
- Department of Biochemistry and Molecular Biology and Simmons Cancer Institute, Southern Illinois University School of Medicine, Carbondale, IL, 62901, USA
| | - Teak Jung Oh
- Department of Biochemistry, University of Illinois Urbana, Champaign, IL, 61820, USA
| | - Judith Davie
- Department of Biochemistry and Molecular Biology and Simmons Cancer Institute, Southern Illinois University School of Medicine, Carbondale, IL, 62901, USA.
| |
Collapse
|
29
|
Lin C, Yuan H, Wang W, Zhu Z, Lu Y, Wang J, Feng F, Wu J. Importance of PNO1 for growth and survival of urinary bladder carcinoma: Role in core-regulatory circuitry. J Cell Mol Med 2019; 24:1504-1515. [PMID: 31800162 PMCID: PMC6991670 DOI: 10.1111/jcmm.14835] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 10/20/2019] [Indexed: 02/06/2023] Open
Abstract
PNO1 (partner of Nob1) was known as a RNA‐binding protein in humans, and its ortholog PNO1 was reported to participate ribosome and proteasome biogenesis in yeasts. Yet there have been few studies about its functions in mammalian cells, and so far its role in human cells has never been reported, especially in urinary bladder cancer (UBC).We interrogated the cellular functions and clinical significance of PNO1 in, and its molecular mechanism through microarrays and bioinformatics analysis. Our findings support that PNO1 participates in promoting proliferation and colonogenesis, while reducing apoptosis of UBC cells, and is also predicted to be associated with the migration and metastasis of UBC PNO1 knockdown (KD) attenuated the tumorigenesis ability of UBC in mouse. PNO1 KD led to the altered expression of 1543 genes that are involved in a number of signalling pathways, biological functions and regulation networks. CD44, PTGS2, cyclin D1, CDK1, IL‐8, FRA1, as well as mTOR, p70 S6 kinase, p38 and Caspase‐3 proteins were all down‐regulated in PNO1 KD cells, suggesting the involvement of PNO1 in inflammatory responses, cell cycle regulation, chemotaxis, cell growth and proliferation, apoptosis, cell migration and invasiveness. This study will enhance our understanding of the molecular mechanism of UBC and may eventually provide novel targets for individualized cancer therapy.
Collapse
Affiliation(s)
- Chunhua Lin
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Hejia Yuan
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Wenting Wang
- The Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Zhe Zhu
- Division of Regenerative Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Youyi Lu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jiahui Wang
- The Central Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Fan Feng
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jitao Wu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| |
Collapse
|
30
|
Shlomovitz I, Speir M, Gerlic M. Flipping the dogma - phosphatidylserine in non-apoptotic cell death. Cell Commun Signal 2019; 17:139. [PMID: 31665027 PMCID: PMC6819419 DOI: 10.1186/s12964-019-0437-0] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/10/2019] [Indexed: 12/18/2022] Open
Abstract
The exposure of phosphatidylserine (PS) on the outer plasma membrane has long been considered a unique feature of apoptotic cells. Together with other "eat me" signals, it enables the recognition and phagocytosis of dying cells (efferocytosis), helping to explain the immunologically-silent nature of apoptosis. Recently, however, PS exposure has also been reported in non-apoptotic forms of regulated inflammatory cell death, such as necroptosis, challenging previous dogma. In this review, we outline the evidence for PS exposure in non-apoptotic cells and extracellular vesicles (EVs), and discuss possible mechanisms based on our knowledge of apoptotic-PS exposure. In addition, we examine the outcomes of non-apoptotic PS exposure, including the reversibility of cell death, efferocytosis, and consequent inflammation. By examining PS biology, we challenge the established approach of distinguishing apoptosis from other cell death pathways by AnnexinV staining of PS externalization. Finally, we re-evaluate how PS exposure is thought to define apoptosis as an immunologically silent process distinct from other non-apoptotic and inflammatory cell death pathways. Ultimately, we suggest that a complete understanding of how regulated cell death processes affect the immune system is far from being fully elucidated.
Collapse
Affiliation(s)
- Inbar Shlomovitz
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mary Speir
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168 Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3800 Australia
| | - Motti Gerlic
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
31
|
Trifan A, Vasincu A, Luca SV, Neophytou C, Wolfram E, Opitz SEW, Sava D, Bucur L, Cioroiu BI, Miron A, Aprotosoaie AC, Cioanca O, Hancianu M, Jitareanu A, Constantinou AI. Unravelling the potential of seaweeds from the Black Sea coast of Romania as bioactive compounds sources. Part I: Cystoseira barbata (Stackhouse) C. Agardh. Food Chem Toxicol 2019; 134:110820. [PMID: 31539616 DOI: 10.1016/j.fct.2019.110820] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 12/30/2022]
Abstract
The Romanian coastlines of the Black Sea have abundant seaweed resources, but little effort has been done to investigate their biological potential. The aim of the present study was to assess the in vitro antioxidant and anti-proliferative effects of Cystoseira barbata (Stackhouse) C. Agardh (Sargassaceae), a brown alga inhabiting the Black Sea coast of Romania. The 70% acetone, methanol and water extracts of C. barbata were evaluated for their total phenolic content, antioxidant activity and anti-proliferative potential against human tumor cell lines (pulmonary A549, colon HT-29, mammary MCF-7) and the non-tumor mammary epithelial MCF-10A cell line. C. barbata 70% acetone extract (CBAE) displayed the highest antioxidant and cytotoxic activities. The mechanism of CBAE anti-proliferative activity involved initially increased intracellular ROS accumulation, followed by increased DNA content in the subG1 phase and DNA fragmentation leading to excessive apoptosis. Thus, our study provides a theoretical basis for the use of CBAE as a tumor preventive agent. Furthermore, UHPLC-DAD-QTOF-MS analysis of CBAE tentatively identified 18 phlorotannins as fucophlorethol and eckol derivatives, containing three up to seven phloroglucinol units. In conclusion, C. barbata represents a valuable source for the development of macroalgal-based products with putative use as nutraceuticals and pharmaceuticals.
Collapse
Affiliation(s)
- Adriana Trifan
- Department of Pharmacognosy, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, Iasi, 700115, Romania
| | - Alexandru Vasincu
- Department of Pharmacodynamics and Clinical Pharmacy, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, Iasi, 700115, Romania.
| | - Simon Vlad Luca
- Department of Pharmacognosy, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, Iasi, 700115, Romania; Biothermodynamics, TUM School of Life and Food Sciences Weihenstephan, Technical University of Munich, Freising, 85354, Germany.
| | | | - Evelyn Wolfram
- Centre for Biochemistry and Bioanalytics, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Wädenswil, 8820, Switzerland
| | - Sebastian E W Opitz
- Phytopharmacy and Natural Products Research Group, Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Wädenswil, 8820, Switzerland
| | - Daciana Sava
- Department of Natural Sciences, Faculty of Natural and Agricultural Sciences, Ovidius University, Constanta, 900470, Romania
| | - Laura Bucur
- Department of Pharmacognosy, Faculty of Pharmacy, Ovidius University, Constanta, 900470, Romania
| | | | - Anca Miron
- Department of Pharmacognosy, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, Iasi, 700115, Romania
| | - Ana Clara Aprotosoaie
- Department of Pharmacognosy, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, Iasi, 700115, Romania
| | - Oana Cioanca
- Department of Pharmacognosy, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, Iasi, 700115, Romania
| | - Monica Hancianu
- Department of Pharmacognosy, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, Iasi, 700115, Romania
| | - Alexandra Jitareanu
- Department of Toxicology, Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, Iasi, 700115, Romania
| | | |
Collapse
|
32
|
Shemorry A, Harnoss JM, Guttman O, Marsters SA, Kőműves LG, Lawrence DA, Ashkenazi A. Caspase-mediated cleavage of IRE1 controls apoptotic cell commitment during endoplasmic reticulum stress. eLife 2019; 8:47084. [PMID: 31453810 PMCID: PMC6711704 DOI: 10.7554/elife.47084] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/14/2019] [Indexed: 12/22/2022] Open
Abstract
Upon detecting endoplasmic reticulum (ER) stress, the unfolded protein response (UPR) orchestrates adaptive cellular changes to reestablish homeostasis. If stress resolution fails, the UPR commits the cell to apoptotic death. Here we show that in hematopoietic cells, including multiple myeloma (MM), lymphoma, and leukemia cell lines, ER stress leads to caspase-mediated cleavage of the key UPR sensor IRE1 within its cytoplasmic linker region, generating a stable IRE1 fragment comprising the ER-lumenal domain and transmembrane segment (LDTM). This cleavage uncouples the stress-sensing and signaling domains of IRE1, attenuating its activation upon ER perturbation. Surprisingly, LDTM exerts negative feedback over apoptotic signaling by inhibiting recruitment of the key proapoptotic protein BAX to mitochondria. Furthermore, ectopic LDTM expression enhances xenograft growth of MM tumors in mice. These results uncover an unexpected mechanism of cross-regulation between the apoptotic caspase machinery and the UPR, which has biologically significant consequences for cell survival under ER stress.
Collapse
Affiliation(s)
- Anna Shemorry
- Cancer Immunology, Genentech, South San Francisco, United States
| | | | - Ofer Guttman
- Cancer Immunology, Genentech, South San Francisco, United States
| | - Scot A Marsters
- Cancer Immunology, Genentech, South San Francisco, United States
| | - László G Kőműves
- Department of Pathology, Genentech, South San Francisco, United States
| | - David A Lawrence
- Cancer Immunology, Genentech, South San Francisco, United States
| | - Avi Ashkenazi
- Cancer Immunology, Genentech, South San Francisco, United States
| |
Collapse
|
33
|
Zhou Z, Zuo CL, Li XS, Ye XP, Zhang QY, Wang P, Zhang RX, Chen G, Yang JL, Chen Y, Ma QY, Song HD. Uterus globulin associated protein 1 (UGRP1) is a potential marker of progression of Graves' disease into hypothyroidism. Mol Cell Endocrinol 2019; 494:110492. [PMID: 31255731 DOI: 10.1016/j.mce.2019.110492] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 01/26/2023]
Abstract
Approximately 20% of Graves' disease (GD) patients may result eventually in hypothyroidism in their natural course. Uterus globulin-associated protein 1 (UGRP1) was associated with GD in our previous study. Here we investigated the role of UGRP1 in the development of autoimmune thyroid disease (AITD). The results showed that UGRP1 was expressed in the thyrocytes of most Hashimoto's thyroiditis (HT) patients and a proportion of GD patients (293 HT and 198 GD). The pathologic features of UGRP1-positive thyrocytes resembled "Hürthle cells", and were surrounded by infiltrated leukocytes. The positivity rate of TPOAb in UGRP1-positive GD patients was much higher than that in -negative GD patients. Moreover, UGRP1 was co-expressed with Fas and HLA-DR in the thyrocytes of AITD patients. We also found IL-1β but not Th1 or Th2 cytokines was able to upregulate the expression of UGRP1. Our findings indicated that UGRP1 may be a novel marker in thyrocytes to predict GD patients who develop hypothyroidism.
Collapse
Affiliation(s)
- Zheng Zhou
- The Core Laboratory in Medical Center of Clinical Research, Department of Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Chun-Lin Zuo
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China.
| | - Xue-Song Li
- Department of Endocrinology, Minhang Hospital, Fudan University, Shanghai, China.
| | - Xiao-Ping Ye
- The Core Laboratory in Medical Center of Clinical Research, Department of Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Qian-Yue Zhang
- The Core Laboratory in Medical Center of Clinical Research, Department of Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Ping Wang
- Department of Pathology, Basic Medical College, Zhejiang Chinese Medical University, Zhejiang, China.
| | - Rong-Xin Zhang
- Department of Thoracic Surgery, The First Hospital Affiliated to University of Science and Technology of China, Anhui, China.
| | - Gang Chen
- Department of Endocrinology, Fujian Province Hospital, Fujian, China.
| | - Jia-Lin Yang
- Department of Endocrinology, Minhang Hospital, Fudan University, Shanghai, China.
| | - Yue Chen
- Department of Endocrinology, Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China.
| | - Qin-Yun Ma
- Department of Endocrinology and Metabolism, Shanghai Institute of Endocrine and Metabolic Diseases, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| | - Huai-Dong Song
- The Core Laboratory in Medical Center of Clinical Research, Department of Endocrinology, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
34
|
Park JE, Kim DH, Ha E, Choi SM, Choi JS, Chun KS, Joo SH. Thymoquinone induces apoptosis of human epidermoid carcinoma A431 cells through ROS-mediated suppression of STAT3. Chem Biol Interact 2019; 312:108799. [PMID: 31433961 DOI: 10.1016/j.cbi.2019.108799] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 08/07/2019] [Accepted: 08/17/2019] [Indexed: 12/21/2022]
Abstract
Black seed (Nigella sativa) oil has been used in various dermatological applications, and its major constituent, thymoquinone (TQ) has been shown to exhibit antiproliferative activity against various cancer cells. In this study, we tried to provide a mechanistic basis of apoptosis induced by TQ. Skin squamous carcinoma A431 cells were treated with TQ to monitor the apoptosis induced by TQ. Western blot analysis was performed to detect expression of apoptotic or anti-apoptotic proteins. Cell viability and apoptosis were measured by using the MTT test and FACS analysis, respectively. The induction of intracellular reactive oxygen species (ROS) by TQ was evaluated by 2',7'-dichlorofluorescein diacetate staining. In vivo xenograft study was followed to confirm the antiproliferative effect of TQ. Treatment of A431 cells with TQ-induced apoptosis, which was associated with the induction of p53 and Bax, inhibition of Mdm2, Bcl-2, and Bcl-xl expression, and activation of caspase-9, -7, and -3. TQ inhibited the constitutive phosphorylation and DNA binding activity of signal transducer and activator of transcription-3 (STAT3) in A431 cells by blocking the phosphorylation of the upstream kinase, Src. Moreover, the expression of STAT3 target gene products, cyclin D1 and survivin, was attenuated by TQ treatment. The generation of ROS was increased during TQ-induced apoptosis, and the pretreatment of N-acetyl cysteine, a ROS scavenger, reversed the apoptotic effect of TQ. In vivo study with NOD scid gamma (NSG) mice confirmed the inhibitory effect of TQ on the growth of A431 cells. Our results provide the first demonstration that TQ induces the apoptosis of A431 cells through generation of ROS and inhibition of STAT3 signaling.
Collapse
Affiliation(s)
- Ji Eun Park
- College of Pharmacy, Keimyung University, Daegu, 42601, South Korea
| | - Do-Hee Kim
- College of Pharmacy, Seoul National University, Seoul, 08826, South Korea
| | - Eunyoung Ha
- Department of Biochemistry, Keimyung University School of Medicine, South Korea
| | - Seung Mi Choi
- College of Pharmacy, Keimyung University, Daegu, 42601, South Korea
| | - Joon-Seok Choi
- Department of Pharmacy, Daegu Catholic University, Gyeongbuk, 38430, South Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu, 42601, South Korea.
| | - Sang Hoon Joo
- Department of Pharmacy, Daegu Catholic University, Gyeongbuk, 38430, South Korea.
| |
Collapse
|
35
|
Meena M, Samal S. Alternaria host-specific (HSTs) toxins: An overview of chemical characterization, target sites, regulation and their toxic effects. Toxicol Rep 2019; 6:745-758. [PMID: 31406682 PMCID: PMC6684332 DOI: 10.1016/j.toxrep.2019.06.021] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 06/18/2019] [Accepted: 06/22/2019] [Indexed: 02/05/2023] Open
Abstract
Alternaria causes pathogenic disease on various economically important crops having saprophytic to endophytic lifecycle. Pathogenic fungi of Alternaria species produce many primary and secondary metabolites (SMs). Alternaria species produce more than 70 mycotoxins. Several species of Alternaria produce various phytotoxins that are host-specific (HSTs) and non-host-specific (nHSTs). These toxins have various negative impacts on cell organelles including chloroplast, mitochondria, plasma membrane, nucleus, Golgi bodies, etc. Non-host-specific toxins such as tentoxin (TEN), Alternaric acid, alternariol (AOH), alternariol 9-monomethyl ether (AME), brefeldin A (dehydro-), Alternuene (ALT), Altertoxin-I, Altertoxin-II, Altertoxin-III, zinniol, tenuazonic acid (TeA), curvularin and alterotoxin (ATX) I, II, III are known toxins produced by Alternaria species. In other hand, Alternaria species produce numerous HSTs such as AK-, AF-, ACT-, AM-, AAL- and ACR-toxin, maculosin, destruxin A, B, etc. are host-specific and classified into different family groups. These mycotoxins are low molecular weight secondary metabolites with various chemical structures. All the HSTs have different mode of actions, biochemical reactions, and signaling mechanisms to causes diseases in the host plants. These HSTs have devastating effects on host plant tissues by affecting biochemical and genetic modifications. Host-specific mycotoxins such as AK-toxin, AF-toxin, and AC-toxin have the devastating effect on plants which causes DNA breakage, cytotoxic, apoptotic cell death, interrupting plant physiology by mitochondrial oxidative phosphorylation and affect membrane permeability. This article will elucidate an understanding of the disease mechanism caused by several Alternaria HSTs on host plants and also the pathways of the toxins and how they caused disease in plants.
Collapse
Key Words
- 1O2, singlet oxygen
- AA, ascorbic acid
- ALT, alternuene
- AME, alternariol 9-monomethyl ether
- AOH, alternariol
- APX, ascorbate peroxidase
- ATX, alterotoxin
- Alternaria species
- CAT, catalase
- CDCs, conditionally dispensable chromosomes
- DHAR, dehydroascorbate reductase
- DHT, dihydrotentoxin
- GPX, guaiacol peroxidase
- GR, glutathione reductase
- GSH, glutathione
- H2O2, hydrogen peroxide
- HR, hypersensitive response
- HSTs, host specific toxins
- Host-specific toxins
- MDHAR, monodehydroascorbate reductase
- NO, nitric oxide
- NRPS, nonribosomal peptide synthetase
- Non-host-specific toxins
- O2˙ˉ, superoxide anion
- PCD, programmed cell death
- PKS, polyketide synthase gene
- Pathogenicity
- REMI, restriction enzyme-mediated integration
- ROS, reactive oxygen species
- SMs, secondary metabolites
- SOD, superoxide dismutase
- Secondary metabolites
- TEN, tentoxin
- TeA, tenuazonic acid
- UGT, UDP-Glucuronosyltransferases
- nHSTs, non-host specific toxins
- ˙OH, hydroxyl radical
Collapse
Affiliation(s)
- Mukesh Meena
- Department of Botany, University College of Science, Mohanlal Sukhadia University, Udaipur, 313001, India
- Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Swarnmala Samal
- Centre of Advanced Study in Botany, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| |
Collapse
|
36
|
Goh JXH, Tan LTH, Goh JK, Chan KG, Pusparajah P, Lee LH, Goh BH. Nobiletin and Derivatives: Functional Compounds from Citrus Fruit Peel for Colon Cancer Chemoprevention. Cancers (Basel) 2019; 11:E867. [PMID: 31234411 PMCID: PMC6627117 DOI: 10.3390/cancers11060867] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 12/19/2022] Open
Abstract
The search for effective methods of cancer treatment and prevention has been a continuous effort since the disease was discovered. Recently, there has been increasing interest in exploring plants and fruits for molecules that may have potential as either adjuvants or as chemopreventive agents against cancer. One of the promising compounds under extensive research is nobiletin (NOB), a polymethoxyflavone (PMF) extracted exclusively from citrus peel. Not only does nobiletin itself exhibit anti-cancer properties, but its derivatives are also promising chemopreventive agents; examples of derivatives with anti-cancer activity include 3'-demethylnobiletin (3'-DMN), 4'-demethylnobiletin (4'-DMN), 3',4'-didemethylnobiletin (3',4'-DMN) and 5-demethylnobiletin (5-DMN). In vitro studies have demonstrated differential efficacies and mechanisms of NOB and its derivatives in inhibiting and killing of colon cancer cells. The chemopreventive potential of NOB has also been well demonstrated in several in vivo colon carcinogenesis animal models. NOB and its derivatives target multiple pathways in cancer progression and inhibit several of the hallmark features of colorectal cancer (CRC) pathophysiology, including arresting the cell cycle, inhibiting cell proliferation, inducing apoptosis, preventing tumour formation, reducing inflammatory effects and limiting angiogenesis. However, these substances have low oral bioavailability that limits their clinical utility, hence there have been numerous efforts exploring better drug delivery strategies for NOB and these are part of this review. We also reviewed data related to patents involving NOB to illustrate the extensiveness of each research area and its direction of commercialisation. Furthermore, this review also provides suggested directions for future research to advance NOB as the next promising candidate in CRC chemoprevention.
Collapse
Affiliation(s)
- Joanna Xuan Hui Goh
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, 47500 Bandar Sunway, Selangor Darul Ehsan, Malaysia.
| | - Loh Teng-Hern Tan
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor Darul Ehsan, Malaysia.
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Joo Kheng Goh
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
| | - Kok Gan Chan
- Division of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia.
- International Genome Centre, Jiangsu University, Zhenjiang 212013, China.
| | - Priyia Pusparajah
- Medical Health and Translational Research Group, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia.
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor Darul Ehsan, Malaysia.
- Asian Centre for Evidence Synthesis in Population, Implementation and Clinical Outcomes (PICO), Health and Well-being Cluster, Global Asia in the 21st Century (GA21) Platform, Monash University Malaysia, Bandar Sunway 47500, Malaysia.
| | - Bey-Hing Goh
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, 47500 Bandar Sunway, Selangor Darul Ehsan, Malaysia.
- Asian Centre for Evidence Synthesis in Population, Implementation and Clinical Outcomes (PICO), Health and Well-being Cluster, Global Asia in the 21st Century (GA21) Platform, Monash University Malaysia, Bandar Sunway 47500, Malaysia.
| |
Collapse
|
37
|
Wei L, He L, Fu J, Liu Y, Ruan J, Liu L, Zhong Q. Molecular characterization of caspase-8-like and its expression induced by microcystin-LR in grass carp (Ctenopharygodon idella). FISH & SHELLFISH IMMUNOLOGY 2019; 89:727-735. [PMID: 30981886 DOI: 10.1016/j.fsi.2019.04.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 03/31/2019] [Accepted: 04/10/2019] [Indexed: 06/09/2023]
Abstract
Caspase-8, an initiator caspase, plays a vital role in apoptosis. In this study, caspase-8-like (named as Cicaspase-8-like), a homologue of caspase-8, was identified in grass carp (Ctenopharygodon idella). The full-length cDNA sequence of CiCaspase-8-like was 1409 bp and contained a 162 bp 5'-UTR, a 239 bp 3'-UTR and a 1008 bp coding sequence. The putative amino acids sequence was 335 residues long, including a large subunit (P20) and a small subunit (P10), but lacking conserved death effector domains. A histidine active site DHSQMDAFVCCVLSHG and a cysteine active-site motif KPKLFFIQACQG were found in P20. Phylogenetic analysis showed that Cicaspase-8-like clustered with the caspase-8 and caspase-8-like of other fish and grouped closely with Carassius auratus caspase-8-like. Quantitative real-time PCR revealed that the Cicaspase-8-like mRNA were expressed constitutively in all tested tissues from healthy grass carp, with high expression level in the blood, spleen, liver and gill, indicating its role in immune reaction. The expression of Cicaspase-8-like mRNA was decreased significantly in the liver because of the stress caused by microcystin-LR (MC-LR) (75 and 100 μg MC-LR/kg BW) at 24 h and 96 h post injection (P < 0.05), but it was increased significantly in grass carp treated with 25 μg MC-LR/kg BW at 24 h (P < 0.05) post injection. Cleaved fragments of Cicaspase-8-like were observed using western blot analysis, and the expression of Cicaspase-8-like protein was increased after MC-LR treatments. Moreover, the expression of both caspase-9 and caspase-3 mRNA increased significantly after treatment with the three doses of MC-LR. TUNEL assay results showed remarkable changes in apoptosis after the MC-LR treatment. These results suggest that Cicaspase-8-like is an important caspase and plays an essential role in MC-LR-induced apoptosis.
Collapse
Affiliation(s)
- LiLi Wei
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, PR China.
| | - Li He
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, PR China
| | - Jianping Fu
- College of Life Sciences, Jiangxi Normal University, Nanchang, Jiangxi Province, 330022, PR China
| | - Yi Liu
- College of Life Sciences, Jiangxi Normal University, Nanchang, Jiangxi Province, 330022, PR China
| | - Jiming Ruan
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, PR China
| | - Lin Liu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, PR China
| | - Qiwang Zhong
- College of Bioscience and Bioengineering, Jiangxi Agricultural University, Nanchang, Jiangxi Province, 330045, PR China.
| |
Collapse
|
38
|
Black phosphorus nanosheets based sensitive protease detection and inhibitor screening. Talanta 2019; 197:270-276. [PMID: 30771934 DOI: 10.1016/j.talanta.2019.01.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 01/01/2019] [Accepted: 01/05/2019] [Indexed: 02/06/2023]
|
39
|
Park BS, Choi NE, Lee JH, Kang HM, Yu SB, Kim HJ, Kang HK, Kim IR. Crosstalk between Fisetin-induced Apoptosis and Autophagy in Human Oral Squamous Cell Carcinoma. J Cancer 2019; 10:138-146. [PMID: 30662534 PMCID: PMC6329852 DOI: 10.7150/jca.28500] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/27/2018] [Indexed: 12/19/2022] Open
Abstract
Fisetin (3,3-,4-,7-tetrahydroxyflavone), a naturally occurring flavonoid, has antioxidant, anti-inflammatory, and anticancer effects. Oral squamous cell carcinoma (OSCC) has a 5-year survival rate lower than that of most other carcinomas, and can create functional and aesthetic problems for the patient. New therapies for OSCC are necessary, and treatment using plant-derived natural substances has recently become a trend. It has been suggested that autophagy may play an important role in cancer therapy. Several studies demonstrated that autophagy inhibition enhances apoptotic cell death. Therefore, autophagy inhibition might be a promising therapeutic method against OSCC. Our results showed that fisetin induced apoptotic cell death in human tongue squamous cell line Ca9-22 could be enhanced by inhibition of autophagy. Thus, autophagy process in fisetin treated OSCC might presumed to play a role of pro-survival. The combination of fisetin and an effective autophagy inhibitor could be a potentially adjuvant and useful treatment for oral cancer.
Collapse
Affiliation(s)
- Bong-Soo Park
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea.,BK21 PLUS Project, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea.,Institute of Translational Dental Sciences, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea
| | - Nak-Eun Choi
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea
| | - Ji Hye Lee
- Department of Oral Pathology, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea.,BK21 PLUS Project, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea.,Institute of Translational Dental Sciences, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea
| | - Hae-Mi Kang
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea.,BK21 PLUS Project, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea
| | - Su-Bin Yu
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea
| | - Hye-Jin Kim
- Department of Dental Hygiene, Dongeui University, Gaya 1-dong, Busanjin-gu, Busan, 47230, South Korea
| | - Hyun-Kyung Kang
- Department of Dental Hygiene, Silla University, 140 Baekyang-daero 700 beon-gil, Busan 46958, South Korea
| | - In-Ryoung Kim
- Department of Oral Anatomy, School of Dentistry, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea.,Institute of Translational Dental Sciences, Pusan National University, Busandaehak-ro, 49, Mulguem-eup, Yangsan-si, Gyeongsangnam-do, 50612, South Korea
| |
Collapse
|
40
|
Sarigöl-Kiliç Z, Ündeğer-Bucurgat Ü. The Apoptotic and Anti-apoptotic Effects of Pendimethalin and Trifluralin on A549 Cells In Vitro. Turk J Pharm Sci 2018; 15:364-369. [PMID: 32454683 DOI: 10.4274/tjps.94695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/17/2017] [Indexed: 12/22/2022]
Abstract
Objectives Pendimethalin and trifluralin are commonly used in many countries to control broadleaf weeds and grassy weed species because of their inhibitor effects on growth and cell division. In this study, we examined the apoptotic and anti-apoptotic potentials of pendimethalin and trifluralin on A549 human non-small lung cancer cells with several concentrations in vitro. Materials and Methods The expression levels of apoptosis-related genes BCL-2, BAX, CAS3, CAS9, P53, BIRC, and PPIA were examined using quantitative RT-PCR after 24 h treatment of 1, 10, 50, 100 and 500 μM pendimethalin and trifluralin. Results The effects of pendimethalin were found more repressive than trifluralin on all studied concentrations. Twenty-four hours' exposure with 100 μM pendimethalin and trifluralin altered the gene expressions, suppressing apoptosis and allowing cancer cells to grow and proliferate. Conclusion Care should be taken not to exceed the permissible values and residue limits in food during pendimethalin and trifluralin use in order to reduce the possible carcinogenic effects on humans.
Collapse
Affiliation(s)
- Zehra Sarigöl-Kiliç
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Ankara, Turkey
| | - Ülkü Ündeğer-Bucurgat
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Toxicology, Ankara, Turkey
| |
Collapse
|
41
|
Lei X, Zheng W, Ding H, Zhu X, Chen Q, Xu H, Zheng T, Tian Y. Effective harvesting of the marine microalga Thalassiosira pseudonana by Marinobacter sp. FL06. BIORESOURCE TECHNOLOGY 2018; 269:127-133. [PMID: 30165270 DOI: 10.1016/j.biortech.2018.08.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/16/2018] [Accepted: 08/19/2018] [Indexed: 06/08/2023]
Abstract
In this study, Marinobacter sp. FL06 was used to effectively harvest the energy-producing microalga Thalassiosira pseudonana through direct flocculation. Strain FL06 showed 92.7% flocculating efficiency against T. pseudonana, and no metal ion was added for the flocculation process, resulting in a more environmentally friendly process. The flocculation efficiency of FL06 was stable over a wide range of pH values and temperatures, indicating that the application of this bacteria has potential advantages under various conditions. Strain FL06 also exhibited flocculation activity against different microalgae, indicating that the strain can be used to harvest multiple types of microalgae. Strain FL06 showed high chemotactic ability toward algal cells, suggesting that chemotaxis is important for flocculation. This study provides the first demonstration that the Marinobacter genus could be used to harvest T. pseudonana biomass. In summary, the results showed that FL06 has the potential for effective harvesting of microalgal biomass.
Collapse
Affiliation(s)
- Xueqian Lei
- State Key Laboratory of Marine Environmental Science and Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, School of Life Sciences, Xiamen University, Xiamen, China
| | - Wei Zheng
- State Key Laboratory of Marine Environmental Science and Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, School of Life Sciences, Xiamen University, Xiamen, China
| | - Hongyan Ding
- State Key Laboratory of Marine Environmental Science and Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, School of Life Sciences, Xiamen University, Xiamen, China
| | - Xiaoying Zhu
- State Key Laboratory of Marine Environmental Science and Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, School of Life Sciences, Xiamen University, Xiamen, China
| | - Qiuliang Chen
- State Key Laboratory of Marine Environmental Science and Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, School of Life Sciences, Xiamen University, Xiamen, China
| | - Hong Xu
- State Key Laboratory of Marine Environmental Science and Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, School of Life Sciences, Xiamen University, Xiamen, China
| | - Tianling Zheng
- State Key Laboratory of Marine Environmental Science and Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yun Tian
- State Key Laboratory of Marine Environmental Science and Key Laboratory of the Ministry of Education for Coastal and Wetland Ecosystems, School of Life Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
42
|
Regulating Apoptosis by Degradation: The N-End Rule-Mediated Regulation of Apoptotic Proteolytic Fragments in Mammalian Cells. Int J Mol Sci 2018; 19:ijms19113414. [PMID: 30384441 PMCID: PMC6274719 DOI: 10.3390/ijms19113414] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/24/2018] [Accepted: 10/27/2018] [Indexed: 12/13/2022] Open
Abstract
A pivotal hallmark of some cancer cells is the evasion of apoptotic cell death. Importantly, the initiation of apoptosis often results in the activation of caspases, which, in turn, culminates in the generation of proteolytically-activated protein fragments with potentially new or altered roles. Recent investigations have revealed that the activity of a significant number of the protease-generated, activated, pro-apoptotic protein fragments can be curbed via their selective degradation by the N-end rule degradation pathways. Of note, previous work revealed that several proteolytically-generated, pro-apoptotic fragments are unstable in cells, as their destabilizing N-termini target them for proteasomal degradation via the N-end rule degradation pathways. Remarkably, previous studies also showed that the proteolytically-generated anti-apoptotic Lyn kinase protein fragment is targeted for degradation by the UBR1/UBR2 E3 ubiquitin ligases of the N-end rule pathway in chronic myeloid leukemia cells. Crucially, the degradation of cleaved fragment of Lyn by the N-end rule counters imatinib resistance in these cells, implicating a possible linkage between the N-end rule degradation pathway and imatinib resistance. Herein, we highlight recent studies on the role of the N-end rule proteolytic pathways in regulating apoptosis in mammalian cells, and also discuss some possible future directions with respect to apoptotic proteolysis signaling.
Collapse
|
43
|
Panickar KS, Jewell DE. The Benefit of Anti-Inflammatory and Renal-Protective Dietary Ingredients on the Biological Processes of Aging in the Kidney. BIOLOGY 2018; 7:biology7040045. [PMID: 30274250 PMCID: PMC6316594 DOI: 10.3390/biology7040045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/27/2018] [Accepted: 09/28/2018] [Indexed: 12/21/2022]
Abstract
One of the significant organ systems which decline in aging is the kidney. While the causes of age-associated decline in renal function are likely multifactorial, oxidative stress and inflammation are hypothesized to play important roles in the structural and functional changes of the kidney. During aging there is a general decline in the glomerular filtration rate (GFR), a primary measurement used to assess kidney function. Inflammation and oxidative stress have been hypothesized to have a significant detrimental effect on renal function in aging and this may be attenuated by renal protective dietary ingredients. These dietary ingredients may affect renal function directly or through a microbiome-mediated secondary product. Likewise, structural changes including renal tubular atrophy, interstitial fibrosis, and glomerulosclerosis have all been described in aging. Such detrimental changes may benefit from dietary ingredients that may delay or attenuate the occurrence of such changes. This review will describe the physiology and pathophysiology of aging in renal function with an emphasis on dogs and cats that develop a decline in kidney function naturally. In addition, the varying biomarkers of health and renal dysfunction will be discussed. Finally, we will evaluate the aid in the management of this normal decline through dietary intervention in animal models.
Collapse
Affiliation(s)
- Kiran S Panickar
- Science & Technology Center, Hill's Pet Nutrition, Inc., Topeka, KS 66617, USA.
| | - Dennis E Jewell
- Science & Technology Center, Hill's Pet Nutrition, Inc., Topeka, KS 66617, USA.
| |
Collapse
|
44
|
Gudipaty SA, Conner CM, Rosenblatt J, Montell DJ. Unconventional Ways to Live and Die: Cell Death and Survival in Development, Homeostasis, and Disease. Annu Rev Cell Dev Biol 2018; 34:311-332. [PMID: 30089222 DOI: 10.1146/annurev-cellbio-100616-060748] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Balancing cell death and survival is essential for normal development and homeostasis and for preventing diseases, especially cancer. Conventional cell death pathways include apoptosis, a form of programmed cell death controlled by a well-defined biochemical pathway, and necrosis, the lysis of acutely injured cells. New types of regulated cell death include necroptosis, pyroptosis, ferroptosis, phagoptosis, and entosis. Autophagy can promote survival or can cause death. Newly described processes of anastasis and resuscitation show that, remarkably, cells can recover from the brink of apoptosis or necroptosis. Important new work shows that epithelia achieve homeostasis by extruding excess cells, which then die by anoikis due to loss of survival signals. This mechanically regulated process both maintains barrier function as cells die and matches rates of proliferation and death. In this review, we describe these unconventional ways in which cells have evolved to die or survive, as well as the contributions that these processes make to homeostasis and cancer.
Collapse
Affiliation(s)
- Swapna A Gudipaty
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Christopher M Conner
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, USA;
| | - Jody Rosenblatt
- Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Denise J Montell
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, California 93106, USA;
| |
Collapse
|
45
|
Zhang S, Chen C, Qin X, Zhang Q, Liu J, Zhu J, Gao Y, Li L, Huang W. Ultrasensitive detection of trypsin activity and inhibitor screening based on the electron transfer between phosphorescence copper nanocluster and cytochrome c. Talanta 2018; 189:92-99. [PMID: 30086981 DOI: 10.1016/j.talanta.2018.06.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/30/2018] [Accepted: 06/09/2018] [Indexed: 11/19/2022]
Abstract
Trypsin, as one of important proteases, is specific for catalyzing the hydrolysis of peptide and ester bonds containing lysine and arginine residues at the C-terminus. The level of trypsin in biological fluids can serve as a reliable and specific diagnostic biomarker for pancreatic function and its pathological changes. Herein, we demonstrate the application of phosphorescent Cu NCs for trypsin detection for the first time depending on the electron transfer between Cu NCs and cyt c. Cyt c and Cu NCs were selected as the quencher and the fluorophore, respectively. Cu NCs could bind to the positively charged cyt c through electrostatic and hydrophobic interactions, and the phosphorescence of Cu NCs was efficiently quenched by the metal-containing heme of cyt c. In the presence of trypsin, cyt c was digested, thus phosphorescence of Cu NCs remained. Therefore, a new and continuous phosphorescence assay for the detection of trypsin activity and its inhibitor screening was established. The plot of relative fluorescence versus trypsin concentration obtains a good linear detection range from 0 to 20 ng/mL (R2 = 0.9657), and a detection limit of 2 ng/mL, which is much lower than 20 ng/mL of the sensor in buffer solution because of urine amplifying the phosphorescence signal of Cu NCs based on the FRET strategy. This assay still has been successfully applied to trypsin inhibitor screening, demonstrating its potential application in drug discovery.
Collapse
Affiliation(s)
- Shiyu Zhang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China
| | - Can Chen
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China
| | - Xiaofei Qin
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China
| | - Qianchen Zhang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China
| | - Jinhua Liu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China; State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha 410082, China.
| | - Jixin Zhu
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China
| | - Yongqian Gao
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China
| | - Lin Li
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China
| | - Wei Huang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, China; Shaanxi Institute of Flexible Electronics (SIFE), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an 710072, China
| |
Collapse
|
46
|
Li Y, Xu Y, Zheng T, Wang H. Amino acids in cell wall of Gram-positive bacterium Micrococcus sp. hsn08 with flocculation activity on Chlorella vulgaris biomass. BIORESOURCE TECHNOLOGY 2018; 249:417-424. [PMID: 29065323 DOI: 10.1016/j.biortech.2017.10.056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/09/2017] [Accepted: 10/13/2017] [Indexed: 06/07/2023]
Abstract
The aim of this work was to investigate the flocculation mechanism by Gram-positive bacterium, Micrococcus sp. hsn08 as a means for harvesting Chlorella vulgaris biomass. Bacterial cells of Micrococcus sp. hsn08 were added into algal culture to harvest algal cells through direct contacting with algae to form flocs. Viability dependence test confirmed that flocculation activity does not depend on live bacteria, but on part of the peptidoglycan. The further investigation has determined that amino acids in cell wall play an important role to flocculate algal cells. Positively charged calcium can combine bacterial and algal cells together, and form a bridge between them, thereby forming the flocs, suggesting that ions bridging is the main flocculation mechanism. These results suggest that bacterial cells of Micrococcus sp. hsn08 can be applied to harvest microalgae biomass with the help of amino acids in cell wall.
Collapse
Affiliation(s)
- Yi Li
- College of Life Sciences, Henan Normal University, Xinxiang 453007, China
| | - Yanting Xu
- College of Life Sciences, Henan Normal University, Xinxiang 453007, China
| | - Tianling Zheng
- State Key Laboratory of Marine Environmental Science, School of Life Sciences, Xiamen University, Xiamen 361005, China
| | - Hailei Wang
- College of Life Sciences, Henan Normal University, Xinxiang 453007, China.
| |
Collapse
|
47
|
Liu L, Zhang H, Song D, Wang Z. An upconversion nanoparticle-based fluorescence resonance energy transfer system for effectively sensing caspase-3 activity. Analyst 2018; 143:761-767. [PMID: 29327008 DOI: 10.1039/c7an01744h] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report a new fluorescence resonance energy transfer (FRET) sensing platform for the sensitive detection of caspase-3 activity in vitro and in cells using NaGdF4:Yb3+,Er3+@NaGdF4 upconversion nanoparticles (UCNPs) as the energy donor and Rhodamine B (RB) as the energy acceptor. The phosphorylated RB-modified peptide containing a caspase-3 cleavage site and cell-penetrating peptide (CPP) motif (sequence, (RB)-DEVDGGS(p)GCGT(p)GRKKRRQRRRPQ) is immobilized on the UCNP surface via the strong coordination interaction between Gd3+ ions with phosphate. After the cleavage of DEVD by caspase-3, the RB is released from the UCNP surface and the reduced upconversion luminescence (UCL) is recovered. Under the optimum conditions, the recovery ratio of the UCL is linearly dependent on the caspase-3 concentration within the range of 0.01 to 1000 pg mL-1 and with a limit of detection (LOD) of 0.01 pg mL-1 (S/N = 3). In particular, the as-proposed UCNP-based FRET sensing platform has reasonable selectivity which is successfully employed to monitor caspase-3 activity in drug-induced apoptosis of HeLa cells.
Collapse
Affiliation(s)
- Lin Liu
- College of Chemistry, Jilin University, Changchun 130012, P. R. China.
| | - Hua Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China.
| | - Daqian Song
- College of Chemistry, Jilin University, Changchun 130012, P. R. China.
| | - Zhenxin Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China.
| |
Collapse
|
48
|
Regulated Cell Death. DAMAGE-ASSOCIATED MOLECULAR PATTERNS IN HUMAN DISEASES 2018. [PMCID: PMC7123501 DOI: 10.1007/978-3-319-78655-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
In this chapter, the various subroutines of regulated cell death are neatly described by highlighting apoptosis and subforms of regulated necrosis such as necroptosis, ferroptosis, pyroptosis, and NETosis. Typically, all forms of regulated necrosis are defined by finite rupture of the plasma cell membrane. Apoptosis is characterized by an enzymatic machinery that consists of caspases which cause the morphologic features of this type of cell death. Mechanistically, apoptosis can be instigated by two major cellular signalling pathways: an intrinsic pathway that is initiated inside cells by mitochondrial release of pro-apoptotic factors or an extrinsic pathway that is initiated at the cell surface by various death receptors. In necroptosis, the biochemical processes are distinct from those found in apoptosis; in particular, there is no caspase activation. As such, necroptosis is a kinase-mediated cell death that relies on “receptor-interacting protein kinase 3” which mediates phosphorylation of the pseudokinase “mixed lineage kinase domain-like protein.” While ferroptosis is an iron-dependent, oxidative form of regulated necrosis that is biochemically characterized by accumulation of ROS from iron metabolism, oxidase activity, and lipid peroxidation products, pyroptosis is defined as a form of cell death (predominantly of phagocytes) that develops during inflammasome activation and is executed by caspase-mediated cleavage of the pore-forming protein gasdermin D. Finally, NETosis refers to a regulated death of neutrophils that is characterized by the release of chromatin-derived weblike structures released into the extracellular space. The chapter ends up with a discussion on the characteristic feature of regulated necrosis: the passive release of large amounts of constitutive DAMPs as a consequence of final plasma membrane rupture as well as the active secretion of inducible DAMPs earlier during the dying process. Notably, per cell death subroutine, the active secretion of inducible DAMPs varies, thereby determining different immunogenicity of dying cells.
Collapse
|
49
|
Senescence and cell death in chronic liver injury: roles and mechanisms underlying hepatocarcinogenesis. Oncotarget 2017; 9:8772-8784. [PMID: 29492237 PMCID: PMC5823588 DOI: 10.18632/oncotarget.23622] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/10/2017] [Indexed: 12/16/2022] Open
Abstract
Chronic liver injury (CLI) is a complex pathological process typically characterized by progressive destruction and regeneration of liver parenchymal cells due to diverse risk factors such as alcohol abuse, drug toxicity, viral infection, and genetic metabolic disorders. When the damage to hepatocytes is mild, the liver can regenerate itself and restore to the normal state; when the damage is irreparable, hepatocytes would undergo senescence or various forms of death including apoptosis, necrosis and necroptosis. These pathological changes not only promote the progression of the existing hepatopathies via various underlying mechanisms but are closely associated with hepatocarcinogenesis. In this review, we discuss the pathological changes that hepatocytes undergo during CLI, and their roles and mechanisms in the progression of hepatopathies and hepatocarcinogenesis. We also give a brief introduction about some animal models currently used for the research of CLI and progress in the research of CLI.
Collapse
|
50
|
Zuhrotun Nisa F, Astuti M, Murdiati A, Mubarika Haryana S. Anti-proliferation and Apoptosis Induction of Aqueous Leaf Extract of Carica papaya L. on Human Breast Cancer Cells MCF-7. Pak J Biol Sci 2017; 20:36-41. [PMID: 29023013 DOI: 10.3923/pjbs.2017.36.41] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND OBJECTIVE Breast cancer is the most frequently diagnosed cancer in women. Chemotherapy is the main method of breast cancer treatment but there are side effects. Carica papaya leaves is vegetable foods consumed by most people of Indonesia have potential as anticancer. The aim of this study was to investigate anti-proliferative and apoptotic induced effect of aqueous papaya leaves extracts on human breast cancer cell lines MCF-7. MATERIALS AND METHODS Inhibitory on cell proliferation was measured by MTT assay while apoptosis induction was measured using Annexin V. RESULTS The results showed that papaya leaf can inhibit the proliferation of human breast cancer cells MCF-7 with IC50 in 1319.25 μg mL-1. The IC50 values of papaya leaf extract was higher than the IC50 value quercetin and doxorubicin. Papaya leaf extract can also induce apoptosis of breast cancer cells MCF-7 about 22.54% for concentration 659.63 μg mL-1 and about 20.73% for concentration 329.81 μg mL-1. The percentage of cell apoptosis of papaya leaf extract lower than doxorubicin but higher than quercetin. CONCLUSION This study indicated that papaya leaf extract have potential as anticancer through mechanism anti-proliferation and apoptosis induction.
Collapse
Affiliation(s)
- Fatma Zuhrotun Nisa
- Department of Health and Nutrition, Faculty of Medicine, Universitas Gadjah Mada, Jl Farmako Sekip Utara, 55281 Yogyakarta, Indonesia
| | - Mary Astuti
- Faculty of Agriculture Technology, Universitas Gadjah Mada, Jl Flora No. 1, 55281 Yogyakarta, Indonesia
| | - Agnes Murdiati
- Faculty of Agriculture Technology, Universitas Gadjah Mada, Jl Flora No. 1, 55281 Yogyakarta, Indonesia
| | - Sofia Mubarika Haryana
- Department of Health and Nutrition, Faculty of Medicine, Universitas Gadjah Mada, Jl Farmako Sekip Utara, 55281 Yogyakarta, Indonesia
| |
Collapse
|