1
|
Horbach N, Kalinka M, Ćwilichowska-Puślecka N, Al Mamun A, Mikołajczyk-Martinez A, Turk B, Snipas SJ, Kasperkiewicz P, Groborz KM, Poręba M. Visualization of calpain-1 activation during cell death and its role in GSDMD cleavage using chemical probes. Cell Chem Biol 2025; 32:603-619.e7. [PMID: 40157358 DOI: 10.1016/j.chembiol.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 01/30/2025] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
Calpain-1, a calcium-dependent cysteine protease, plays a vital role in cellular processes such as cell death, cytoskeletal remodeling, signal transduction, and cell cycle progression. While its role in apoptosis, including substrate cleavage for orderly disassembly, is well established, its involvement in pyroptosis remains less understood. This study focused on developing chemical tools to detect calpain-1 activity. Using the hybrid combinatorial substrate library (HyCoSuL) approach with unnatural amino acids, we designed fluorescent substrates, inhibitors, and fluorescent activity-based probe (ABP) specific to calpain-1, enabling its visualization in living cells. We further investigated calpain-1's expression alongside cell death proteins in immune cells using mass cytometry and observed strong colocalization with gasdermin D (GSDMD). Additionally, we demonstrated that calpain-1 can hydrolyze GSDMD in vitro. Through fluorescence-based substrate assays and mass spectrometry, we identified putative cleavage sites within the GSDMD sequence that may promote pyroptosis. These findings underscore calpain-1's multifaceted role in cell death pathways, extending beyond apoptosis.
Collapse
Affiliation(s)
- Natalia Horbach
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | - Małgorzata Kalinka
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | | | - Abdulla Al Mamun
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | - Agata Mikołajczyk-Martinez
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland; Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| | - Boris Turk
- Jozef Stefan Institute, Sl-1000 Ljubljana, Slovenia
| | - Scott J Snipas
- SBP Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Paulina Kasperkiewicz
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | - Katarzyna M Groborz
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland; SBP Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Marcin Poręba
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland; Faculty of Medicine, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland.
| |
Collapse
|
2
|
Bourne CM, Raniszewski NR, Kulkarni M, Exconde PM, Liu S, Yost W, Wrong TJ, Patio RC, Mahale A, Kardhashi M, Shosanya T, Kambayashi M, Discher BM, Brodsky IE, Burslem GM, Taabazuing CY. Chemical Tools Based on the Tetrapeptide Sequence of IL-18 Reveals Shared Specificities between Inflammatory and Apoptotic Initiator Caspases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.23.639785. [PMID: 40060427 PMCID: PMC11888271 DOI: 10.1101/2025.02.23.639785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Caspases are a family of cysteine proteases that act as molecular scissors to cleave substrates and regulate biological processes such as programmed cell death and inflammation. Extensive efforts have been made to identify caspase substrates and to determine factors that dictate substrate specificity. We recently discovered that that the human inflammatory caspases (caspases-1, -4, and -5) cleave the cytokines IL-1β and IL-18 in a sequence-dependent manner. Here, we report the development of a new peptide-based probe and inhibitor based on the tetrapeptide sequence of IL-18 (LESD). We found that this inhibitor was most selective and potent at inhibiting caspase-8 activity (IC50 = 50 nM). We also discovered that our LESD-based inhibitor is more potent than the currently used z-IETD-FMK inhibitor that is thought to be the most selective and potent inhibitor of caspase-8. Accordingly, we demonstrate that the LESD based inhibitor prevents caspase-8 activation during Yersinia pseudotuberculosis infection in primary bone-marrow derived macrophages. Furthermore, we characterize the selectivity and potency of currently known substrates and inhibitors for the apoptotic and inflammatory caspases using the same activity units of each caspase. Our findings reveal that VX-765, a known caspase-1 inhibitor, also inhibits caspase-8 (IC50 = 1 μM) and even when specificities are shared, the caspases have different efficiencies and potencies for shared substrates and inhibitors. Altogether, we report the development of new tools that will facilitate the study of caspases and their roles in biology.
Collapse
Affiliation(s)
- Christopher M. Bourne
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Nicole R. Raniszewski
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Madhura Kulkarni
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Patrick M. Exconde
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Sherry Liu
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Winslow Yost
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Tristan J. Wrong
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Robert C. Patio
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Ashutosh Mahale
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Matilda Kardhashi
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Teni Shosanya
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Mirai Kambayashi
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Bohdana M. Discher
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Igor E. Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - George M. Burslem
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Cancer Biology and Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Cornelius Y. Taabazuing
- Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| |
Collapse
|
3
|
Wang Y, Liang Y, Huang Y, Wang W, Long X, Jiang L, Cheng T, Du J, Luo X. The mechanism of tea tree oil regulating the damage of hydrogen sulfide to spleen and intestine of chicken. Poult Sci 2025; 104:104605. [PMID: 39626606 PMCID: PMC11652936 DOI: 10.1016/j.psj.2024.104605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/04/2024] [Accepted: 11/26/2024] [Indexed: 01/25/2025] Open
Abstract
Intensification of poultry industry has led to a surge in animal product output, but this has also revealed issues with environmental management in poultry houses, particularly the harmful effects of high hydrogen sulfide (H2S) levels on poultry health. The study aimed to assess the therapeutic impact of tea tree oil (TTO) on H2S-induced spleen and intestinal injuries in chickens. A total of 240 one-day-old Lohmann Brown chicks were randomly divided into three groups: the control group (CON), the H2S exposure group (AVG), and the TTO treatment group (TTG), with four replicates, each consisting of 20 chicks. The experiment lasted 42 days. Results showed that TTO treatment alleviated tissue damage in the thymus, kidneys, spleen, and bursa of Fabricius, and improved the organ index (P < 0.05) compared with the AVG. Serum analysis revealed that TTO lowered levels of alanine aminotransferase(ALT), aspartate aminotransferase(AST), triglycerides(TG), CD3 positive CD4 positive T cells(CD3+CD4+), CD4 positive to CD8 positive Rratio(CD4+/CD8+), and alkaline phosphatase(AKP), while increasing albumin(ALB), globulin(GLO), immunoglobulin A(IgA), and immunoglobulin G(IgG) levels (P < 0.05). Intestinal findings indicated that TTO treatment enhanced villus height, reduced crypt depth, and upregulated the expression of Claudin 1, Occludin, and ZO-1 mRNA in the jejunum (P < 0.05). After TTO treatment, H2S-induced oxidative stress injury and apoptosis protein expression in spleen were improved (P < 0.05). TTO also reduced interferon-γ(INF-γ), tumor necrosis factor-α(TNF-α) and interleukin-1β(IL-1β) proteins (P < 0.05), while raising CD3+CD8+ T-cell subsets (P < 0.05). Compared with CON, TTO treatment alleviated serum biochemical disorders and intestinal damage caused by H2S exposure and restored them to normal (P > 0.05). In conclusion, TTO can improve spleen and intestinal function and reduce the effects of H2S on growth performance and health of chickens.
Collapse
Affiliation(s)
- Yachao Wang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China; Biomass Center, School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China.
| | - Yilei Liang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China; Biomass Center, School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China
| | - Yan Huang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China; Biomass Center, School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China
| | - Wei Wang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China; Biomass Center, School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China
| | - Xiaoyan Long
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China; Biomass Center, School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China
| | - Li Jiang
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China
| | - Tingting Cheng
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China; Biomass Center, School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China
| | - Jinfeng Du
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China
| | - Xuegang Luo
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China; Biomass Center, School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan 621000, PR China
| |
Collapse
|
4
|
Zhao J, Wang J, Zhao K, Zhang Y, Hu W. Protopanaxadiols Eliminate Behavioral Impairments and Mitochondrial Dysfunction in Parkinson's Disease Mice Model. Neurochem Res 2024; 49:1751-1761. [PMID: 38551796 PMCID: PMC11144128 DOI: 10.1007/s11064-024-04132-w] [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: 11/15/2023] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 06/02/2024]
Abstract
Currently, there are no effective therapies to cure Parkinson's disease (PD), which is the second most common neurodegenerative disease primarily characterized by motor dysfunction and degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNc). Protopanaxadiols (PPDs), including 20 (R)- protopanaxadiol (R-PPD) and 20 (S)- protopanaxadiol (S-PPD), are main metabolites of ginsenosides. The role of ginsenosides in neurodegenerative diseases has been thoroughly studied, however, it is unknown whether PPDs can attenuate behavioral deficits and dopaminergic neuron injury in PD model mice to date. Here, we administered PPDs to MPTP-induced PD model mice and monitored the effects on behavior and dopaminergic neurons to investigate the effects of R-PPD and S-PPD against PD. Our results showed that R-PPD and S-PPD (at a dose of 20 mg/kg, i.g.) treatment alleviated MPTP (30 mg/kg, i.p.) induced behavioral deficits. Besides, R-PPD and S-PPD protected MPP+-induced neuron injury and mitochondrial dysfunction, and reduced the abnormal expression of Cyt C, Bax, caspase-3 and Bcl-2. These findings demonstrate that R-PPD and S-PPD were potentially useful to ameliorate PD.
Collapse
Affiliation(s)
- Jindong Zhao
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, People's Republic of China
- College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, People's Republic of China
| | - Ji Wang
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, People's Republic of China
- School of Chinese Materia Medica &Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, People's Republic of China
| | - Kunying Zhao
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, People's Republic of China
- College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, People's Republic of China
| | - Yuxiao Zhang
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, People's Republic of China
- College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, People's Republic of China
| | - Weiyan Hu
- School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, People's Republic of China.
- College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, People's Republic of China.
| |
Collapse
|
5
|
Wei SJ, Schell JR, Chocron ES, Varmazyad M, Xu G, Chen WH, Martinez GM, Dong FF, Sreenivas P, Trevino R, Jiang H, Du Y, Saliba A, Qian W, Lorenzana B, Nazarullah A, Chang J, Sharma K, Munkácsy E, Horikoshi N, Gius D. Ketogenic diet induces p53-dependent cellular senescence in multiple organs. SCIENCE ADVANCES 2024; 10:eado1463. [PMID: 38758782 PMCID: PMC11100565 DOI: 10.1126/sciadv.ado1463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/12/2024] [Indexed: 05/19/2024]
Abstract
A ketogenic diet (KD) is a high-fat, low-carbohydrate diet that leads to the generation of ketones. While KDs improve certain health conditions and are popular for weight loss, detrimental effects have also been reported. Here, we show mice on two different KDs and, at different ages, induce cellular senescence in multiple organs, including the heart and kidney. This effect is mediated through adenosine monophosphate-activated protein kinase (AMPK) and inactivation of mouse double minute 2 (MDM2) by caspase-2, leading to p53 accumulation and p21 induction. This was established using p53 and caspase-2 knockout mice and inhibitors to AMPK, p21, and caspase-2. In addition, senescence-associated secretory phenotype biomarkers were elevated in serum from mice on a KD and in plasma samples from patients on a KD clinical trial. Cellular senescence was eliminated by a senolytic and prevented by an intermittent KD. These results have important clinical implications, suggesting that the effects of a KD are contextual and likely require individual optimization.
Collapse
Affiliation(s)
- Sung-Jen Wei
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - Joseph R. Schell
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - E. Sandra Chocron
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - Mahboubeh Varmazyad
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - Guogang Xu
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - Wan Hsi Chen
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - Gloria M. Martinez
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - Felix F. Dong
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - Prethish Sreenivas
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - Rolando Trevino
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - Haiyan Jiang
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - Yan Du
- Center for Precision Medicine, UT Health San Antonio, San Antonio, TX, USA
- School of Nursing, UT Health San Antonio, San Antonio, TX, USA
| | - Afaf Saliba
- Center for Precision Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Wei Qian
- Houston Methodist Cancer Center, Houston, TX, USA
- Houston Methodist Research Institute, Houston, TX, USA
| | - Brandon Lorenzana
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - Alia Nazarullah
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - Jenny Chang
- Houston Methodist Cancer Center, Houston, TX, USA
- Houston Methodist Research Institute, Houston, TX, USA
| | - Kumar Sharma
- Center for Precision Medicine, UT Health San Antonio, San Antonio, TX, USA
- Division of Nephrology, Department of Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Erin Munkácsy
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - Nobuo Horikoshi
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| | - David Gius
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, San Antonio, TX, USA
- Barshop Institute for Longevity and Aging Studies at UT Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
6
|
Wang C, Wen L, Wang K, Wu R, Li M, Zhang Y, Gao Z. Visualization of ferroptosis in brain diseases and ferroptosis-inducing nanomedicine for glioma. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2023; 13:179-194. [PMID: 38023817 PMCID: PMC10656630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 09/08/2023] [Indexed: 12/01/2023]
Abstract
A remarkable body of new data establishes that many degenerative brain diseases and some acute injury situations in the brain may be associated with ferroptosis. In recent years, ferroptosis has also attracted great interest in the cancer research community, partly because it is a unique mode of cell death distinct from other forms and thus has great therapeutic potential for brain cancer. Glioblastoma is a highly aggressive and fatal human cancer, accounting for 60% of all primary brain tumors. Despite the development of various pharmacological and surgical modalities, the survival rates of high-grade gliomas have remained poor over the past few decades. Recent evidence has revealed that ferroptosis is involved in tumor initiation, progression, and metastasis, and manipulating ferroptosis could offer a novel strategy for glioma management. Nanoparticles have been exploited as multifunctional platforms that can cross the blood-brain barrier and deliver therapeutic agents to the brain to address the pressing need for accurate visualization of ferroptosis and glioma treatment. To create efficient and durable ferroptosis inducers, many researchers have engineered nanocomposites to induce a more effective ferroptosis for therapy. In this review, we present the mechanism of ferroptosis and outline the current strategies of imaging and nanotherapy of ferroptosis in brain diseases, especially glioma. We aim to provide up-to-date information on ferroptosis and emphasize the potential clinical implications of ferroptosis for glioma diagnosis and treatment. However, regulation of ferroptosis in vivo remains challenging due to a lack of compounds.
Collapse
Affiliation(s)
- Chenyang Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
- Hubei Key Laboratory of Molecular ImagingWuhan 430022, Hubei, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of EducationWuhan 430022, Hubei, China
| | - Li Wen
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
- Hubei Key Laboratory of Molecular ImagingWuhan 430022, Hubei, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of EducationWuhan 430022, Hubei, China
| | - Kun Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
- Hubei Key Laboratory of Molecular ImagingWuhan 430022, Hubei, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of EducationWuhan 430022, Hubei, China
| | - Ruolin Wu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
- Hubei Key Laboratory of Molecular ImagingWuhan 430022, Hubei, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of EducationWuhan 430022, Hubei, China
| | - Mengting Li
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
- Hubei Key Laboratory of Molecular ImagingWuhan 430022, Hubei, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of EducationWuhan 430022, Hubei, China
| | - Yajing Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
- Hubei Key Laboratory of Molecular ImagingWuhan 430022, Hubei, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of EducationWuhan 430022, Hubei, China
| | - Zairong Gao
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, Hubei, China
- Hubei Key Laboratory of Molecular ImagingWuhan 430022, Hubei, China
- Key Laboratory of Biological Targeted Therapy, The Ministry of EducationWuhan 430022, Hubei, China
| |
Collapse
|
7
|
Lentini G, Famà A, De Gaetano GV, Coppolino F, Mahjoub AK, Ryan L, Lien E, Espevik T, Beninati C, Teti G. Caspase-8 inhibition improves the outcome of bacterial infections in mice by promoting neutrophil activation. Cell Rep Med 2023:101098. [PMID: 37390829 PMCID: PMC10394171 DOI: 10.1016/j.xcrm.2023.101098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/07/2023] [Accepted: 06/08/2023] [Indexed: 07/02/2023]
Abstract
During differentiation, neutrophils undergo a spontaneous pro-inflammatory program that is hypothesized here to be under caspase-8 control. In mice, intraperitoneal administration of the caspase-8 inhibitor z-IETD-fmk is sufficient to unleash the production of pro-inflammatory cytokines and neutrophil influx in the absence of cell death. These effects are due to selective inhibition of caspase-8 and require tonic interferon-β (IFN-β) production and RIPK3 but not MLKL, the essential downstream executioner of necroptotic cell death. In vitro, stimulation with z-IETD-fmk is sufficient to induce significant cytokine production in murine neutrophils but not in macrophages. Therapeutic administration of z-IETD-fmk improves clinical outcome in models of lethal bacterial peritonitis and pneumonia by augmenting cytokine release, neutrophil influx, and bacterial clearance. Moreover, the inhibitor protects mice against high-dose endotoxin shock. Collectively, our data unveil a RIPK3- and IFN-β-dependent pathway that is constitutively activated in neutrophils and can be harnessed therapeutically using caspase-8 inhibition.
Collapse
Affiliation(s)
- Germana Lentini
- Department of Human Pathology, University of Messina, Messina, Italy
| | - Agata Famà
- Department of Human Pathology, University of Messina, Messina, Italy
| | | | - Francesco Coppolino
- Department of Chemical, Biological and Pharmaceutical Sciences, University of Messina, Messina, Italy
| | | | - Liv Ryan
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Egil Lien
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Division of Infectious Diseases and Immunology, Program in Innate Immunity, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Terje Espevik
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Concetta Beninati
- Department of Human Pathology, University of Messina, Messina, Italy; Scylla Biotech Srl, Messina, Italy
| | | |
Collapse
|
8
|
Markowski AR, Żbikowski A, Zabielski P, Chlabicz U, Sadowska P, Pogodzińska K, Błachnio-Zabielska AU. The Effect of Silencing the Genes Responsible for the Level of Sphingosine-1-phosphate on the Apoptosis of Colon Cancer Cells. Int J Mol Sci 2023; 24:ijms24087197. [PMID: 37108361 PMCID: PMC10138425 DOI: 10.3390/ijms24087197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/10/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) and ceramides (Cer) are engaged in key events of signal transduction, but their involvement in the pathogenesis of colorectal cancer is not conclusive. The aim of our study was to investigate how the modulation of sphingolipid metabolism through the silencing of the genes involved in the formation (SPHK1) and degradation (SGPL1) of sphingosine-1-phosphate would affect the sphingolipid profile and apoptosis of HCT-116 human colorectal cancer cells. Silencing of SPHK1 expression decreased S1P content in HCT-116 cells, which was accompanied by an elevation in sphingosine, C18:0-Cer, and C18:1-Cer, increase in the expression and activation of Caspase-3 and -9, and augmentation of apoptosis. Interestingly, silencing of SGLP1 expression increased cellular content of both the S1P and Cer (C16:0-; C18:0-; C18:1-; C20:0-; and C22:0-Cer), yet inhibited activation of Caspase-3 and upregulated protein expression of Cathepsin-D. The above findings suggest that modulation of the S1P level and S1P/Cer ratio regulates both cellular apoptosis and CRC metastasis through Cathepsin-D modulation. The cellular ratio of S1P/Cer seems to be a crucial component of the above mechanism.
Collapse
Affiliation(s)
- Adam R Markowski
- Department of Internal Medicine and Gastroenterology, Polish Red Cross Memorial Municipal Hospital, 79 Henryk Sienkiewicz Street, 15-003 Bialystok, Poland
| | - Arkadiusz Żbikowski
- Department of Medical Biology, Medical University of Bialystok, 2C Adam Mickiewicz Street, 15-222 Bialystok, Poland
| | - Piotr Zabielski
- Department of Medical Biology, Medical University of Bialystok, 2C Adam Mickiewicz Street, 15-222 Bialystok, Poland
| | - Urszula Chlabicz
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, 2C Adam Mickiewicz Street, 15-222 Bialystok, Poland
| | - Patrycja Sadowska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, 2C Adam Mickiewicz Street, 15-222 Bialystok, Poland
| | - Karolina Pogodzińska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, 2C Adam Mickiewicz Street, 15-222 Bialystok, Poland
| | - Agnieszka U Błachnio-Zabielska
- Department of Hygiene, Epidemiology and Metabolic Disorders, Medical University of Bialystok, 2C Adam Mickiewicz Street, 15-222 Bialystok, Poland
| |
Collapse
|
9
|
Pockes S, Walters MA, Ashe KH. Targeting caspase-2 interactions with tau in Alzheimer's disease and related dementias. Transl Res 2023; 254:34-40. [PMID: 36343883 PMCID: PMC9991976 DOI: 10.1016/j.trsl.2022.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/26/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
Targeting amyloid-β plaques and tau tangles has failed to provide effective treatments for Alzheimer's disease and related dementias (ADRD). A more fruitful pathway to ADRD therapeutics may be the development of therapies that target common signaling pathways that disrupt synaptic connections and impede communication between neurons. In this review, we present our characterization of a signaling pathway common to several neurological diseases featuring dementia including Alzheimer's disease, frontotemporal dementia, Lewy body dementia, and Huntington's disease. This signaling pathway features the cleavage of tau by caspase-2 (Casp2) yielding Δtau314 (Casp2/tau/Δtau314). Through a not yet fully delineated mechanism, Δtau314 catalyzes the mislocalization and accumulation of tau to dendritic spines leading to the internalization of AMPA receptors and the concomitant weakening of synaptic transmission. Here, we review the accumulated evidence supporting Casp2 as a druggable target and its importance in ADRD. Additionally, we provide a brief overview of our initial medicinal chemistry explorations aimed at the preparation of novel, brain penetrant Casp2 inhibitors. We anticipate that this review will spark broader interest in Casp2 as a target for restoring synaptic dysfunction in ADRD.
Collapse
Affiliation(s)
- Steffen Pockes
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany; Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, Minnesota; Department of Neurology, University of Minnesota, Minneapolis, Minnesota.
| | - Michael A Walters
- Department of Medicinal Chemistry, Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, Minnesota.
| | - Karen H Ashe
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
10
|
Groborz KM, Kalinka M, Grzymska J, Kołt S, Snipas SJ, Poręba M. Selective chemical reagents to investigate the role of caspase 6 in apoptosis in acute leukemia T cells. Chem Sci 2023; 14:2289-2302. [PMID: 36873853 PMCID: PMC9977399 DOI: 10.1039/d2sc05827h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/02/2023] [Indexed: 01/04/2023] Open
Abstract
Activated effector caspases 3, 6 and 7 are responsible for cleaving a number of target substrates, leading to the ultimate destruction of cells via apoptosis. The functions of caspases 3 and 7 in apoptosis execution have been widely studied over the years with multiple chemical probes for both of these enzymes. In contrast, caspase 6 seems to be largely neglected when compared to the heavily studied caspases 3 and 7. Therefore, the development of new small-molecule reagents for the selective detection and visualization of caspase 6 activity can improve our understanding of molecular circuits of apoptosis and shed new light on how they intertwine with other types of programmed cell death. In this study, we profiled caspase 6 substrate specificity at the P5 position and discovered that, similar to caspase 2, caspase 6 prefers pentapeptide substrates over tetrapeptides. Based on these data, we developed a set of chemical reagents for caspase 6 investigation, including coumarin-based fluorescent substrates, irreversible inhibitors and selective aggregation-induced emission luminogens (AIEgens). We showed that AIEgens are able to distinguish between caspase 3 and caspase 6 in vitro. Finally, we validated the efficiency and selectivity of the synthesized reagents by monitoring lamin A and PARP cleavage via mass cytometry and western blot analysis. We propose that our reagents may provide new research prospects for single-cell monitoring of caspase 6 activity to reveal its function in programmed cell death pathways.
Collapse
Affiliation(s)
- Katarzyna M Groborz
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology Wyb. Wyspiańskiego 27 50-370 Wroclaw Poland
- Genetech Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Małgorzata Kalinka
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology Wyb. Wyspiańskiego 27 50-370 Wroclaw Poland
| | - Justyna Grzymska
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology Wyb. Wyspiańskiego 27 50-370 Wroclaw Poland
| | - Sonia Kołt
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology Wyb. Wyspiańskiego 27 50-370 Wroclaw Poland
| | - Scott J Snipas
- NCI Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute 10901 North Torrey Pines Road La Jolla CA 92037 USA
| | - Marcin Poręba
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology Wyb. Wyspiańskiego 27 50-370 Wroclaw Poland
| |
Collapse
|
11
|
Janiszewski T, Kołt S, Ciastoń I, Vizovisek M, Poręba M, Turk B, Drąg M, Kozieł J, Kasperkiewicz P. Investigation of osteoclast cathepsin K activity in osteoclastogenesis and bone loss using a set of chemical reagents. Cell Chem Biol 2023; 30:159-174.e8. [PMID: 36696904 DOI: 10.1016/j.chembiol.2023.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/28/2022] [Accepted: 01/04/2023] [Indexed: 01/26/2023]
Abstract
Cathepsin K (CatK) is a lysosomal cysteine protease whose highest expression is found in osteoclasts, which are the cells responsible for bone resorption. Investigations of the functions and physiological relevance of CatK have often relied on antibody-related techniques, which makes studying its activity patterns a challenging task. Hence, we developed a set of chemical tools for the investigation of CatK activity. We show that our probe is a valuable tool for monitoring the proteolytic activation of CatK during osteoclast formation. Moreover, we demonstrate that our inhibitor of CatK impedes osteoclastogenesis and bone resorption and that CatK is stored in its active form in osteoclasts within their lysosomal compartment and mainly in the ruffled borders of osteoclasts. Given that our probe recognizes active CatK within living cells without exhibiting any observed cytotoxicity in the several models tested, we expect that it would be well suited to theranostic applications in CatK-related diseases.
Collapse
Affiliation(s)
- Tomasz Janiszewski
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Sonia Kołt
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Izabela Ciastoń
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland
| | - Matej Vizovisek
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Marcin Poręba
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, 1000 Ljubljana, Slovenia
| | - Marcin Drąg
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Joanna Kozieł
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland
| | - Paulina Kasperkiewicz
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland.
| |
Collapse
|
12
|
Ćwilichowska N, Świderska KW, Dobrzyń A, Drąg M, Poręba M. Diagnostic and therapeutic potential of protease inhibition. Mol Aspects Med 2022; 88:101144. [PMID: 36174281 DOI: 10.1016/j.mam.2022.101144] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 07/20/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022]
Abstract
Proteases are enzymes that hydrolyze peptide bonds in proteins and peptides; thus, they control virtually all biological processes. Our understanding of protease function has advanced considerably from nonselective digestive enzymes to highly specialized molecular scissors that orchestrate complex signaling networks through a limited proteolysis. The catalytic activity of proteases is tightly regulated at several levels, ranging from gene expression through trafficking and maturation to posttranslational modifications. However, when this delicate balance is disturbed, many diseases develop, including cancer, inflammatory disorders, diabetes, and neurodegenerative diseases. This new understanding of the role of proteases in pathologic physiology indicates that these enzymes represent excellent molecular targets for the development of therapeutic inhibitors, as well as for the design of chemical probes to visualize their redundant activity. Recently, numerous platform technologies have been developed to identify and optimize protease substrates and inhibitors, which were further used as lead structures for the development of chemical probes and therapeutic drugs. Due to this considerable success, the clinical potential of proteases in therapeutics and diagnostics is rapidly growing and is still not completely explored. Therefore, small molecules that can selectively target aberrant protease activity are emerging in diseases cells. In this review, we describe modern trends in the design of protease drugs as well as small molecule activity-based probes to visualize selected proteases in clinical settings.
Collapse
Affiliation(s)
- Natalia Ćwilichowska
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Karolina W Świderska
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Agnieszka Dobrzyń
- Nencki Institute of Experimental Biology, Ludwika Pasteura 3, 02-093, Warsaw, Poland
| | - Marcin Drąg
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland.
| | - Marcin Poręba
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland.
| |
Collapse
|
13
|
Li C, Salmen SH, Awad Alahmadi T, Priya Veeraraghavan V, Krishna Mohan S, Natarajan N, Subramanian S. Anticancer effect of Selenium/Chitosan/Polyethylene glycol/Allyl isothiocyanate nanocomposites against diethylnitrosamine-induced liver cancer in rats. Saudi J Biol Sci 2022; 29:3354-3365. [PMID: 35844425 PMCID: PMC9280227 DOI: 10.1016/j.sjbs.2022.02.012] [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: 01/19/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Background Nano-based drug delivery systems have shown several advantages in cancer treatment like specific targeting of cancer cells, good pharmacokinetics, and lesser adverse effects. Liver cancer is a fifth most common cancer and third leading cause of cancer-related mortalities worldwide. Objective The present study focusses to formulate the selenium (S)/chitosan (C)/polyethylene glycol (Pg)/allyl isothiocyanate (AI) nanocomposites (SCPg-AI-NCs) and assess its therapeutic properties against the diethylnitrosamine (DEN)-induced liver cancer in rats via inhibition of oxidative stress and tumor markers. Methodology The SCPg-AI-NCs were synthesized by ionic gelation technique and characterized by various characterization techniques. The liver cancer was induced to the rats by injecting a DEN (200 mg/kg) on the 8th day of experiment. Then DEN-induced rats treated with 10 mg/kg of formulated SCPg-AI-NCs an hour before DEN administration for 16 weeks. The 8-hydroxy-2′ -deoxyguanosine (8-OHdG) content, albumin, globulin, and total protein were examined by standard methods. The level of glutathione (GSH), vitamin-C & -E, and superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and glutathione reductase (GR) activities were examined using assay kits. The liver marker enzymes i.e., alanine transaminase (ALT), aspartate tansaminase (AST), γ-glutamyl transaminase (GGT), lactate dehydrogenase (LDH), and alkaline phosphatase (ALP) activities, alpha fetoprotein (AFP) and carcinoembryonic antigen (CEA), Bax, and Bcl-2 levels, and caspase-3&9 activities was examined using assay kits and the liver histopathology was assessed microscopically by hematoxylin and eosin staining method. The effect of formulated SCPg-AI-NCs on the viability and apoptotic cell death on the HepG2 cells were examined using MTT and dual staining assays, respectively. Results The results of different characterization studies demonstrated the formation of SCPg-AI-NCs with tetragonal shape, narrowed distribution, and size ranging from 390 to 450 nm. The formulated SCPg-AI-NCs treated liver cancer rats indicated the reduced levels of 8-OHdG, albumin, globulin, and total protein. The SCPg-AI-NCs treatment appreciably improved the GSH, vitamin-C & -E contents, and SOD, CAT, GPx, and GR activities in the serum of liver cancer rats. The SCPg-AI-NCs treatment remarkably reduced the liver marker enzyme activities in the DEN-induced rats. The SCPg-AI-NCs treatment decreased the AFP and CEA contents and enhanced the Bax and caspase 3&9 activities in the DEN-induced rats. The SCPg-AI-NCs effectively decreased the cell viability and induced apoptosis in the HepG2 cells. Conclusion The present findings suggested that the formulated SCPg-AI-NCs remarkably inhibited the DEN-induced liver carcinogenesis in rats. These findings provide an evidence that SCPg-AI-NCs can be a promising anticancer nano-drug in the future to treat the liver carcinogenesis.
Collapse
|
14
|
The roles of cellular protease interactions in viral infections and programmed cell death: a lesson learned from the SARS-CoV-2 outbreak and COVID-19 pandemic. Pharmacol Rep 2022; 74:1149-1165. [PMID: 35997950 PMCID: PMC9395814 DOI: 10.1007/s43440-022-00394-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/03/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022]
Abstract
The unprecedented pandemic of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), which leads to COVID-19, is threatening global health. Over the last 2 years, we have witnessed rapid progress in research focusing on developing new antiviral vaccines and drugs, as well as in academic and clinical efforts to understand the biology and pathology of COVID-19. The roles of proteases among master regulators of SARS-CoV-2 invasion and replication and their pivotal roles in host defence against this pathogen, including programmed cell death, have not been well established. Our understanding of protease function in health and disease has increased considerably over the last two decades, with caspases, matrix metalloproteases, and transmembrane serine proteases representing the most prominent examples. Therefore, during the COVID-19 pandemic, these enzymes have been investigated as potential molecular targets for therapeutic interventions. Proteases that are responsible for SARS-CoV-2 cell entry and replication, such as TMPRSS2, ACE2 or cathepsins, are screened with inhibitor libraries to discover lead structures for further drug design that would prevent virus multiplication. On the other hand, proteases that orchestrate programmed cell death can also be harnessed to enhance the desired demise of infected cells through apoptosis or to attenuate highly inflammatory lytic cell death that leads to undesired cytokine storms, a major hallmark of severe COVID-19. Given the prominent role of proteases in SARS-CoV-2-induced cell death, we discuss the individual roles of these enzymes and their catalytic interactions in the pathology of COVID-19 in this article. We provide a rationale for targeting proteases participating in cell death as potential COVID-19 treatments and identify knowledge gaps that might be investigated to better understand the mechanism underlying SARS-CoV-2-induced cell death.
Collapse
|
15
|
Lovell S, Zhang L, Kryza T, Neodo A, Bock N, De Vita E, Williams ED, Engelsberger E, Xu C, Bakker AT, Maneiro M, Tanaka RJ, Bevan CL, Clements JA, Tate EW. A Suite of Activity-Based Probes To Dissect the KLK Activome in Drug-Resistant Prostate Cancer. J Am Chem Soc 2021; 143:8911-8924. [PMID: 34085829 PMCID: PMC9282638 DOI: 10.1021/jacs.1c03950] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
![]()
Kallikrein-related
peptidases (KLKs) are a family of secreted serine
proteases, which form a network (the KLK activome) with an important
role in proteolysis and signaling. In prostate cancer (PCa), increased
KLK activity promotes tumor growth and metastasis through multiple
biochemical pathways, and specific quantification and tracking of
changes in the KLK activome could contribute to validation of KLKs
as potential drug targets. Herein we report a technology platform
based on novel activity-based probes (ABPs) and inhibitors enabling
simultaneous orthogonal analysis of KLK2, KLK3, and KLK14 activity
in hormone-responsive PCa cell lines and tumor homogenates. Importantly,
we identifed a significant decoupling of KLK activity and abundance
and suggest that KLK proteolysis should be considered as an additional
parameter, along with the PSA blood test, for accurate PCa diagnosis
and monitoring. Using selective inhibitors and multiplexed fluorescent
activity-based protein profiling (ABPP), we dissect the KLK activome
in PCa cells and show that increased KLK14 activity leads to a migratory
phenotype. Furthermore, using biotinylated ABPs, we show that active
KLK molecules are secreted into the bone microenvironment by PCa cells
following stimulation by osteoblasts suggesting KLK-mediated signaling
mechanisms could contribute to PCa metastasis to bone. Together our
findings show that ABPP is a powerful approach to dissect dysregulation
of the KLK activome as a promising and previously underappreciated
therapeutic target in advanced PCa.
Collapse
Affiliation(s)
- Scott Lovell
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Leran Zhang
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Thomas Kryza
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.,Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Anna Neodo
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Nathalie Bock
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Elena De Vita
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Elizabeth D Williams
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Elisabeth Engelsberger
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Congyi Xu
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Alexander T Bakker
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Maria Maneiro
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Reiko J Tanaka
- Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K
| | - Charlotte L Bevan
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, U.K
| | - Judith A Clements
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Edward W Tate
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.,The Francis Crick Institute, London NW1 1AT, U.K
| |
Collapse
|
16
|
Zainodini N, Hajizadeh MR, Mirzaei MR. Evaluation of Apoptotic Gene Expression in Hepatoma Cell Line (HepG2) Following Nisin Treatment. Asian Pac J Cancer Prev 2021; 22:1413-1419. [PMID: 34048169 PMCID: PMC8408378 DOI: 10.31557/apjcp.2021.22.5.1413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 01/10/2023] Open
Abstract
Objective: The present study aims to examine the effects of nisin on the survival and apoptosis of the hepatoma cell line HepG2 and to investigate possible apoptosis pathways activated by nisin. Materials and Methods: For this purpose, viability and apoptosis of the cells were accomplished by the nisin treatment using the MTT assay and Annexin-V-fluorescein/propidium iodide (PI) double staining, respectively. Additionally, the human apoptosis PCR array was performed to determine pathways or genes activated by nisin during possible apoptosis. Results: The results of the present study showed that nisin was able to decrease cell viability (IC50 ~ 40 µg/ml) in a dose-dependent manner and could induce apoptosis in HepG2 cells. PCR data indicated a considerable increase in the expression of genes, such as caspase and BCL2 families, involved in the induction of apoptosis. Conclusions: The data from this study showed that overexpression of genes involved in the intrinsic pathway of apoptosis, especially caspase-9 and BID, increased apoptosis in HepG2 cells treated by nisin, compared to the control group.
Collapse
Affiliation(s)
- Nahid Zainodini
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Reza Hajizadeh
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Reza Mirzaei
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
17
|
Kasperkiewicz P. Peptidyl Activity-Based Probes for Imaging Serine Proteases. Front Chem 2021; 9:639410. [PMID: 33996745 PMCID: PMC8117214 DOI: 10.3389/fchem.2021.639410] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/15/2021] [Indexed: 01/12/2023] Open
Abstract
Proteases catalyze the hydrolysis of peptide bonds. Products of this breakdown mediate signaling in an enormous number of biological processes. Serine proteases constitute the most numerous group of proteases, accounting for 40%, and they are prevalent in many physiological functions, both normal and disease-related functions, making them one of the most important enzymes in humans. The activity of proteases is controlled at the expression level by posttranslational modifications and/or endogenous inhibitors. The study of serine proteases requires specific reagents not only for detecting their activity but also for their imaging. Such tools include inhibitors or substrate-related chemical molecules that allow the detection of proteolysis and visual observation of active enzymes, thus facilitating the characterization of the activity of proteases in the complex proteome. Peptidyl activity-based probes (ABPs) have been extensively studied recently, and this review describes the basic principles in the design of peptide-based imaging agents for serine proteases, provides examples of activity-based probe applications and critically discusses their strengths, weaknesses, challenges and limitations.
Collapse
Affiliation(s)
- Paulina Kasperkiewicz
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| |
Collapse
|
18
|
Kim HR, Tagirasa R, Yoo E. Covalent Small Molecule Immunomodulators Targeting the Protease Active Site. J Med Chem 2021; 64:5291-5322. [PMID: 33904753 DOI: 10.1021/acs.jmedchem.1c00172] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cells of the immune system utilize multiple proteases to regulate cell functions and orchestrate innate and adaptive immune responses. Dysregulated protease activities are implicated in many immune-related disorders; thus, protease inhibitors have been actively investigated for pharmaceutical development. Although historically considered challenging with concerns about toxicity, compounds that covalently modify the protease active site represent an important class of agents, emerging not only as chemical probes but also as approved drugs. Here, we provide an overview of technologies useful for the study of proteases with the focus on recent advances in chemoproteomic methods and screening platforms. By highlighting covalent inhibitors that have been designed to target immunomodulatory proteases, we identify opportunities for the development of small molecule immunomodulators.
Collapse
Affiliation(s)
- Hong-Rae Kim
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ravichandra Tagirasa
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Euna Yoo
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
19
|
Vizovisek M, Ristanovic D, Menghini S, Christiansen MG, Schuerle S. The Tumor Proteolytic Landscape: A Challenging Frontier in Cancer Diagnosis and Therapy. Int J Mol Sci 2021; 22:ijms22052514. [PMID: 33802262 PMCID: PMC7958950 DOI: 10.3390/ijms22052514] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
In recent decades, dysregulation of proteases and atypical proteolysis have become increasingly recognized as important hallmarks of cancer, driving community-wide efforts to explore the proteolytic landscape of oncologic disease. With more than 100 proteases currently associated with different aspects of cancer development and progression, there is a clear impetus to harness their potential in the context of oncology. Advances in the protease field have yielded technologies enabling sensitive protease detection in various settings, paving the way towards diagnostic profiling of disease-related protease activity patterns. Methods including activity-based probes and substrates, antibodies, and various nanosystems that generate reporter signals, i.e., for PET or MRI, after interaction with the target protease have shown potential for clinical translation. Nevertheless, these technologies are costly, not easily multiplexed, and require advanced imaging technologies. While the current clinical applications of protease-responsive technologies in oncologic settings are still limited, emerging technologies and protease sensors are poised to enable comprehensive exploration of the tumor proteolytic landscape as a diagnostic and therapeutic frontier. This review aims to give an overview of the most relevant classes of proteases as indicators for tumor diagnosis, current approaches to detect and monitor their activity in vivo, and associated therapeutic applications.
Collapse
|
20
|
Li Q, Shi N, Cai C, Zhang M, He J, Tan Y, Fu W. The Role of Mitochondria in Pyroptosis. Front Cell Dev Biol 2021; 8:630771. [PMID: 33553170 PMCID: PMC7859326 DOI: 10.3389/fcell.2020.630771] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/28/2020] [Indexed: 11/25/2022] Open
Abstract
Pyroptosis is a recently discovered aspartic aspart-specific cysteine protease (Caspase-1/4/5/11) dependent mode of gene-regulated cell death cell death, which is represented by the rupture of cell membrane perforations and the production of proinflammatory mediaters like interleukin-18(IL-18) and interleukin-1β (IL-1β). Mitochondria also play an important role in apoptotic cell death. When it comes to apoptosis of mitochondrion, mitochondrial outer membrane permeabilization (MOMP) is commonly known to cause cell death. As a downstream pathological process of apoptotic signaling, MOMP participates in the leakage of cytochrome-c from mitochondrion to the cytosol and subsequently activate caspase proteases. Hence, targeting MOMP for the sake of manipulating cell death presents potential therapeutic effects among various types of diseases, such as autoimmune disorders, neurodegenerative diseases, and cancer. In this review, we highlights the roles and significance of mitochondria in pyroptosis to provide unexplored strategies that target the mitochondria to regulate cell death for clinical benefits.
Collapse
Affiliation(s)
- Qian Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Nengxian Shi
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chen Cai
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mingming Zhang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing He
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Tan
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weijun Fu
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
21
|
Thermococcus kodakarensis-derived L-asparaginase: a candidate for the treatment of glioblastoma. Biologia (Bratisl) 2021. [DOI: 10.2478/s11756-021-00678-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Hong W, Gu Y, Guan R, Xie D, Zhou H, Yu M. Pan-cancer analysis of the CASP gene family in relation to survival, tumor-infiltrating immune cells and therapeutic targets. Genomics 2020; 112:4304-4315. [PMID: 32682809 DOI: 10.1016/j.ygeno.2020.07.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/04/2020] [Accepted: 07/13/2020] [Indexed: 01/04/2023]
Abstract
The cysteinyl aspartate protease (caspase, or CASP) gene family plays a significant role in programmed cell death, inflammation and immunity. However, the correlation between CASP family members and prognosis and tumor-infiltrating lymphocytes in different tumors has not been determined. We investigated the role of CASP genes in cancer prognosis and their relationship with clinicopathological parameters. We also evaluated the correlation between the expression of CASP family members and cancer immune infiltration and evaluated whether these molecules can be used as targets for immunotherapy. The CASP1/2/4/5/7/9 genes may represent prognostic factors and therapeutic targets for breast cancer, hepatocellular carcinoma and pancreatic cancer. Another finding is that the CASP1/4/5 genes help to regulate innate immunity and T cell immunity and may also have an important effect on tumor checkpoint inhibition. These findings may elucidate the roles played by CASP family members in cancer progression and identify strategies to promote collaborative activities in the context of immunotherapy.
Collapse
Affiliation(s)
- Weifeng Hong
- Department of Medical Imaging, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Morning Star Academic Cooperation, Shanghai, China
| | - YuJun Gu
- Department of Ultrasonic Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - RenGuo Guan
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Daipeng Xie
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Haiyu Zhou
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Min Yu
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
23
|
Bin J, Bai T, Zhao Q, Duan X, Deng S, Xu Y. Parkin overexpression reduces inflammation-mediated cardiomyocyte apoptosis through activating Nrf2/ARE signaling pathway. J Recept Signal Transduct Res 2020; 41:451-456. [PMID: 33012239 DOI: 10.1080/10799893.2020.1825488] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Inflammation has been acknowledged as one of the pathological alterations in various cardiovascular disorders. Parkin has been found to be associated with mitochondrial protection. In the present study, we explored the influence of Parkin overexpression on cardiomyocyte induced by LPS-mediated inflammation response. Our results demonstrated that cardiomyocyte viability was reduced and apoptotic rate was increased upon LPS treatment, an effect that may be caused by cardiomyocyte oxidative stress. At the molecular levels, LPS treatment promoted ROS production, a result that was followed by a drop in the levels of anti-oxidants. Interestingly, Parkin overexpression significantly promoted cardiomyocyte survival and this cardioprotective was attributable to the anti-oxidative property. Parkin overexpression enhanced the expression of anti-oxidative factors such as GSH, SOD and GPX, resulting into depressed ROS production. Further, we found that Parkin modulated cellular anti-oxidative capacity through the Nrf2/ARE signaling pathway. This finding demonstrates that oxidative stress could be considered as the core of inflammation response. Further, therapeutic approaches targeting Parkin would improve cardiomyocyte anti-oxidative capacity through activating Nrf2/ARE signaling pathway.
Collapse
Affiliation(s)
- Jianguo Bin
- Department of Cardiology, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), Hengyang, China
| | - Taizhu Bai
- Department of Cardiology, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), Hengyang, China
| | - Qingxi Zhao
- Department of Cardiology, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), Hengyang, China
| | - Xiaohua Duan
- Department of Cardiology, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), Hengyang, China
| | - Suxin Deng
- Department of Cardiology, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), Hengyang, China
| | - Yunjun Xu
- Department of Cardiology, Affiliated Hengyang Hospital, Southern Medical University (Hengyang Central Hospital), Hengyang, China
| |
Collapse
|
24
|
Poreba M, Groborz KM, Rut W, Pore M, Snipas SJ, Vizovisek M, Turk B, Kuhn P, Drag M, Salvesen GS. Multiplexed Probing of Proteolytic Enzymes Using Mass Cytometry-Compatible Activity-Based Probes. J Am Chem Soc 2020; 142:16704-16715. [PMID: 32870676 PMCID: PMC7595764 DOI: 10.1021/jacs.0c06762] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The subset of the proteome that contains enzymes in their catalytically active form can be interrogated by using probes targeted toward individual specific enzymes. A subset of such enzymes are proteases that are frequently studied with activity-based probes, small inhibitors equipped with a detectable tag, commonly a fluorophore. Due to the spectral overlap of these commonly used fluorophores, multiplex analysis becomes limited. To overcome this, we developed a series of protease-selective lanthanide-labeled probes compatible with mass cytometry giving us the ability to monitor the activity of multiple proteases in parallel. Using these probes, we were able to identify the distribution of four proteases with different active site geometries in three cell lines and peripheral blood mononuclear cells. This provides a framework for the use of mass cytometry for multiplexed enzyme activity detection.
Collapse
Affiliation(s)
- Marcin Poreba
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Katarzyna M. Groborz
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Wioletta Rut
- Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Milind Pore
- University of Southern California, USC Michelson Center for Convergent Biosciences, Los Angeles, CA, USA
| | - Scott J. Snipas
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Boris Turk
- Jozef Stefan Institute, Ljubljana, Slovenia
| | - Peter Kuhn
- University of Southern California, USC Michelson Center for Convergent Biosciences, Los Angeles, CA, USA
| | - Marcin Drag
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Guy S. Salvesen
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
25
|
Chen S, Yim JJ, Bogyo M. Synthetic and biological approaches to map substrate specificities of proteases. Biol Chem 2020; 401:165-182. [PMID: 31639098 DOI: 10.1515/hsz-2019-0332] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 10/11/2019] [Indexed: 02/07/2023]
Abstract
Proteases are regulators of diverse biological pathways including protein catabolism, antigen processing and inflammation, as well as various disease conditions, such as malignant metastasis, viral infection and parasite invasion. The identification of substrates of a given protease is essential to understand its function and this information can also aid in the design of specific inhibitors and active site probes. However, the diversity of putative protein and peptide substrates makes connecting a protease to its downstream substrates technically difficult and time-consuming. To address this challenge in protease research, a range of methods have been developed to identify natural protein substrates as well as map the overall substrate specificity patterns of proteases. In this review, we highlight recent examples of both synthetic and biological methods that are being used to define the substrate specificity of protease so that new protease-specific tools and therapeutic agents can be developed.
Collapse
Affiliation(s)
- Shiyu Chen
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joshua J Yim
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
26
|
Orning P, Lien E. Multiple roles of caspase-8 in cell death, inflammation, and innate immunity. J Leukoc Biol 2020; 109:121-141. [PMID: 32531842 DOI: 10.1002/jlb.3mr0420-305r] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/16/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
Caspase-8 is an apical caspase involved in the programmed form of cell death called apoptosis that is critically important for mammalian development and immunity. Apoptosis was historically described as immunologically silent in contrast to other types of programmed cell death such as necroptosis or pyroptosis. Recent reports suggest considerable crosstalk between these different forms of cell death. It is becoming increasingly clear that caspase-8 has many non-apoptotic roles, participating in multiple processes including regulation of necroptosis (mediated by receptor-interacting serine/threonine kinases, RIPK1-RIPK3), inflammatory cytokine expression, inflammasome activation, and cleavage of IL-1β and gasdermin D, and protection against shock and microbial infection. In this review, we discuss the involvement of caspase-8 in cell death and inflammation and highlight its role in innate immune responses and in the relationship between different forms of cell death. Caspase-8 is one of the central components in this type of crosstalk.
Collapse
Affiliation(s)
- Pontus Orning
- UMass Medical School, Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, Worcester, Massachusetts, USA.,Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Egil Lien
- UMass Medical School, Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, Worcester, Massachusetts, USA.,Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
27
|
Sladky VC, Villunger A. Uncovering the PIDDosome and caspase-2 as regulators of organogenesis and cellular differentiation. Cell Death Differ 2020; 27:2037-2047. [PMID: 32415279 PMCID: PMC7308375 DOI: 10.1038/s41418-020-0556-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 02/08/2023] Open
Abstract
The PIDDosome is a multiprotein complex that drives activation of caspase-2, an endopeptidase originally implicated in apoptosis. Yet, unlike other caspases involved in cell death and inflammation, caspase-2 seems to exert additional versatile functions unrelated to cell death. These emerging roles range from control of transcription factor activity to ploidy surveillance. Thus, caspase-2 and the PIDDosome act as a critical regulatory unit controlling cellular differentiation processes during organogenesis and regeneration. These newly established functions of the PIDDosome and its downstream effector render its components attractive targets for drug-development aiming to prevent fatty liver diseases, neurodegenerative disorders or osteoporosis. ![]()
Collapse
Affiliation(s)
- Valentina C Sladky
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Andreas Villunger
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria. .,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, 1090, Vienna, Austria. .,CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria.
| |
Collapse
|
28
|
Poreba M. Protease-activated prodrugs: strategies, challenges, and future directions. FEBS J 2020; 287:1936-1969. [PMID: 31991521 DOI: 10.1111/febs.15227] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/14/2020] [Accepted: 01/23/2020] [Indexed: 02/06/2023]
Abstract
Proteases play critical roles in virtually all biological processes, including proliferation, cell death and survival, protein turnover, and migration. However, when dysregulated, these enzymes contribute to the progression of multiple diseases, with cancer, neurodegenerative disorders, inflammation, and blood disorders being the most prominent examples. For a long time, disease-associated proteases have been used for the activation of various prodrugs due to their well-characterized catalytic activity and ability to selectively cleave only those substrates that strictly correspond with their active site architecture. To date, versatile peptide sequences that are cleaved by proteases in a site-specific manner have been utilized as bioactive linkers for the targeted delivery of multiple types of cargo, including fluorescent dyes, photosensitizers, cytotoxic drugs, antibiotics, and pro-antibodies. This platform is highly adaptive, as multiple protease-labile conjugates have already been developed, some of which are currently in clinical use for cancer treatment. In this review, recent advancements in the development of novel protease-cleavable linkers for selective drug delivery are described. Moreover, the current limitations regarding the selectivity of linkers are discussed, and the future perspectives that rely on the application of unnatural amino acids for the development of highly selective peptide linkers are also presented.
Collapse
Affiliation(s)
- Marcin Poreba
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Poland
| |
Collapse
|
29
|
Li P, Wang J, Zhao X, Ru J, Tian T, An Y, Tang L, Bai Y. PTEN inhibition attenuates endothelial cell apoptosis in coronary heart disease via modulating the AMPK-CREB-Mfn2-mitophagy signaling pathway. J Cell Physiol 2020; 235:4878-4889. [PMID: 31654396 DOI: 10.1002/jcp.29366] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 10/07/2019] [Indexed: 12/11/2022]
Abstract
Atherosclerosis (AS) is a major pathogenic factor in patients with cardiovascular diseases, and endothelial dysfunction (ED) plays a primary role in the occurrence and development of AS. In our study, we attempted to evaluate the role of phosphatase and tensin homolog (PTEN) in endothelial cell apoptosis under oxidized low-density lipoprotein (ox-LDL) stimulation and identify the associated mechanisms. The results of our study demonstrated that ox-LDL induced human umbilical vein endothelial cell (HUVEC) death via mitochondrial apoptosis, as evidenced by reduced mitochondrial potential, increased mitochondria permeability transition pore opening, cellular calcium overload, and caspase-9/-3 activation. In addition, ox-LDL also suppressed cellular energy production via downregulating the mitochondrial respiratory complex. Moreover, ox-LDL impaired HUVECs migration. Western blot analysis showed that PTEN expression was upregulated after exposure to ox-LDL and knockdown of PTEN could attenuate ox-LDL-mediated endothelial cell damage. Furthermore, we found that ox-LDL impaired mitophagy activity, whereas PTEN deletion could improve mitophagic flux and this effect relied on the activity of the AMP-activated protein kinase (AMPK)-cAMP-response element-binding protein (CREB)-Mitofusin-2 (Mfn2) axis. When the AMPK-CREB-Mfn2 pathway was inhibited, PTEN deletion-associated HUVECs protection was significantly reduced, suggesting that the AMPK-CREB-Mfn2-mitophagy axis is required for PTEN deletion-mediated endothelial cell survival under ox-LDL. Taken together, our results indicate that ox-LDL-induced endothelial cell damage is associated with PTEN overexpression, and inhibition of PTEN could promote endothelial survival via activating the AMPK-CREB-Mfn2-mitophagy signaling pathway.
Collapse
Affiliation(s)
- Pei Li
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Jing Wang
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xia Zhao
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Jing Ru
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Tian Tian
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yun An
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Liying Tang
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yuzhi Bai
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Tian Y, Tang L, Yi P, Pan Q, Han Y, Shi Y, Rao S, Tan S, Xia L, Lin J, Oyang L, Tang Y, Liang J, Luo X, Liao Q, Wang H, Zhou Y. MiRNAs in Radiotherapy Resistance of Nasopharyngeal Carcinoma. J Cancer 2020; 11:3976-3985. [PMID: 32328201 PMCID: PMC7171507 DOI: 10.7150/jca.42734] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/27/2020] [Indexed: 02/06/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is one of the most common malignant tumors of the head and neck in Southeast Asia and southern China. Although the comprehensive treatment based on intensity-modulated radiation therapy improves outcomes, the five-year survival rate of NPC patients is low, and the recurrence remains high. Radiotherapy resistance is the main cause of poor prognosis in NPC patients. MicroRNAs (miRNAs) are a class of endogenous non-coding RNAs regulating various biological functions in eukaryotes. These miRNAs can regulate the development and progression of nasopharyngeal carcinoma by affecting the proliferation, apoptosis, movement, invasion and metastasis of NPC cells. The abnormal expression of miRNAs is closely related to radiotherapy sensitivity and prognosis of NPC patients, which can affect the transmission of related signaling pathways by regulating the expression of tumor suppressor genes and / or oncogenes, and therefore participate in radiotherapy resistance in nasopharyngeal carcinoma. Here, we review the mechanisms by which miRNAs may be involved in the radiotherapy resistance of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Yutong Tian
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Lu Tang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Pin Yi
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Qing Pan
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China.,University of South China, Hengyang, 421001, Hunan, China
| | - Yaqian Han
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China
| | - Yingrui Shi
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China
| | - Shan Rao
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China
| | - Shiming Tan
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China
| | - Longzheng Xia
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China
| | - Jinguan Lin
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China
| | - Linda Oyang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China
| | - Yanyan Tang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China
| | - Jiaxin Liang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China
| | - Xia Luo
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China
| | - Qianjin Liao
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China
| | - Hui Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China
| | - Yujuan Zhou
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University and Hunan Cancer Hospital, Key Laboratory of Translational Radiation Oncology, Hunan Province, 283 Tongzipo Road, Changsha 410013, Hunan, China
| |
Collapse
|
31
|
Zhou D, Zhang M, Min L, Jiang K, Jiang Y. Cerebral ischemia-reperfusion is modulated by macrophage-stimulating 1 through the MAPK-ERK signaling pathway. J Cell Physiol 2020; 235:7067-7080. [PMID: 32017081 DOI: 10.1002/jcp.29603] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/22/2020] [Indexed: 01/04/2023]
Abstract
Cerebral ischemia-reperfusion (IR) injury is associated with mitochondrial damage. Macrophage-stimulating 1 (MST1) reportedly stimulates mitochondrial apoptosis by suppressing BCL-2. We investigated whether MST1 promotes the progression of cerebral IR injury by inducing mitochondrial dysfunction in vivo and in vitro. Western blot analysis, quantitative polymerase chain reaction, immunofluorescence, and mitochondrial function assays were conducted in cells from wild-type and Mst1-knockout mice subjected to cerebral IR injury. MST1 expression in wild-type glial cells increased following cerebral IR injury. Cerebral IR injury reduced the mitochondrial membrane potential and mitochondrial metabolism in glial cells, while it enhanced mitochondrial reactive oxygen species generation and mitochondrial calcium levels in these cells. The deletion of Mst1 attenuated cerebral IR injury by improving mitochondrial function and reducing mitochondrial damage. The mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway was suppressed in wild-type glial cell upon cerebral IR injury but was reactivated in Mst1-knockout glial cell. Accordingly, blocking the MAPK/ERK pathway abolished the beneficial effects of Mst1 deletion during cerebral IR injury by inducing mitochondrial damage in glial cells. Our results suggest that cerebral IR injury is associated with MST1 upregulation in the brain, while the genetic ablation of Mst1 can attenuate mitochondrial damage and sustain brain function following cerebral IR injury.
Collapse
Affiliation(s)
- Dingzhou Zhou
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mingming Zhang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liu Min
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kaiyuan Jiang
- Department of Neurosurgery, The Central Hospital of Shaoyang, Shaoyang, Hunan, China
| | - Yugang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
32
|
Elvas F, Vanden Berghe T, Adriaenssens Y, Vandenabeele P, Augustyns K, Staelens S, Stroobants S, Van der Veken P, Wyffels L. Caspase-3 probes for PET imaging of apoptotic tumor response to anticancer therapy. Org Biomol Chem 2020; 17:4801-4824. [PMID: 31033991 DOI: 10.1039/c9ob00657e] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Apoptosis is a highly regulated process involved in the normal organism development and homeostasis. In the context of anticancer therapy, apoptosis is also studied intensively in an attempt to induce cell death in cancer cells. Caspase activation is a known key event in the apoptotic process. In particular, active caspase-3 and -7 are the common effectors in several apoptotic pathways, therefore effector caspase activation may be a promising biomarker for response evaluation to anticancer therapy. Quantitative imaging of apoptosis in vivo could provide early assessment of therapeutic effectiveness and could also be used in drug development to evaluate the efficacy as well as potential toxicity of novel treatments. Positron Emission Tomography (PET) is a highly sensitive molecular imaging modality that allows non-invasive in vivo imaging of biological processes such as apoptosis by using radiolabeled probes. Here we describe the development and evaluation of fluorine-18-labeled caspase-3 activity-based probes (ABPs) for PET imaging of apoptosis. ABPs were selected by screening of a small library of fluorine-19-labeled DEVD peptides containing different electrophilic warhead groups. An acyloxymethyl ketone was identified with low nanomolar affinity for caspase-3 and was radiolabeled with fluorine-18. The resulting radiotracer, [18F]MICA-302, showed good labeling of active caspase-3 in vitro and favorable pharmacokinetic properties. A μPET imaging experiment in colorectal tumor xenografts demonstrated an increased tumor accumulation of [18F]MICA-302 in drug-treated versus control animals. Therefore, our data suggest this radiotracer may be useful for clinical PET imaging of response to anticancer therapy.
Collapse
Affiliation(s)
- Filipe Elvas
- Molecular Imaging Center Antwerp, University of Antwerp, 2610 Wilrijk, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
DNA-PKcs promotes cardiac ischemia reperfusion injury through mitigating BI-1-governed mitochondrial homeostasis. Basic Res Cardiol 2020; 115:11. [PMID: 31919590 DOI: 10.1007/s00395-019-0773-7] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 12/27/2019] [Indexed: 01/24/2023]
Abstract
DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a novel inducer to promote mitochondrial apoptosis and suppress tumor growth in a variety of cells although its role in cardiovascular diseases remains obscure. This study was designed to examine the role of DNA-PKcs in cardiac ischemia reperfusion (IR) injury and mitochondrial damage. Cardiomyocyte-specific DNA-PKcs knockout (DNA-PKcsCKO) mice were subjected to IR prior to assessment of myocardial function and mitochondrial apoptosis. Our data revealed that IR challenge, hypoxia-reoxygenation (HR) or H2O2-activated DNA-PKcs through post-transcriptional phosphorylation in murine hearts or cardiomyocytes. Mice deficient in DNA-PKcs in cardiomyocytes were protected against cardiomyocyte death, infarct area expansion and cardiac dysfunction. DNA-PKcs ablation countered IR- or HR-induced oxidative stress, mPTP opening, mitochondrial fission, mitophagy failure and Bax-mediated mitochondrial apoptosis, possibly through suppression of Bax inhibitor-1 (BI-1) activity. A direct association between DNA-PKcs and BI-1 was noted where DNA-PKcs had little effect on BI-1 transcription but interacted with BI-1 to promote its degradation. Loss of DNA-PKcs stabilized BI-1, thus offering resistance of mitochondria and cardiomyocytes against IR insult. Moreover, DNA-PKcs ablation-induced beneficial cardioprotection against IR injury was mitigated by concurrent knockout of BI-1. Double deletion of DNA-PKcs and BI-1 failed to exert protection against global IR injury and mitochondrial damage, confirming a permissive role of BI-1 in DNA-PKcs deletion-elicited cardioprotection against IR injury. DNA-PKcs serves as a novel causative factor for mitochondrial damage via suppression of BI-1, en route to the onset and development of cardiac IR injury.
Collapse
|
34
|
Januszyk K, Januszyk P, Grabarek BO, Boroń D, Oplawski M. The Influence of Salinomycin on the Expression Profile of mRNAs Encoding Selected Caspases and MiRNAs Regulating their Expression in Endometrial Cancer Cell Line. Curr Pharm Biotechnol 2020; 21:1505-1515. [PMID: 32407273 PMCID: PMC8206191 DOI: 10.2174/1389201021666200514095043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/19/2020] [Accepted: 04/17/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Apoptosis could take place in the pathway dependent on death receptors or pathways dependent on mitochondria. In both, a key role is played by enzymes with protease activity, known as caspases. AIM The aim of this study was to assess the variances in the expression pattern of caspase-dependent signaling pathways in the endometrial cancer cell line when treated with salinomycin. Additionally, the changes in the level of miRNA that potentially regulate these mRNAs were evaluated. MATERIALS AND METHODS Endometrial cancer cells were treated with 1 μM of salinomycin for 12, 24 and 48 hours. Untreated cells made up the control culture. The molecular analysis consisted of screening mRNA and miRNA microarray expression profiles of caspases, and the evaluation of the expression of caspases 3,8 and 9 by RTqPCR, also on the protein level. RESULTS AND DISCUSSION It was observed that 5 of the 14 differentiating mRNAs were commonly found for all incubation times of the cells and they corresponded with CASP3, CASP8, and CASP9 genes. The highest impact probability was determined between CASP3(up-regulated) and hsa- miR- 30d (FC -2.01), CASP8 (down-regulated) and hsa-miR-21 (FC +1.39) and between CASP9 (upregulated) and hsa-miR-1271 (FC +1.71). CONCLUSION Salinomycin induces the apoptosis of endometrial cancer cells. The largest increase in activity was noted for caspases 3 and 9, while the expression of caspase 8 was decreased. Salinomycin causes a regulatory effect on the transcriptomes of mRNA and miRNA in in vitro endometrial cancer cells.
Collapse
Affiliation(s)
- Krzysztof Januszyk
- Address correspondence to this author at the Faculty of Health Science, Public Higher Medical Professional School in Opole, Poland;, E-mail:
| | | | | | | | | |
Collapse
|
35
|
Maluch I, Czarna J, Drag M. Applications of Unnatural Amino Acids in Protease Probes. Chem Asian J 2019; 14:4103-4113. [PMID: 31593336 DOI: 10.1002/asia.201901152] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 10/01/2019] [Indexed: 12/11/2022]
Abstract
Since proteases are involved in a wide range of physiological and disease states, the development of novel tools for imaging proteolytic enzyme activity is attracting increasing interest from scientists. Peptide substrates containing proteinogenic amino acids are often the first line of defining enzyme specificity. This Minireview outlines examples of major recent advances in probing proteases using unnatural amino acid residues, which greatly expands the possibilities for designing substrate probes and inhibitory activity-based probes. This approach already yielded innovative probes that selectively target only one active protease within the group of enzymes exhibiting similar specificity both in cellular assays and in bioimaging research.
Collapse
Affiliation(s)
- Izabela Maluch
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw, University of Science and Technology, Wyb. Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Justyna Czarna
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw, University of Science and Technology, Wyb. Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Marcin Drag
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw, University of Science and Technology, Wyb. Wyspianskiego 27, 50-370, Wroclaw, Poland
| |
Collapse
|
36
|
Fan L, Wang J, Ma C. miR125a attenuates BMSCs apoptosis via the MAPK‐ERK pathways in the setting of craniofacial defect reconstruction. J Cell Physiol 2019; 235:2857-2865. [PMID: 31578723 DOI: 10.1002/jcp.29191] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 09/03/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Longkun Fan
- Department of Medical Plastic Surgery, Cangzhou Central Hospital, Hebei, China
| | - Jingxian Wang
- Department of Medical Plastic Surgery, Cangzhou Central Hospital, Hebei, China
| | - Chao Ma
- Department of Medical Plastic Surgery, Cangzhou Central Hospital, Hebei, China
| |
Collapse
|
37
|
Poreba M, Groborz K, Vizovisek M, Maruggi M, Turk D, Turk B, Powis G, Drag M, Salvesen GS. Fluorescent probes towards selective cathepsin B detection and visualization in cancer cells and patient samples. Chem Sci 2019; 10:8461-8477. [PMID: 31803426 PMCID: PMC6839509 DOI: 10.1039/c9sc00997c] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/29/2019] [Indexed: 12/23/2022] Open
Abstract
Highly selective fluorescent activity-based probe for the visualization of cathepsin B in cancer cells.
Human cysteine cathepsins constitute an 11-membered family of proteases responsible for degradation of proteins in cellular endosomal–lysosomal compartments as such, they play important roles in antigen processing, cellular stress signaling, autophagy, and senescence. Moreover, for many years these enzymes were also linked to tumor growth, invasion, angiogenesis and metastasis when upregulated. Individual biological roles of each cathepsin are difficult to establish, because of their redundancy and similar substrate specificities. Selective chemical tools that enable imaging of individual cathepsin activities in living cells, tumors, and the tumor microenvironment may provide a better insight into their functions. In this work, we used HyCoSuL technology to profile the substrate specificity of human cathepsin B. The use of unnatural amino acids in the substrate library enabled us to uncover the broad cathepsin B preferences that we utilized to design highly-selective substrates and fluorescent activity-based probes (ABPs). We further demonstrated that Cy5-labeled MP-CB-2 probe can selectively label cathepsin B in eighteen cancer cell lines tested, making this ABP highly suitable for other biological setups. Moreover, using Cy5-labelled MP-CB-2 we were able to demonstrate by fluorescence microscopy that in cancer cells cathepsins B and L share overlapping, but not identical subcellular localization.
Collapse
Affiliation(s)
- Marcin Poreba
- Sanford Burnham Prebys Medical Discovery Institute , 10901 North Torrey Pines Road , La Jolla , CA 92037 , USA . ; ; .,Department of Bioorganic Chemistry , Faculty of Chemistry , Wroclaw University of Technology , Wyb. Wyspianskiego 27 , 50-370 Wroclaw , Poland
| | - Katarzyna Groborz
- Department of Bioorganic Chemistry , Faculty of Chemistry , Wroclaw University of Technology , Wyb. Wyspianskiego 27 , 50-370 Wroclaw , Poland
| | - Matej Vizovisek
- Department of Biochemistry and Molecular and Structural Biology , Jožef Stefan Institute , SI-1000 Ljubljana , Slovenia
| | - Marco Maruggi
- Sanford Burnham Prebys Medical Discovery Institute , 10901 North Torrey Pines Road , La Jolla , CA 92037 , USA . ; ;
| | - Dusan Turk
- Department of Biochemistry and Molecular and Structural Biology , Jožef Stefan Institute , SI-1000 Ljubljana , Slovenia
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology , Jožef Stefan Institute , SI-1000 Ljubljana , Slovenia.,Faculty of Chemistry and Chemical Technology , University of Ljubljana , SI-1000 Ljubljana , Slovenia
| | - Garth Powis
- Sanford Burnham Prebys Medical Discovery Institute , 10901 North Torrey Pines Road , La Jolla , CA 92037 , USA . ; ;
| | - Marcin Drag
- Sanford Burnham Prebys Medical Discovery Institute , 10901 North Torrey Pines Road , La Jolla , CA 92037 , USA . ; ; .,Department of Bioorganic Chemistry , Faculty of Chemistry , Wroclaw University of Technology , Wyb. Wyspianskiego 27 , 50-370 Wroclaw , Poland
| | - Guy S Salvesen
- Sanford Burnham Prebys Medical Discovery Institute , 10901 North Torrey Pines Road , La Jolla , CA 92037 , USA . ; ;
| |
Collapse
|
38
|
de Vries LE, Sanchez MI, Groborz K, Kuppens L, Poreba M, Lehmann C, Nevins N, Withers-Martinez C, Hirst DJ, Yuan F, Arastu-Kapur S, Horn M, Mares M, Bogyo M, Drag M, Deu E. Characterization of P. falciparum dipeptidyl aminopeptidase 3 specificity identifies differences in amino acid preferences between peptide-based substrates and covalent inhibitors. FEBS J 2019; 286:3998-4023. [PMID: 31177613 PMCID: PMC6851853 DOI: 10.1111/febs.14953] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 04/12/2019] [Accepted: 06/06/2019] [Indexed: 12/31/2022]
Abstract
Malarial dipeptidyl aminopeptidases (DPAPs) are cysteine proteases important for parasite development thus making them attractive drug targets. In order to develop inhibitors specific to the parasite enzymes, it is necessary to map the determinants of substrate specificity of the parasite enzymes and its mammalian homologue cathepsin C (CatC). Here, we screened peptide-based libraries of substrates and covalent inhibitors to characterize the differences in specificity between parasite DPAPs and CatC, and used this information to develop highly selective DPAP1 and DPAP3 inhibitors. Interestingly, while the primary amino acid specificity of a protease is often used to develop potent inhibitors, we show that equally potent and highly specific inhibitors can be developed based on the sequences of nonoptimal peptide substrates. Finally, our homology modelling and docking studies provide potential structural explanations of the differences in specificity between DPAP1, DPAP3, and CatC, and between substrates and inhibitors in the case of DPAP3. Overall, this study illustrates that focusing the development of protease inhibitors solely on substrate specificity might overlook important structural features that can be exploited to develop highly potent and selective compounds.
Collapse
Affiliation(s)
- Laura E de Vries
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mateo I Sanchez
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Katarzyna Groborz
- Division of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw, Poland
| | - Laurie Kuppens
- Chemical Biology Approaches to Malaria Laboratory, The Francis Crick Institute, London, UK
| | - Marcin Poreba
- Division of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw, Poland
| | - Christine Lehmann
- Chemical Biology Approaches to Malaria Laboratory, The Francis Crick Institute, London, UK
| | - Neysa Nevins
- Computational Sciences, GlaxoSmithKline, Collegeville, PA, USA
| | | | - David J Hirst
- Crick-GSK Biomedical LinkLabs, GlaxoSmithKline, Stevenage, UK
| | - Fang Yuan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Shirin Arastu-Kapur
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Martin Horn
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Michael Mares
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Marcin Drag
- Division of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw, Poland
| | - Edgar Deu
- Chemical Biology Approaches to Malaria Laboratory, The Francis Crick Institute, London, UK
| |
Collapse
|
39
|
Potent and selective caspase-2 inhibitor prevents MDM-2 cleavage in reversine-treated colon cancer cells. Cell Death Differ 2019; 26:2695-2709. [PMID: 30976094 DOI: 10.1038/s41418-019-0329-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/17/2019] [Accepted: 03/25/2019] [Indexed: 01/02/2023] Open
Abstract
Most caspases can be positioned unambiguously within the regulated cell death networks of apoptosis and pyroptosis, but the role of caspase-2, a highly conserved protease within the family, remains enigmatic. This is mainly due to lack of selective chemical and biochemical tools for the investigation of this protease. In this study, we used our hybrid combinatorial substrate library (HyCoSuL) approach to broadly profile caspase-2 substrate specificity using peptide scanning libraries. This screen uncovered previously unknown caspase-2 peptidyl substrate preferences, which were further used to develop caspase-2 selective fluorogenic substrates and covalent, irreversible AOMK inhibitors. Finally, we used the champion inhibitor (NH-23-C2) in reversine-treated HCT-116 colon cancer cells to selectively block caspase-2 activity and caspase-2-mediated MDM-2 cleavage. In addition, we showed that NH-23-C2 does not block caspase-3 or caspase-8, which makes it a powerful chemical tool to dissect the true role of caspase-2 in various biological setups.
Collapse
|
40
|
Zou H, Sevigny MB, Liu S, Madden DT, Louie MC. Novel flexible heteroarotinoid, SL-1-39, inhibits HER2-positive breast cancer cell proliferation by promoting lysosomal degradation of HER2. Cancer Lett 2019; 443:157-166. [PMID: 30503556 DOI: 10.1016/j.canlet.2018.11.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/21/2018] [Accepted: 11/24/2018] [Indexed: 10/27/2022]
Abstract
SL-1-39 [1-(4-chloro-3-methylphenyl)-3-(4-nitrophenyl)thiourea] is a new flexible heteroarotinoid (Flex-Het) analog derived from the parental compound, SHetA2, previously shown to inhibit cell growth across multiple cancer types. The current study aims to determine growth inhibitory effects of SL-1-39 across the different subtypes of breast cancer cells and delineate its molecular mechanism. Our results demonstrate that while SL-1-39 blocks cell proliferation of all breast cancer subtypes tested, it has the highest efficacy against HER2+ breast cancer cells. Molecular analyses suggest that SL-1-39 prevents S phase progression of HER2+ breast cancer cells (SKBR3 and MDA-MB-453), which is consistent with reduced expression of key cell-cycle regulators at both the protein and transcriptional levels. SL-1-39 treatment also decreases the protein levels of HER2 and pHER2 as well as its downstream effectors, pMAPK and pAKT. Reduction of HER2 and pHER2 at the protein level is attributed to increased lysosomal degradation of total HER2 levels. This is the first study to show that a flexible heteroarotinoid analog modulates the HER2 signaling pathway through lysosomal degradation, and thus further warrants the development of SL-1-39 as a therapeutic option for HER2+ breast cancer.
Collapse
Affiliation(s)
- Hongye Zou
- Department of Natural Sciences and Mathematics, Dominican University of California, 50 Acacia Avenue, San Rafael, CA, 94901, USA.
| | - Mary B Sevigny
- Department of Natural Sciences and Mathematics, Dominican University of California, 50 Acacia Avenue, San Rafael, CA, 94901, USA.
| | - Shengquan Liu
- College of Pharmacy, Touro University California, 1310 Club Drive, Vallejo, CA, 94594, USA.
| | - David T Madden
- College of Pharmacy, Touro University California, 1310 Club Drive, Vallejo, CA, 94594, USA; Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA, 94945, USA.
| | - Maggie C Louie
- Department of Natural Sciences and Mathematics, Dominican University of California, 50 Acacia Avenue, San Rafael, CA, 94901, USA; College of Pharmacy, Touro University California, 1310 Club Drive, Vallejo, CA, 94594, USA.
| |
Collapse
|
41
|
Cogo F, Poreba M, Rut W, Groborz K, Smyth P, Johnston MC, Williams R, Longley DB, Burden RE, Salvesen GS, Drag M, Scott CJ. Development of an advanced nanoformulation for the intracellular delivery of a caspase-3 selective activity-based probe. NANOSCALE 2019; 11:742-751. [PMID: 30566168 PMCID: PMC7331821 DOI: 10.1039/c8nr07859a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The ability to label active caspase-3 represents a useful pharmacodynamic strategy to determine the efficacy of anti-tumour drugs. Activity-based probes (ABPs) provide a method for the labelling of activated caspases and the recent development of hybrid combinatorial substrate libraries (HyCoSuL) has allowed for the generation of highly selective ABPs to discriminately label these proteases. Here using this approach, a novel caspase-3 selective ABP (CS1) has been developed and validated in apoptotic cells to selectively bind caspase-3 over the closely related caspase-7. However, a critical bottleneck for ABPs is their cell penetrance and therefore this cell-impermeable CS1 probe was subsequently formulated into PLGA-based nanoparticles (CS1-NPs). We demonstrate the ability of these particles to be taken up by the cells and facilitate intracellular delivery of the ABP to effectively label caspase 3 in response to apoptotic stimuli. This work forms the foundation of a novel approach for the labelling of caspase 3 and may have downstream utility to measure real time apoptosis in tumours and other organs.
Collapse
Affiliation(s)
- Francesco Cogo
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Vizovišek M, Vidmar R, Drag M, Fonović M, Salvesen GS, Turk B. Protease Specificity: Towards In Vivo Imaging Applications and Biomarker Discovery. Trends Biochem Sci 2018; 43:829-844. [PMID: 30097385 DOI: 10.1016/j.tibs.2018.07.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/05/2018] [Accepted: 07/12/2018] [Indexed: 02/06/2023]
Abstract
Proteases are considered of major importance in biomedical research because of their crucial roles in health and disease. Their ability to hydrolyze their protein and peptide substrates at single or multiple sites, depending on their specificity, makes them unique among the enzymes. Understanding protease specificity is therefore crucial to understand their biology as well as to develop tools and drugs. Recent advancements in the fields of proteomics and chemical biology have improved our understanding of protease biology through extensive specificity profiling and identification of physiological protease substrates. There are growing efforts to transfer this knowledge into clinical modalities, but their success is often limited because of overlapping protease features, protease redundancy, and chemical tools lacking specificity. Herein, we discuss the current trends and challenges in protease research and how to exploit the growing information on protease specificities for understanding protease biology, as well as for development of selective substrates, cleavable linkers, and activity-based probes and for biomarker discovery.
Collapse
Affiliation(s)
- Matej Vizovišek
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia; These authors contributed equally to this work
| | - Robert Vidmar
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia; These authors contributed equally to this work
| | - Marcin Drag
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Marko Fonović
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Guy S Salvesen
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, SI-1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
43
|
Abstract
This question of whether fungi undergo apoptosis-like programmed cell death can be separated into two questions. One question is about applying the term "apoptosis" to fungi, and the other is a more challenging question of whether fungi have evolved mechanisms that inflict self-injury. The answers to both questions depend on the definitions applied to "apoptosis" and "programmed cell death." Considering how these and other cell death terms originated and are currently defined for animals, some confusion arises when the terms are applied to fungi. While it is difficult to defend the concept of fungal apoptosis, the more interesting issue is whether fungi will eventually be found to encode programmed or extemporaneous self-destructive processes, as suggested by intriguing new findings.
Collapse
Affiliation(s)
- J Marie Hardwick
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|