1
|
Zhang S, Zhang T, Cao Z, Yang Y, Lü P. Hijacking the autophagy-apoptosis crosstalk: African swine fever virus orchestrates immune evasion via host remodeling for viral pathogenesis. Microb Pathog 2025; 204:107609. [PMID: 40250498 DOI: 10.1016/j.micpath.2025.107609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 04/13/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
African swine fever (ASF) is an acute, highly fatal hemorrhagic disease of domestic and wild pigs caused by African swine fever virus (ASFV). ASFV, a large double-stranded DNA virus of the Asfarviridae family, is highly infectious and pathogenic. Through modulation of host apoptosis and autophagy pathways, the virus subverts innate immune surveillance to promote its replication and dissemination. Following ASFV infection, domestic pigs may exhibit 100 % morbidity and mortality rates with highly virulent strains, constituting a major threat to the global pork industry. Nowadays, ASF is listed as a notifiable terrestrial animal disease by the World Organisation for Animal Health (WOAH). Therefore, elucidating ASFV's pathogenic mechanisms, particularly its molecular regulation of apoptosis and autophagy, is crucial for developing effective ASF control and prevention strategies. This review comprehensively summarizes the pathogenic mechanisms of ASFV, with particular focus on the autophagy-apoptosis crosstalk and viral manipulation of these cellular processes. These insights not only improve our understanding of ASFV-mediated immune evasion mechanisms but also provide valuable references for developing ASF control strategies targeting apoptosis and autophagy pathways.
Collapse
Affiliation(s)
- Simeng Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, PR China
| | - Tiancheng Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, PR China
| | - Zhaoxiao Cao
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, PR China
| | - Yanhua Yang
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, PR China.
| | - Peng Lü
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, PR China
| |
Collapse
|
2
|
Oliveira MS, Fernandes RA, Pinto LS, Moreira FA, Castro OWD, Santos VR. Balancing efficacy and safety: The dual impact of antiseizure medications on the developing brain. Epilepsy Behav 2025; 167:110400. [PMID: 40187052 DOI: 10.1016/j.yebeh.2025.110400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/25/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
The number of neurons in the developing brain is greater than typically found in adulthood, and the brain possesses delicate mechanisms to induce the death of excess cells and refine neural circuitry. The correct tuning between the processes of neuronal death and survival generates a mature and functional brain in its complexity and plastic capacity. Epilepsy is a highly prevalent neurological condition worldwide, including among young individuals. However, exposure to the main treatment approaches, the long-term use of Antiseizure Medication (ASM), during the critical period of development can induce a series of changes in this delicate balance. Acting by various mechanisms of action, ASMs may induce an increase in neuronal death, something that translates into deleterious neuropsychiatric effects in adulthood. Several investigations conducted in recent years have brought to light new aspects related to this dynamic, yet many questions, such as the cellular mechanisms of death and the pathophysiology of late effects, still have unresolved elements. In this review, we aimed to explore the mechanisms of action of the most widely used ASMs in the treatment of neonatal epilepsy, the broad aspects of neuronal death in the developing brain and the repercussions of this death and other effects in adulthood. We review the evidence indicating a relationship between exposure to ASMs and the manifestation of associated psychiatric comorbidities in adulthood and discuss some possible mechanisms underlying the induction of this process by morphological and physiological changes in the related behaviors.
Collapse
Affiliation(s)
- M S Oliveira
- Department of Morphology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - R A Fernandes
- Department of Morphology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - L S Pinto
- Department of Morphology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - F A Moreira
- Department of Pharmacology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| | - O W de Castro
- Departament of Physiology, Institute of Biological Science and Health, Universidade Federal de Alagoas - UFAL, Brazil
| | - V R Santos
- Department of Morphology, Institute of Biological Science, Universidade Federal de Minas Gerais (ICB/UFMG), Belo Horizonte, Brazil
| |
Collapse
|
3
|
Willard PA, Kornbluth J. The ubiquitin ligase NKLAM promotes apoptosis and suppression of cell growth. J Biol Chem 2025; 301:108527. [PMID: 40273985 DOI: 10.1016/j.jbc.2025.108527] [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: 01/30/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Natural killer lytic-associated molecule (NKLAM), also known as RNF19b, is a member of the RING-in between-RING-RING (RBR) E3 ubiquitin ligase family and plays a pivotal role in immune regulation. We identified a critical cysteine residue at position 301 essential for NKLAM's ubiquitin ligase function. Site-directed mutagenesis of this residue to serine or alanine abrogated the ligase activity of NKLAM. Utilizing inducible expression systems in two different cell lines, HEK293 embryonic kidney cells and K562 myeloid leukemia cells, we demonstrated that wild-type (WT) NKLAM, but not the catalytically inactive NKLAM alanine mutant (C301A), inhibited cellular proliferation, as evidenced by reduced cell numbers and decreased metabolic activity. Moreover, NKLAM expression led to a significant decrease in the abundance and stability of the proto-oncogene c-Myc, a key regulator of proliferation. NKLAM facilitated the proteasomal degradation of c-Myc, with a reduction in c-Myc half-life from 27 min to 12 min and restoration of c-Myc levels upon proteasome inhibition. Notably, prolonged NKLAM expression induced apoptosis, measured by annexin-V staining and caspase activation. Strikingly, the serine mutant, C301S, while lacking ubiquitin ligase activity, induced apoptosis comparable to WT NKLAM, highlighting an alternative pathway for NKLAM-mediated inhibition of cellular homeostasis. Our findings indicate that NKLAM is a cytolytic protein with multifaceted roles in cellular proliferation and apoptosis.
Collapse
Affiliation(s)
- Paul A Willard
- Department of Pathology, Saint Louis University School of Medicine, St Louis, Missouri, USA
| | - Jacki Kornbluth
- Department of Pathology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Research and Development Service, St Louis VA Medical Center, St Louis, Missouri, USA.
| |
Collapse
|
4
|
Prata RBDS, Pinheiro RO. Cell Death Mechanisms in Mycobacterium abscessus Infection: A Double-Edged Sword. Pathogens 2025; 14:391. [PMID: 40333197 PMCID: PMC12030298 DOI: 10.3390/pathogens14040391] [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: 03/07/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025] Open
Abstract
Infections caused by non-tuberculous mycobacteria (NTM), such as Mycobacterium abscessus, elicit diverse cell death mechanisms including apoptosis, necrosis, and pyroptosis, which play key roles in immunopathogenesis. NTM can manipulate these cell death pathways to evade host immune responses, ensuring their intracellular survival and persistence. Apoptosis may aid in antigen presentation and immune activation, while necrosis and pyroptosis trigger excessive inflammation, leading to tissue damage. Autophagy, a crucial cellular defense mechanism, is often induced in response to NTM infection; however, M. abscessus has evolved mechanisms to inhibit autophagic processes, enhancing its ability to survive within host cells. This manipulation of cell death pathways, particularly the dysregulation of autophagy and ferroptosis, contributes to chronic infection, immune evasion, and tissue damage, complicating disease management. Understanding these mechanisms offers potential therapeutic targets for improving treatment strategies against M. abscessus infections.
Collapse
Affiliation(s)
| | - Roberta Olmo Pinheiro
- Leprosy Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro 21040-360, Brazil;
| |
Collapse
|
5
|
Saadh MJ, Muhammad FA, Alazzawi TS, Fahdil AA, Athab ZH, Tuxtayev J, Alsaikhan F, Farhood B. Regulation of Apoptotic Pathways by MicroRNAs: A Therapeutic Strategy for Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04833-5. [PMID: 40220245 DOI: 10.1007/s12035-025-04833-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/09/2025] [Indexed: 04/14/2025]
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder marked by a gradual decline in memory and cognitive functions. It is characterized by the presence of senile plaques, neurofibrillary tangles, and neuronal degeneration, affecting a significant portion of the human population. A key feature of various nervous system disorders, including AD, is extensive cellular death caused by apoptosis, which affects not only neurons but also glial cells. While apoptosis plays a vital role in eliminating certain cells and supporting normal development, alterations or disruptions in apoptotic pathways can lead to harmful neurodegenerative conditions such as AD. Thus, targeting apoptosis presents a promising therapeutic approach for these diseases. MicroRNAs (miRNAs), a class of non-coding RNA, play diverse roles in cellular functions, including proliferation, gene expression regulation, programmed cell death, intercellular communication, and angiogenesis. By modulating regulatory genes, miRNAs can influence apoptosis, either promoting or inhibiting it. Aberrant expression of miRNAs can impact multiple apoptotic pathways, potentially driving the progression of AD and related health issues. This review summarizes recent research on miRNAs and their dual role in exacerbating or protecting against neural cell damage in AD by altering apoptotic pathways. The regulation of apoptosis by miRNAs offers a prospective therapeutic strategy for Alzheimer's disease.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | - Tuqa S Alazzawi
- College of Dentist, National University of Science and Technology, Dhi Qar, Iraq
| | - Ali A Fahdil
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Jamshid Tuxtayev
- Department of Surgical Diseases, Faculty of Pediatrics, Samarkand State Medical Institute, Samarkand, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
6
|
Singh R, Gaur SK, Nagar R, Kaul R. Insights into the different mechanisms of Autophagy and Apoptosis mediated by Morbilliviruses. Virology 2025; 603:110371. [PMID: 39742556 DOI: 10.1016/j.virol.2024.110371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/10/2024] [Accepted: 12/20/2024] [Indexed: 01/03/2025]
Abstract
Viruses are obligate intracellular parasites that have co-evolved with the host. During the course of evolution, viruses have acquired abilities to abrogate the host's immune responses by modulating the host proteins which play a pivotal role in various biological processes. One such process is the programmed cell death in virus-infected cells, which can occur via autophagy or apoptosis. Morbilliviruses are known to modulate both autophagy and apoptosis. Upon infecting a cell, the morbilliviruses can utilize autophagosomes as their nest and delay the host defense apoptotic response, and/or can promote apoptosis to escalate the virus dissemination. Moreover, there is an active interplay between these two pathways which eventually decides the fate of a virus-infected cell. Recent advances in our understanding of these processes provide a potential rationale to further explore morbilliviruses for therapeutic purposes.
Collapse
Affiliation(s)
- Rashmi Singh
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Sharad Kumar Gaur
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Rakhi Nagar
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India
| | - Rajeev Kaul
- Department of Microbiology, University of Delhi South Campus, New Delhi, 110021, India.
| |
Collapse
|
7
|
Yang S, Zou Y, Zhong C, Zhou Z, Peng X, Tang C. Dual role of pyroptosis in liver diseases: mechanisms, implications, and therapeutic perspectives. Front Cell Dev Biol 2025; 13:1522206. [PMID: 39917567 PMCID: PMC11798966 DOI: 10.3389/fcell.2025.1522206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/10/2025] [Indexed: 02/09/2025] Open
Abstract
Pyroptosis, a form of programmed cell death induced by inflammasome with a mechanism distinct from that of apoptosis, occurs via one of the three pathway types: classical, non-classical, and granzyme A/B-dependent pyroptosis pathways. Pyroptosis is implicated in various diseases, notably exhibiting a dual role in liver diseases. It facilitates the clearance of damaged hepatocytes, preventing secondary injury, and triggers immune responses to eliminate pathogens and damaged cells. Conversely, excessive pyroptosis intensifies inflammatory responses, exacerbates hepatocyte damage and promotes the activation and proliferation of hepatic stellate cells, accelerating liver fibrosis. Furthermore, by sustaining an inflammatory state, impacts the survival and proliferation of cancer cells. This review comprehensively summarizes the dual role of pyroptosis in liver diseases and its therapeutic strategies, offering new theoretical foundations and practical guidance for preventing and treating of liver diseases.
Collapse
Affiliation(s)
| | | | | | - Zuoqiong Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Xiyang Peng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| | - Changfa Tang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, China
| |
Collapse
|
8
|
Wei Y, Ma HK, Wong ME, Papasavvas E, Konnikova L, Tebas P, Morgenstern R, Montaner LJ, Ho YC. BACH2-driven tissue resident memory programs promote HIV-1 persistence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.16.628794. [PMID: 39763845 PMCID: PMC11702684 DOI: 10.1101/2024.12.16.628794] [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: 01/12/2025]
Abstract
Transcription repressor BACH2 redirects short-lived terminally differentiated effector into long-lived memory cells. We postulate that BACH2-mediated long-lived memory programs promote HIV-1 persistence in gut CD4+ T cells. We coupled single-cell DOGMA-seq and TREK-seq to capture chromatin accessibility, transcriptome, surface proteins, T cell receptor, HIV-1 DNA and HIV-1 RNA in 100,744 gut T cells from ten aviremic HIV-1+ individuals and five HIV-1- donors. BACH2 was the leading transcription factor that shaped gut tissue resident memory T cells (TRMs) into long-lived memory with restrained interferon-induced effector function. We found that HIV-1-infected cells were enriched in TRMs (80.8%). HIV-1-infected cells had increased BACH2 transcription factor accessibility, TRM (CD49a, CD69, CD103) and survival (IL7R) gene expression, and Th17 polarization (RORC, CCR6). In vitro gut CD4+ T cell infection revealed preferential infection and persistence of HIV-1 in CCR6+ TRMs. Overall, we found BACH2-driven TRM program promotes HIV-1 persistence and BACH2 as a new therapeutic target.
Collapse
Affiliation(s)
- Yulong Wei
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Haocong Katherine Ma
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Michelle E. Wong
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06519, USA
| | | | - Liza Konnikova
- Departments of Pediatrics, Yale University School of Medicine, New Haven, CT 06519, USA
| | - Pablo Tebas
- Presbyterian Hospital-University of Pennsylvania Hospital, Philadelphia, PA 19104, USA
| | - Ricardo Morgenstern
- Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | | - Ya-Chi Ho
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06519, USA
| |
Collapse
|
9
|
Tu W, Guo M, Zhang Z, Li C. Pathogen-induced apoptosis in echinoderms: A review. FISH & SHELLFISH IMMUNOLOGY 2024; 155:109990. [PMID: 39481501 DOI: 10.1016/j.fsi.2024.109990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/16/2024] [Accepted: 10/26/2024] [Indexed: 11/02/2024]
Abstract
Echinoderms possess unique biological traits that make them valuable models in immunology, regeneration, and developmental biology studies. As a class rich in active substances with significant nutritional and medicinal value, echinoderms face threats from marine pathogens, including bacteria, viruses, fungi, protozoa, and parasites, which have caused substantial economic losses in echinoderm aquaculture. Echinoderms counteract pathogen invasion through innate immunity and programmed cell death, in particular, with apoptosis being essential for eliminating infected or damaged cells and maintaining homeostasis in many echinoderm cell types. Despite the importance of this process, there is a lack of comprehensive and updated reviews on this topic. This review underscores that echinoderm apoptotic pathways exhibit a complexity comparable to that of vertebrates, featuring proteins with unique domains that may indicate the presence of novel signaling mechanisms. We synthesize current knowledge on how echinoderms utilize diverse transcriptional and post-transcriptional mechanisms to regulate apoptosis in response to pathogen infections and explore how pathogens have evolved strategies to manipulate echinoderm apoptosis, either by inhibiting it to create survival niches or by inducing excessive apoptosis to weaken the host. By elucidating the primary apoptotic pathways in echinoderms and the host-pathogen interactions that modulate these pathways, this review aims to reveal new mechanisms of apoptosis in animal immune defense and provide insights into the evolutionary arms race between hosts and pathogens.
Collapse
Affiliation(s)
- Weitao Tu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Ming Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China.
| | - Zhen Zhang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, China.
| |
Collapse
|
10
|
Santana PT, de Lima IS, da Silva e Souza KC, Barbosa PHS, de Souza HSP. Persistent Activation of the P2X7 Receptor Underlies Chronic Inflammation and Carcinogenic Changes in the Intestine. Int J Mol Sci 2024; 25:10874. [PMID: 39456655 PMCID: PMC11507540 DOI: 10.3390/ijms252010874] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Aberrant signaling through damage-associated molecular patterns (DAMPs) has been linked to several health disorders, attracting considerable research interest over the last decade. Adenosine triphosphate (ATP), a key extracellular DAMP, activates the purinergic receptor P2X7, which acts as a danger sensor in immune cells and is implicated in distinct biological functions, including cell death, production of pro-inflammatory cytokines, and defense against microorganisms. In addition to driving inflammation mediated by immune and non-immune cells, the persistent release of endogenous DAMPs, including ATP, has been shown to result in epigenetic modifications. In intestinal diseases such as inflammatory bowel disease (IBD) and colorectal cancer (CRC), consequent amplification of the inflammatory response and the resulting epigenetic reprogramming may impact the development of pathological changes associated with specific disease phenotypes. P2X7 is overexpressed in the gut mucosa of patients with IBD, whereas the P2X7 blockade prevents the development of chemically induced experimental colitis. Recent data suggest a role for P2X7 in determining gut microbiota composition. Regulatory mechanisms downstream of the P2X7 receptor, combined with signals from dysbiotic microbiota, trigger intracellular signaling pathways and inflammasomes, intensify inflammation, and foster colitis-associated CRC development. Preliminary studies targeting the ATP-P2X7 pathway have shown favorable therapeutic effects in human IBD and experimental colitis.
Collapse
Affiliation(s)
- Patricia Teixeira Santana
- Department of Clinical Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (P.T.S.); (I.S.d.L.); (K.C.d.S.e.S.); (P.H.S.B.)
- D’Or Institute for Research and Education (IDOR), Rua Diniz Cordeiro 30, Botafogo, Rio de Janeiro 22281-100, Brazil
| | - Isadora Schmukler de Lima
- Department of Clinical Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (P.T.S.); (I.S.d.L.); (K.C.d.S.e.S.); (P.H.S.B.)
| | - Karen Cristina da Silva e Souza
- Department of Clinical Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (P.T.S.); (I.S.d.L.); (K.C.d.S.e.S.); (P.H.S.B.)
| | - Pedro Henrique Sales Barbosa
- Department of Clinical Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (P.T.S.); (I.S.d.L.); (K.C.d.S.e.S.); (P.H.S.B.)
| | - Heitor Siffert Pereira de Souza
- Department of Clinical Medicine, Federal University of Rio de Janeiro, Rio de Janeiro 21941-913, Brazil; (P.T.S.); (I.S.d.L.); (K.C.d.S.e.S.); (P.H.S.B.)
- D’Or Institute for Research and Education (IDOR), Rua Diniz Cordeiro 30, Botafogo, Rio de Janeiro 22281-100, Brazil
| |
Collapse
|
11
|
Wu S, Ding D, Wang D. Regulated Cell Death Pathways in Pathological Cardiac Hypertrophy. Rev Cardiovasc Med 2024; 25:366. [PMID: 39484135 PMCID: PMC11522757 DOI: 10.31083/j.rcm2510366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/19/2024] [Accepted: 06/03/2024] [Indexed: 11/03/2024] Open
Abstract
Cardiac hypertrophy is characterized by an increased volume of individual cardiomyocytes rather than an increase in their number. Myocardial hypertrophy due to pathological stimuli encountered by the heart, which reduces pressure on the ventricular walls to maintain cardiac function, is known as pathological hypertrophy. This eventually progresses to heart failure. Certain varieties of regulated cell death (RCD) pathways, including apoptosis, pyroptosis, ferroptosis, necroptosis, and autophagy, are crucial in the development of pathological cardiac hypertrophy. This review summarizes the molecular mechanisms and signaling pathways underlying these RCD pathways, focusing on their mechanism of action findings for pathological cardiac hypertrophy. It intends to provide new ideas for developing therapeutic approaches targeted at the cellular level to prevent or reverse pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Shengnan Wu
- Department of Geriatrics, The First Affiliated Hospital of Wannan Medical College, 241001 Wuhu, Anhui, China
| | - Ding Ding
- Department of Geriatrics, The First Affiliated Hospital of Wannan Medical College, 241001 Wuhu, Anhui, China
| | - Deguo Wang
- Department of Geriatrics, The First Affiliated Hospital of Wannan Medical College, 241001 Wuhu, Anhui, China
| |
Collapse
|
12
|
Kim HY, Ha H. Distinct granzyme k expression in immune cells: a single-cell rna-seq meta-analysis. Genes Genomics 2024; 46:1097-1106. [PMID: 39115674 DOI: 10.1007/s13258-024-01555-1] [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: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Granzymes are essential serine proteases in cytotoxic T cells and natural killer (NK) cells, with GZMK's expression being less understood. This study aims to uncover GZMK expression profiles across various immune cell types using single-cell RNA sequencing meta-analysis. OBJECTIVE This study aims to uncover GZMK expression profiles across various immune cell types using single-cell RNA sequencing meta-analysis. METHODS We conducted a meta-analysis using cellxgene, an interactive data exploration platform developed by the Chan Zuckerberg Initiative. We focused on mature T cells, NK cells, B cells, and NKT cells. We also checked transcription factor binding sites at the granzyme gene promoter regions using JASPAR. Comparative analysis was also done using mouse single-cell RNA sequencing data. RESULTS GZMK was the most lowly expressed in NK cells and mature NKT cells in most tissues except for colon and lymph nodes. In mature T cells, GZMK is similarly or more highly expressed than other granzymes. HBCA data revealed weak expression of GZMK in NK cells but strong expression in effector memory CD8-positive, alpha-beta T cells. Combined data shows no significant difference in GZMK expression between cell types. Subtype analysis shows that GZMK expression was higher in CD16-negative, CD56-bright NK cells when compared to CD16-positive, CD56-dim NK cells. We also identified unique transcription factor binding sites for GZMK. While this pattern in mouse data with low Gzmk expression in NK cells and higher T cells was repeated. CONCLUSION GZMK expression is distinctively regulated among immune cells and tissues, with unique promoter regions and transcription factor binding sites contributing to this differential expression. These insights into GZMK's role in immune function and regulation offer potential therapeutic targets.
Collapse
Affiliation(s)
- Hyeon-Young Kim
- Department of Molecular and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Hongseok Ha
- Institute of Endemic Disease, Seoul National University Medical Research Center, Seoul, Republic of Korea.
| |
Collapse
|
13
|
Lenárt M, Bober P, Marcin M, Tkáčiková S, Kacírová M, Alexovič M, Tóth D, Madárová N, Radoňak J, Urdzík P, Fedačko J, Sabo J. Peripheral Blood CD8 + T-Lymphocyte Immune Response in Benign and Subpopulations of Breast Cancer Patients. Int J Mol Sci 2024; 25:6423. [PMID: 38928129 PMCID: PMC11204132 DOI: 10.3390/ijms25126423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Peripheral blood CD8+ T lymphocytes play a crucial role in cell-mediated immunity and tumor-related immune responses in breast cancer. In this study, label-free quantification analysis and gene set enrichment analysis (GSEA) of CD8+ T lymphocytes in the peripheral blood of benign patients and patients with different breast cancer (BC) subtypes, i.e., luminal A, luminal B, and triple-negative breast cancer (TNBC), were performed using nano-UHPLC and Orbitrap mass spectrometry. Differential protein expression in CD8+ T lymphocytes revealed significant downregulation (log2 FC ≥ 0.38 or ≤-0.38, adj. p < 0.05), particularly in proteins involved in cytotoxicity, cytolysis, and proteolysis, such as granzymes (GZMs) and perforin 1 (PRF1). This downregulation was observed in the benign group (GZMH, GZMM, and PRF1) and luminal B (GZMA, GZMH) subtypes, whereas granzyme K (GZMK) was upregulated in TNBC in comparison to healthy controls. The RNA degradation pathway was significantly downregulated (p < 0.05, normalized enrichment score (NES) from -1.47 to -1.80) across all BC subtypes, suggesting a potential mechanism for regulating gene expression during T cell activation. Also, the Sm-like proteins (LSM2, LSM3, and LSM5) were significantly downregulated in the RNA degradation pathway. Proteomic analysis of CD8+ T lymphocytes in peripheral blood across different breast cancer subtypes provides a comprehensive view of the molecular mechanisms of the systemic immune response that can significantly contribute to advancements in the diagnosis, treatment, and prognosis of this disease.
Collapse
Affiliation(s)
- Marek Lenárt
- 1st Department of Surgery, Faculty of Medicine, University of Pavol Jozef Šafárik and UNLP in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (M.L.); (N.M.); (J.R.)
| | - Peter Bober
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of Pavol Jozef Šafárik in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (M.M.); (S.T.); (M.A.)
| | - Miroslav Marcin
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of Pavol Jozef Šafárik in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (M.M.); (S.T.); (M.A.)
| | - Soňa Tkáčiková
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of Pavol Jozef Šafárik in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (M.M.); (S.T.); (M.A.)
| | - Mária Kacírová
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, University of Pavol Jozef Šafárik in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (M.K.); (J.F.)
| | - Michal Alexovič
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of Pavol Jozef Šafárik in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (M.M.); (S.T.); (M.A.)
| | - Dávid Tóth
- Department of Gynaecology and Obstetrics, Faculty of Medicine, University of Pavol Jozef Šafárik and UNLP in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (D.T.); (P.U.)
| | - Natália Madárová
- 1st Department of Surgery, Faculty of Medicine, University of Pavol Jozef Šafárik and UNLP in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (M.L.); (N.M.); (J.R.)
| | - Jozef Radoňak
- 1st Department of Surgery, Faculty of Medicine, University of Pavol Jozef Šafárik and UNLP in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (M.L.); (N.M.); (J.R.)
| | - Peter Urdzík
- Department of Gynaecology and Obstetrics, Faculty of Medicine, University of Pavol Jozef Šafárik and UNLP in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (D.T.); (P.U.)
| | - Ján Fedačko
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, University of Pavol Jozef Šafárik in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (M.K.); (J.F.)
| | - Ján Sabo
- Department of Medical and Clinical Biophysics, Faculty of Medicine, University of Pavol Jozef Šafárik in Košice, Trieda SNP 1, 04011 Košice, Slovakia; (M.M.); (S.T.); (M.A.)
| |
Collapse
|
14
|
Mosquera-Sulbaran JA, Pedreañez A, Vargas R, Hernandez-Fonseca JP. Apoptosis in post-streptococcal glomerulonephritis and mechanisms for failed of inflammation resolution. Pediatr Nephrol 2024; 39:1709-1724. [PMID: 37775580 DOI: 10.1007/s00467-023-06162-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 10/01/2023]
Abstract
Post-streptococcal glomerulonephritis is a condition resulting from infection by group A beta-hemolytic streptococcus. The main mechanism involves the formation of immune complexes formed in the circulation or in situ on the glomerular basement membrane, which activates complement and causes various inflammatory processes. Cellular mechanisms have been reported in the induction of kidney damage represented by the infiltration of innate cells (neutrophils and monocyte/macrophages) and adaptive cells (CD4 + lymphocytes and CD8 + lymphocytes) of the immune system. These cells induce kidney damage through various mechanisms. It has been reported that nephritogenic antigens are capable of inducing inflammatory processes early, even before the formation of immune complexes. Usually, this disease progresses towards clinical and renal normalization; however, in a smaller number of patients, it evolves into chronicity and persistent kidney damage. Hypotheses have been proposed regarding the mechanisms underlying this progression to chronicity including failure to induce apoptosis and failure to phagocytose apoptotic cells, allowing these cells to undergo membrane permeabilization and release pro-inflammatory molecules into the environment, thereby perpetuating renal inflammation. Other mechanisms involved include persistent infection, genetic background of the host's complement system, tubulointerstitial changes, and pre-existing kidney damage due to old age and comorbidities.
Collapse
Affiliation(s)
- Jesús A Mosquera-Sulbaran
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette," Facultad de Medicina, Universidad del Zulia, Apartado Postal: 23, Maracaibo, 4001-A, Zulia, Venezuela.
| | - Adriana Pedreañez
- Escuela de Bioanálisis, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | - Renata Vargas
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette," Facultad de Medicina, Universidad del Zulia, Apartado Postal: 23, Maracaibo, 4001-A, Zulia, Venezuela
| | - Juan Pablo Hernandez-Fonseca
- Instituto de Investigaciones Clínicas "Dr. Américo Negrette," Facultad de Medicina, Universidad del Zulia, Apartado Postal: 23, Maracaibo, 4001-A, Zulia, Venezuela
- Servicio de Microscopia Electrónica del Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| |
Collapse
|
15
|
Arya GC, Khalid M, Mehla S, Jakhmola V. A review of synthetic strategy, SAR, docking, simulation studies, and mechanism of action of isoxazole derivatives as anticancer agents. J Biomol Struct Dyn 2024; 42:4909-4935. [PMID: 37315986 DOI: 10.1080/07391102.2023.2220819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 05/29/2023] [Indexed: 06/16/2023]
Abstract
Breast cancer (BC) is a global health concern and the leading cause of cancerous death among women across the world, BC has been characterized by fresh lump in the breast or underarm (armpit), thickened or swollen. Worldwide estimated 9.6 million deaths in 2018-2019. Numerous drugs have been approved by FDA for BC treatment but showed numerous adverse effects like bioavailability issues, selectivity issues, and toxicity issues. Therefore, there is an immediate need to develop new molecules that are non-toxic and more efficient for treating cancer. Isoxazole derivatives have gained popularity over the few years due to their effective antitumor potential. These derivatives work against cancer by inhibiting the thymidylate enzyme, inducing apoptosis, inhibiting tubulin polymerization, protein kinase inhibition, and aromatase inhibition. In this study, we have concentrated on the isoxazole derivative with structure-activity relationship study, various synthesis techniques, mechanism of action, docking, and simulation studies pertaining to BC receptors. Hence the development of isoxazole derivatives with improved therapeutic efficacy will inspire further progress in improving human health.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Girish Chandra Arya
- University Institute of Pharmaceutical Sciences (UIPS), Chandigarh University, Mohali, India
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Shefali Mehla
- University Institute of Pharmaceutical Sciences (UIPS), Chandigarh University, Mohali, India
| | - Vikash Jakhmola
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| |
Collapse
|
16
|
Cigalotto L, Martinvalet D. Granzymes in health and diseases: the good, the bad and the ugly. Front Immunol 2024; 15:1371743. [PMID: 38646541 PMCID: PMC11026543 DOI: 10.3389/fimmu.2024.1371743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Granzymes are a family of serine proteases, composed of five human members: GA, B, H, M and K. They were first discovered in the 1980s within cytotoxic granules released during NK cell- and T cell-mediated killing. Through their various proteolytic activities, granzymes can trigger different pathways within cells, all of which ultimately lead to the same result, cell death. Over the years, the initial consideration of granzymes as mere cytotoxic mediators has changed due to surprising findings demonstrating their expression in cells other than immune effectors as well as new intracellular and extracellular activities. Additional roles have been identified in the extracellular milieu, following granzyme escape from the immunological synapse or their release by specific cell types. Outside the cell, granzyme activities mediate extracellular matrix alteration via the degradation of matrix proteins or surface receptors. In certain contexts, these processes are essential for tissue homeostasis; in others, excessive matrix degradation and extensive cell death contribute to the onset of chronic diseases, inflammation, and autoimmunity. Here, we provide an overview of both the physiological and pathological roles of granzymes, highlighting their utility while also recognizing how their unregulated presence can trigger the development and/or worsening of diseases.
Collapse
Affiliation(s)
- Lavinia Cigalotto
- Laboratory of Reactive Oxygen Species and Cytotoxic Immunity, Department Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute Of Molecular Medicine (VIMM), Padova, Italy
| | - Denis Martinvalet
- Laboratory of Reactive Oxygen Species and Cytotoxic Immunity, Department Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute Of Molecular Medicine (VIMM), Padova, Italy
| |
Collapse
|
17
|
Karisola P, Nikkola V, Joronen H, Ylianttila L, Grönroos M, Partonen T, Snellman E, Alenius H. Narrow-band UVB radiation triggers diverse changes in the gene expression and induces the accumulation of M1 macrophages in human skin. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 253:112887. [PMID: 38460430 DOI: 10.1016/j.jphotobiol.2024.112887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/22/2024] [Accepted: 03/05/2024] [Indexed: 03/11/2024]
Abstract
BACKGROUND The underlying molecular mechanisms that determine the biological effects of UVB radiation exposure on human skin are still only partially comprehended. OBJECTIVES Our goal is to examine the human skin transcriptome and related molecular mechanisms following a single exposure to UVB in the morning versus evening. METHODS We exposed 20 volunteer females to four-fold standard erythema doses (SED4) of narrow-band UVB (309-313 nm) in the morning or evening and studied skin transcriptome 24 h after the exposure. We performed enrichment analyses of gene pathways, predicted changes in skin cell composition using cellular deconvolution, and correlated cell proportions with gene expression. RESULTS In the skin transcriptome, UVB exposure yielded 1384 differentially expressed genes (DEGs) in the morning and 1295 DEGs in the evening, of which the most statistically significant DEGs enhanced proteasome and spliceosome pathways. Unexposed control samples showed difference by 321 DEGs in the morning vs evening, which was related to differences in genes associated with the circadian rhythm. After the UVB exposure, the fraction of proinflammatory M1 macrophages was significantly increased at both timepoints, and this increase was positively correlated with pathways on Myc targets and mTORC1 signaling. In the evening, the skin clinical erythema was more severe and had stronger positive correlation with the number of M1 macrophages than in the morning after UVB exposure. The fractions of myeloid and plasmacytoid dendritic cells and CD8 T cells were significantly decreased in the morning but not in the evening. CONCLUSIONS NB-UVB-exposure causes changes in skin transcriptome, inhibiting cell division, and promoting proteasome activity and repair responses, both in the morning and in the evening. Inflammatory M1 macrophages may drive the UV-induced skin responses by exacerbating inflammation and erythema. These findings highlight how the same UVB exposure influences skin responses differently in morning versus evening and presents a possible explanation to the differences in gene expression in the skin after UVB irradiation at these two timepoints.
Collapse
Affiliation(s)
- Piia Karisola
- Faculty of Medicine, Human Microbiome Research Program, University of Helsinki, Finland.
| | - Veera Nikkola
- Tampere University, Faculty of Medicine and Health Technology, Department of Dermatology and Venereology, Tampere, Finland
| | - Heli Joronen
- Tampere University, Faculty of Medicine and Health Technology, Department of Dermatology and Venereology, Tampere, Finland; Päijät-Häme Social and Health Care Group, Department of Dermatology and Allergology, Lahti, Finland.
| | - Lasse Ylianttila
- Radiation and Nuclear Safety Authority (STUK), Helsinki, Finland.
| | - Mari Grönroos
- Päijät-Häme Social and Health Care Group, Department of Dermatology and Allergology, Lahti, Finland.
| | - Timo Partonen
- Finnish Institute for Health and Welfare, Department of Public Health and Welfare, Finland.
| | - Erna Snellman
- Tampere University, Faculty of Medicine and Health Technology, Department of Dermatology and Venereology, Tampere, Finland.
| | - Harri Alenius
- Faculty of Medicine, Human Microbiome Research Program, University of Helsinki, Finland; Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
18
|
Rasi V, Phelps KR, Paulson KR, Eickhoff CS, Chinnaraj M, Pozzi N, Di Gioia M, Zanoni I, Shakya S, Carlson HL, Ford DA, Kolar GR, Hoft DF. Homodimeric Granzyme A Opsonizes Mycobacterium tuberculosis and Inhibits Its Intracellular Growth in Human Monocytes via Toll-Like Receptor 4 and CD14. J Infect Dis 2024; 229:876-887. [PMID: 37671668 PMCID: PMC10938207 DOI: 10.1093/infdis/jiad378] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/18/2023] [Accepted: 09/04/2023] [Indexed: 09/07/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb)-specific γ9δ2 T cells secrete granzyme A (GzmA) protective against intracellular Mtb growth. However, GzmA-enzymatic activity is unnecessary for pathogen inhibition, and the mechanisms of GzmA-mediated protection remain unknown. We show that GzmA homodimerization is essential for opsonization of mycobacteria, altered uptake into human monocytes, and subsequent pathogen clearance within the phagolysosome. Although monomeric and homodimeric GzmA bind mycobacteria, only homodimers also bind cluster of differentiation 14 (CD14) and Toll-like receptor 4 (TLR4). Without access to surface-expressed CD14 and TLR4, GzmA fails to inhibit intracellular Mtb. Upregulation of Rab11FIP1 was associated with inhibitory activity. Furthermore, GzmA colocalized with and was regulated by protein disulfide isomerase AI (PDIA1), which cleaves GzmA homodimers into monomers and prevents Mtb inhibitory activity. These studies identify a previously unrecognized role for homodimeric GzmA structure in opsonization, phagocytosis, and elimination of Mtb in human monocytes, and they highlight PDIA1 as a potential host-directed therapy for prevention and treatment of tuberculosis, a major human disease.
Collapse
Affiliation(s)
- Valerio Rasi
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
- Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Kathleen R Phelps
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
- Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Keegan R Paulson
- Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Christopher S Eickhoff
- Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Mathivanan Chinnaraj
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Nicola Pozzi
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Marco Di Gioia
- Harvard Medical School and Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Ivan Zanoni
- Harvard Medical School and Division of Immunology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Shubha Shakya
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Haley L Carlson
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - David A Ford
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Grant R Kolar
- Department of Pathology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Daniel F Hoft
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
- Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
19
|
Montalvo MJ, Bandey IN, Rezvan A, Wu KL, Saeedi A, Kulkarni R, Li Y, An X, Sefat KMSR, Varadarajan N. Decoding the mechanisms of chimeric antigen receptor (CAR) T cell-mediated killing of tumors: insights from granzyme and Fas inhibition. Cell Death Dis 2024; 15:109. [PMID: 38307835 PMCID: PMC10837176 DOI: 10.1038/s41419-024-06461-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 02/04/2024]
Abstract
Chimeric antigen receptor (CAR) T cell show promise in cancer treatments, but their mechanism of action is not well understood. Decoding the mechanisms used by individual T cells can help improve the efficacy of T cells while also identifying mechanisms of T cell failure leading to tumor escape. Here, we used a suite of assays including dynamic single-cell imaging of cell-cell interactions, dynamic imaging of fluorescent reporters to directly track cytotoxin activity in tumor cells, and scRNA-seq on patient infusion products to investigate the cytotoxic mechanisms used by individual CAR T cells in killing tumor cells. We show that surprisingly, overexpression of the Granzyme B (GZMB) inhibitor, protease inhibitor-9 (PI9), does not alter the cytotoxicity mediated by CD19-specific CAR T cells against either the leukemic cell line, NALM6; or the ovarian cancer cell line, SkOV3-CD19. We designed and validated reporters to directly assay T cell delivered GZMB activity in tumor cells and confirmed that while PI9 overexpression inhibits GZMB activity at the molecular level, this is not sufficient to impact the kinetics or magnitude of killing mediated by the CAR T cells. Altering cytotoxicity mediated by CAR T cells required combined inhibition of multiple pathways that are tumor cell specific: (a) B-cell lines like NALM6, Raji and Daudi were sensitive to combined GZMB and granzyme A (GZMA) inhibition; whereas (b) solid tumor targets like SkOV3-CD19 and A375-CD19 (melanoma) were sensitive to combined GZMB and Fas ligand inhibition. We realized the translational relevance of these findings by examining the scRNA-seq profiles of Tisa-cel and Axi-cel infusion products and show a significant correlation between GZMB and GZMA expression at the single-cell level in a T cell subset-dependent manner. Our findings highlight the importance of the redundancy in killing mechanisms of CAR T cells and how this redundancy is important for efficacious T cells.
Collapse
Affiliation(s)
- Melisa J Montalvo
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Irfan N Bandey
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Ali Rezvan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Kwan-Ling Wu
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Arash Saeedi
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Rohan Kulkarni
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Yongshuai Li
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Xingyue An
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - K M Samiur Rahman Sefat
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Navin Varadarajan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA.
| |
Collapse
|
20
|
Liu LY, He SJ, Chen Z, Ge M, Lyu CY, Gao D, Yu JP, Cai MH, Yuan JX, Zhang JL. The Role of Regulatory Cell Death in Vitiligo. DNA Cell Biol 2024; 43:61-73. [PMID: 38153369 DOI: 10.1089/dna.2023.0188] [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] [Indexed: 12/29/2023] Open
Abstract
Vitiligo is one of the common chronic autoimmune skin diseases in clinic, which is characterized by localized or generalized depigmentation and seriously affects the physical and mental health of patients. At present, the pathogenesis of vitiligo is not clear; mainly, heredity, autoimmunity, oxidative stress, melanocyte (MC) self-destruction, and the destruction, death, or dysfunction of MCs caused by various reasons are always the core of vitiligo. Regulatory cell death (RCD) is an active and orderly death mode of cells regulated by genes, which widely exists in various life activities, plays a pivotal role in maintaining the homeostasis of the organism, and is closely related to the occurrence and development of many diseases. With the deepening of the research and understanding of RCD, people gradually found that there are many different forms of RCD in the lesions and perilesional skin of vitiligo patients, such as apoptosis, autophagy, pyroptosis, ferroptosis, and so on. Different cell death modes have different mechanisms in vitiligo, and different RCDs can interact and regulate each other. In this article, the mechanism related to RCD in the pathogenesis of vitiligo is reviewed, which provides new ideas for exploring the pathogenesis and targeted treatment of vitiligo.
Collapse
Affiliation(s)
- Lyu-Ye Liu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Si-Jia He
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, People's Republic of China
| | - Zhao Chen
- First Clinical Medical College Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Man Ge
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Chun-Yi Lyu
- First Clinical Medical College Shandong University of Traditional Chinese Medicine, Jinan, People's Republic of China
| | - Dandan Gao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Ji-Peng Yu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Meng-Han Cai
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Jin-Xiang Yuan
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Jun-Ling Zhang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, People's Republic of China
| |
Collapse
|
21
|
He S, Su L, Hu H, Liu H, Xiong J, Gong X, Chi H, Wu Q, Yang G. Immunoregulatory functions and therapeutic potential of natural killer cell-derived extracellular vesicles in chronic diseases. Front Immunol 2024; 14:1328094. [PMID: 38239346 PMCID: PMC10795180 DOI: 10.3389/fimmu.2023.1328094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
Extracellular vesicles (EVs) have been proven to play a significant immunoregulatory role in many chronic diseases, such as cancer and immune disorders. Among them, EVs derived from NK cells are an essential component of the immune cell functions. These EVs have been demonstrated to carry a variety of toxic proteins and nucleic acids derived from NK cells and play a therapeutic role in diseases like malignancies, liver fibrosis, and lung injury. However, natural NK-derived EVs (NKEVs) have certain limitations in disease treatment, such as low yield and poor targeting. Concurrently, NK cells exhibit characteristics of memory-like NK cells, which have stronger proliferative capacity, increased IFN-γ production, and enhanced cytotoxicity, making them more advantageous for disease treatment. Recent research has shifted its focus towards engineered extracellular vesicles and their potential to improve the efficiency, specificity, and safety of disease treatments. In this review, we will discuss the characteristics of NK-derived EVs and the latest advancements in disease therapy. Specifically, we will compare different cellular sources of NKEVs and explore the current status and prospects of memory-like NK cell-derived EVs and engineered NKEVs.
Collapse
Affiliation(s)
- Shuang He
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Lanqian Su
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Haiyang Hu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Haiqi Liu
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jingwen Xiong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Xiangjin Gong
- Department of Sports Rehabilitation, Southwest Medical University, Luzhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Qibiao Wu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Guanhu Yang
- Department of Specialty Medicine, Ohio University, Athens, OH, United States
| |
Collapse
|
22
|
Wang Y, Liu Y, Fang J, Xing X, Wang H, Shi X, Liu X, Niu T, Liu K. Small-molecule agonist AdipoRon alleviates diabetic retinopathy through the AdipoR1/AMPK/EGR4 pathway. J Transl Med 2024; 22:2. [PMID: 38166990 PMCID: PMC10759471 DOI: 10.1186/s12967-023-04783-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is a progressive disease that involves multiple organs due to increased blood glucose, and diabetic retinopathy (DR) is the main complication of DM in the eyes and causes irreversible vision loss. In the pathogenesis of diabetic vascular disease, oxidative stress caused by hyperglycemia plays an important role in Müller cell impairment. In recent years, AdipoRon, an adiponectin analog that demonstrated important physiological functions in obesity, diabetes, inflammation, and cardiovascular diseases, demonstrated cellular protection from apoptosis and reduced inflammatory damage through a receptor-dependent mechanism. Here, we investigated how AdipoRon reduced oxidative stress and apoptosis in Müller glia in a high glucose environment. RESULTS By binding to adiponectin receptor 1 on Müller glia, AdipoRon activated 5' adenosine monophosphate-activated protein kinase (AMPK)/acetyl-CoA carboxylase phosphorylation downstream, thereby alleviating oxidative stress and eventual apoptosis of cells and tissues. Transcriptome sequencing revealed that AdipoRon promoted the synthesis and expression of early growth response factor 4 (EGR4) and inhibited the cellular protective effects of AdipoRon in a high-glucose environment by reducing the expression of EGR4. This indicated that AdipoRon played a protective role through the EGR4 and classical AMPK pathways. CONCLUSIONS This provides a new target for the early treatment of DR.
Collapse
Affiliation(s)
- Yihan Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
- Department of Ophthalmology, Shanghai Renji Hospital, School of Medicine, Shanghai, 200127, China
| | - Yujuan Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
| | - Junwei Fang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
| | - Xindan Xing
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
| | - Hanying Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
| | - Xin Shi
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
| | - Xinyi Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
| | - Tian Niu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China.
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China.
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China.
| | - Kun Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China.
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China.
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China.
| |
Collapse
|
23
|
Li Y, Zhong G, Li L, Li T, Li H, Li Y, Zhang H, Pan J, Hu L, Liao J, Yu W, Tang Z. MitomiR-1736-3p regulates copper-induced mitochondrial pathway apoptosis by inhibiting AATF in chicken hepatocytes. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 906:167825. [PMID: 37839473 DOI: 10.1016/j.scitotenv.2023.167825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 10/17/2023]
Abstract
Copper (Cu) is a toxic heavy metal pollutant. The hepatic toxicity of Cu has attracted widespread attention from researchers. However, its underlying mechanism remains elusive. Mitochondrial microRNAs (mitomiRs) are considered important factors in regulating mitochondrial and cellular functions, and their roles have been implicated in the mechanisms of metal toxicity. Therefore, this research revealed the changes in the mitomiRs expression profile of chicken liver after Cu exposure. It was ultimately determined that mitomiR-1736-3p can be involved in Cu-induced chicken liver damage by targeting AATF. In particular, our investigations have uncovered that exposure to Cu can trigger heightened levels of apoptosis in the hepatic tissue of chickens and primary chicken embryo hepatocytes (CEHs). It is noteworthy that we found upregulation of miR-1736-3p expression can exacerbate Cu-induced cell apoptosis, while inhibition of miR-1736-3p can effectively reduce apoptosis occurrence. Subsequently, we found that Cu-induced cell apoptosis could be restored by overexpressing AATF, while silencing AATF exacerbated the level of apoptosis. Fascinatingly, this change in apoptotic level is directly influenced by AATF on Bax and Bak1, rather than on p53 and Bcl-2. Overall, these findings suggest that the mitomiR-1736-3p/AATF axis mediates the mitochondrial pathway of cell apoptosis potentially involved in Cu-induced chicken liver toxicity.
Collapse
Affiliation(s)
- Yuanxu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Gaolong Zhong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Lei Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Tingyu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Huayu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Jiaqiang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Wenlan Yu
- Laboratory Animal Center, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, Guangdong, PR China.
| |
Collapse
|
24
|
Shan Y, Guan C, Wang J, Qi W, Chen A, Liu S. Impact of ferroptosis on preeclampsia: A review. Biomed Pharmacother 2023; 167:115466. [PMID: 37729725 DOI: 10.1016/j.biopha.2023.115466] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/03/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
Preeclampsia (PE) is usually associated with the accumulation of reactive oxygen species (ROS) resulting from heightened oxidative stress (OS). Ferroptosis is a unique type of lipid peroxidation-induced iron-dependent cell death distinct from traditional apoptosis, necroptosis, and pyroptosis and most likely contributes considerable to PE pathogenesis. At approximately 10-12 weeks of gestation, trophoblasts create an environment rich in oxygen and iron. In patients with PE, ferroptosis-related genes such as HIF1 and MAPK8 are downregulated, whereas PLIN2 is upregulated. Furthermore, miR-30b-5p overexpression inhibits solute carrier family 11 member 2, resulting in a decrease in glutathione levels and an increase in the labile iron pool. At the maternal-fetal interface, physiological hypoxia/reperfusion and excessive iron result in lipid peroxidation and ROS production. Owing to the high expression of Fpn and polyunsaturated fatty acid-containing phospholipid-related enzymes, including acyl-CoA synthetase long-chain family member 4, lysophosphatidylcholine acyl-transferase 3, and spermidine/spermine N1-acetyltransferase 1, trophoblasts become more susceptible to OS and ROS damage. In stage 1, the injured trophoblasts exhibit poor invasion and incomplete uterine spiral artery remodeling caused by ferroptosis, leading to placental ischemia and hypoxia. Subsequently, ferroptosis marked by OS occurs in stage 2, eventually causing PE. We aimed to explore the new therapeutic target of PE through OS in ferroptosis.
Collapse
Affiliation(s)
- Yuping Shan
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chengcheng Guan
- Laboratory Department, Qingdao Haici Hospital, Qingdao, China
| | - Jingli Wang
- Department of Medical Genetics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Weihong Qi
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Aiping Chen
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Shiguo Liu
- Department of Medical Genetics, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
25
|
Ge W, Wang H, Wu X, Dong B, Zhang R, Tian M. Construction of a Dual-Emissive Probe for Discriminative Visualization of Lysosomal and Mitochondrial Dysfunction. Anal Chem 2023; 95:14787-14796. [PMID: 37726214 DOI: 10.1021/acs.analchem.3c03024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
Discriminatively visualizing mitochondrial and lysosomal dysfunction is crucial for an in-depth understanding of cell apoptosis regulation and relative biology. However, fluorescent probes for the separate visualization of lysosomal and mitochondria damages have not been reported yet. Herein, we have constructed a fluorescent probe [2-(4-hydroxystyryl)-1,3,3-trimethyl-3H-indol-1-ium iodide (HBSI)] for labeling mitochondria and lysosomes in dual emission colors and discriminatively imaging mitochondrial and lysosomal damage in two different sets of fluorescent signals. In living cells, HBSI targeted both lysosomes and mitochondria to give green and red emission, respectively. During mitochondrial damages, HBSI immigrated into lysosomes, and the red emission decreased. During lysosomal damage, HBSI immigrated into mitochondria, and the green emission decreased. With the robust probe, the different damaging sequences of mitochondria and lysosomes under different amounts of H2O2 and chloral hydrate have been revealed. The sequential damage of lysosomes and mitochondria during cell apoptosis induced by rotenone, paclitaxel, and colchicine has been discovered. Furthermore, the regulation of mitochondria, lysosome, and their interplay during autophagy was also observed with the probe.
Collapse
Affiliation(s)
- Wei Ge
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China
| | - Huina Wang
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China
| | - Xiaofen Wu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China
| | - Baoli Dong
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China
| | - Ruoyao Zhang
- School of Medical Technology, Institute of Engineering Medicine, School of Life Science, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Minggang Tian
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China
| |
Collapse
|
26
|
Du H, Meng S, Geng M, Zhao P, Gong L, Zheng X, Li X, Yuan Z, Yang H, Zhao Y, Dai L. Detachable MOF-Based Core/Shell Nanoreactor for Cancer Dual-Starvation Therapy With Reversing Glucose and Glutamine Metabolisms. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2303253. [PMID: 37330663 DOI: 10.1002/smll.202303253] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/19/2023] [Indexed: 06/19/2023]
Abstract
Tumor-dependent glucose and glutamine metabolisms are essential for maintaining survival, while the accordingly metabolic suppressive therapy is limited by the compensatory metabolism and inefficient delivery efficiency. Herein, a functional metal-organic framework (MOF)-based nanosystem composed of the weakly acidic tumor microenvironment-activated detachable shell and reactive oxygen species (ROS)-responsive disassembled MOF nanoreactor core is designed to co-load glycolysis and glutamine metabolism inhibitors glucose oxidase (GOD) and bis-2-(5-phenylacetmido-1,2,4-thiadiazol-2-yl) ethyl sulfide (BPTES) for tumor dual-starvation therapy. The nanosystem excitingly improves tumor penetration and cellular uptake efficiency via integrating the pH-responsive size reduction and charge reversal and ROS-sensitive MOF disintegration and drug release strategy. Furthermore, the degradation of MOF and cargoes release can be self-amplified via additional self-generation H2 O2 mediated by GOD. Last, the released GOD and BPTES collaboratively cut off the energy supply of tumors and induce significant mitochondrial damage and cell cycle arrest via simultaneous restriction of glycolysis and compensatory glutamine metabolism pathways, consequently realizing the remarkable triple negative breast cancer killing effect in vivo with good biosafety via the dual starvation therapy.
Collapse
Affiliation(s)
- Huiping Du
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Siyu Meng
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Meijuan Geng
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Pan Zhao
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Liyang Gong
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Xinmin Zheng
- School of Life Science, Northwestern Polytechnical University, Xian, 710072, China
| | - Xiang Li
- School of Life Science, Northwestern Polytechnical University, Xian, 710072, China
| | - Zhang Yuan
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| | - Hui Yang
- School of Life Science, Northwestern Polytechnical University, Xian, 710072, China
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Liangliang Dai
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, P. R. China
| |
Collapse
|
27
|
Rao S, Liu M, Iosef C, Knutsen C, Alvira CM. Endothelial-specific loss of IKKβ disrupts pulmonary endothelial angiogenesis and impairs postnatal lung growth. Am J Physiol Lung Cell Mol Physiol 2023; 325:L299-L313. [PMID: 37310763 PMCID: PMC10625829 DOI: 10.1152/ajplung.00034.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/14/2023] Open
Abstract
Pulmonary angiogenesis drives alveolarization, but the transcriptional regulators directing pulmonary angiogenesis remain poorly defined. Global, pharmacological inhibition of nuclear factor-kappa B (NF-κB) impairs pulmonary angiogenesis and alveolarization. However, establishing a definitive role for NF-κB in pulmonary vascular development has been hindered by embryonic lethality induced by constitutive deletion of NF-κB family members. We created a mouse model allowing inducible deletion of the NF-κB activator, IKKβ, in endothelial cells (ECs) and assessed the effect on lung structure, endothelial angiogenic function, and the lung transcriptome. Embryonic deletion of IKKβ permitted lung vascular development but resulted in a disorganized vascular plexus, while postnatal deletion significantly decreased radial alveolar counts, vascular density, and proliferation of both endothelial and nonendothelial lung cells. Loss of IKKβ impaired survival, proliferation, migration, and angiogenesis in primary lung ECs in vitro, in association with decreased expression of VEGFR2 and activation of downstream effectors. Loss of endothelial IKKβ in vivo induced broad changes in the lung transcriptome with downregulation of genes related to mitotic cell cycle, extracellular matrix (ECM)-receptor interaction, and vascular development, and the upregulation of genes related to inflammation. Computational deconvolution suggested that loss of endothelial IKKβ decreased general capillary, aerocyte capillary, and alveolar type I cell abundance. Taken together, these data definitively establish an essential role for endogenous endothelial IKKβ signaling during alveolarization. A deeper understanding of the mechanisms directing this developmental, physiological activation of IKKβ in the lung vasculature may provide novel targets for the development of strategies to enhance beneficial proangiogenic signaling in lung development and disease.NEW & NOTEWORTHY This study highlights the cell-specific complexity of nuclear factor kappa B signaling in the developing lung by demonstrating that inducible loss of IKKβ in endothelial cells impairs alveolarization, disrupts EC angiogenic function, and broadly represses genes important for vascular development.
Collapse
Affiliation(s)
- Shailaja Rao
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Min Liu
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Cristiana Iosef
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Carsten Knutsen
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| | - Cristina M Alvira
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, United States
- Stanford Center for Excellence in Pulmonary Biology, Palo Alto, California, United States
| |
Collapse
|
28
|
Duquette D, Harmon C, Zaborowski A, Michelet X, O'Farrelly C, Winter D, Koay HF, Lynch L. Human Granzyme K Is a Feature of Innate T Cells in Blood, Tissues, and Tumors, Responding to Cytokines Rather than TCR Stimulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:633-647. [PMID: 37449888 DOI: 10.4049/jimmunol.2300083] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/02/2023] [Indexed: 07/18/2023]
Abstract
NK cells and CD8 T cells use cytotoxic molecules to kill virally infected and tumor cell targets. While perforin and granzyme B (GzmB) are the most commonly studied lytic molecules, less is known about granzyme K (GzmK). However, this granzyme has been recently associated with improved prognosis in solid tumors. In this study, we show that, in humans, GzmK is predominantly expressed by innate-like lymphocytes, as well as a newly identified population of GzmK+CD8+ non- mucosal-associated invariant T cells with innate-like characteristics. We found that GzmK+ T cells are KLRG1+EOMES+IL-7R+CD62L-Tcf7int, suggesting that they are central memory T and effector memory T cells. Furthermore, GzmK+ cells are absent/low in cord blood, suggesting that GzmK is upregulated with immune experience. Surprisingly, GzmK+ cells respond to cytokine stimuli alone, whereas TCR stimulation downregulates GzmK expression, coinciding with GzmB upregulation. GzmK+ cells have reduced IFN-γ production compared with GzmB+ cells in each T cell lineage. Collectively, this suggests that GzmK+ cells are not naive, and they may be an intermediate memory-like or preterminally differentiated population. GzmK+ cells are enriched in nonlymphoid tissues such as the liver and adipose. In colorectal cancer, GzmK+ cells are enriched in the tumor and can produce IFN-γ, but GzmK+ expression is mutually exclusive with IL-17a production. Thus, in humans, GzmK+ cells are innate memory-like cells that respond to cytokine stimulation alone and may be important effector cells in the tumor.
Collapse
Affiliation(s)
- Danielle Duquette
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Cathal Harmon
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | | | - Xavier Michelet
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
| | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Des Winter
- St. Vincent's University Hospital, Dublin, Ireland
| | - Hui-Fern Koay
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Austria
| | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- St. Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
29
|
Wang X, Shields CA, Ekperikpe U, Amaral LM, Williams JM, Cornelius DC. VASCULAR AND RENAL MECHANISMS OF PREECLAMPSIA. CURRENT OPINION IN PHYSIOLOGY 2023; 33:100655. [PMID: 37009057 PMCID: PMC10062189 DOI: 10.1016/j.cophys.2023.100655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Preeclampsia (PE) is a multisystem obstetric disorder that affects 2-10% of pregnancies worldwide and it is a leading cause of maternal and fetal morbidity and mortality. The etiology of PE development is not clearly delineated, but since delivery of the fetus and placenta often leads to symptom resolution in the most cases of PE, it is hypothesized that the placenta is the inciting factor of the disease. Current management strategies for PE focus on treating the maternal symptoms to stabilize the mother in an attempt to prolong the pregnancy. However, the efficacy of this management strategy is limited. Therefore, identification of novel therapeutic targets and strategies is needed. Here, we provide a comprehensive overview of the current state of knowledge regarding mechanisms of vascular and renal pathophysiology during PE and discuss potential therapeutic targets directed at improving maternal vascular and renal function.
Collapse
Affiliation(s)
- Xi Wang
- Department of Pharmacology, University of Mississippi Medical Center
| | - Corbin A Shields
- Department of Emergency Medicine, University of Mississippi Medical Center
| | - Ubong Ekperikpe
- Department of Pharmacology, University of Mississippi Medical Center
| | - Lorena M Amaral
- Department of Pharmacology, University of Mississippi Medical Center
| | | | - Denise C Cornelius
- Department of Pharmacology, University of Mississippi Medical Center
- Department of Emergency Medicine, University of Mississippi Medical Center
| |
Collapse
|
30
|
Sobierajska P, Wiatrak B, Jawien P, Janeczek M, Wiglusz K, Szeląg A, Wiglusz RJ. Imatinib-Functionalized Galactose Hydrogels Loaded with Nanohydroxyapatite as a Drug Delivery System for Osteosarcoma: In Vitro Studies. ACS OMEGA 2023; 8:17891-17900. [PMID: 37251195 PMCID: PMC10210190 DOI: 10.1021/acsomega.3c00986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/26/2023] [Indexed: 05/31/2023]
Abstract
This study reports an impact of structure (XRPD, FT-IR) and surface morphology (SEM-EDS) of imatinib-functionalized galactose hydrogels, loaded and unloaded with nHAp, on osteosarcoma cell (Saos-2 and U-2OS) viability, levels of free oxygen radicals, and nitric oxide, levels of BCL-2, p53, and caspase 3 and 9, as well as glycoprotein-P activity. It was investigated how the rough surface of the crystalline hydroxyapatite-modified hydrogel affected amorphous imatinib (IM) release. The imatinib drug effect on cell cultures has been demonstrated in different forms of administration-directly to the culture or the hydrogels. Administration of IM and hydrogel composites could be expected to reduce the risk of multidrug resistance development by inhibiting Pgp.
Collapse
Affiliation(s)
- Paulina Sobierajska
- Institute
of Low Temperature and Structure Research, Polish Academy of Sciences, Okolna 2, Wroclaw 50-422, Poland
| | - Benita Wiatrak
- Department
of Pharmacology, Wroclaw Medical University, Mikulicza-Radeckiego 2, Wroclaw 50-345, Poland
| | - Paulina Jawien
- Department
of Biostructure and Animal Physiology, Wroclaw
University of Environmental and Life Sciences, Norwida 25/27, 50-375 Wroclaw, Poland
| | - Maciej Janeczek
- Department
of Biostructure and Animal Physiology, Wroclaw
University of Environmental and Life Sciences, Norwida 25/27, 50-375 Wroclaw, Poland
| | - Katarzyna Wiglusz
- Department
of Basic Chemical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211 A, 50-566 Wroclaw, Poland
| | - Adam Szeląg
- Department
of Pharmacology, Wroclaw Medical University, Mikulicza-Radeckiego 2, Wroclaw 50-345, Poland
| | - Rafal J. Wiglusz
- Institute
of Low Temperature and Structure Research, Polish Academy of Sciences, Okolna 2, Wroclaw 50-422, Poland
| |
Collapse
|
31
|
Cuesta LM, Verna A, Pérez S. Modulation of granzymes mRNA expression in neural tissue of BoAHV-1 and BoAHV-5-infected cattle. Vet Immunol Immunopathol 2023; 260:110610. [PMID: 37196596 DOI: 10.1016/j.vetimm.2023.110610] [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: 04/20/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/19/2023]
Abstract
Bovine alphaherpesviruses (BoAHV)-1 and 5 are neurotropic viruses of cattle which differ in their neuropathogenic potential. BoAHV-5 is responsible for non-suppurative meningoencephalitis in calves while BoAHV-1 can occasionally cause encephalitis. Granzymes (GZMs) are serine-proteases that participate in CD8+ T cells-mediated killing of virally-infected cells upon release through perforin (PFN)-formed pores in the cell membrane. Recently, six GZMs have been identified in cattle (A, B, K, H, M and O). However, their expression in bovine tissues has not been evaluated. In this study, the mRNA expression levels of PFN and GZMs A, B, K, H and M in the nervous system of calves experimentally-inoculated with BoAHV-1 or BoAHV-5 were analyzed at the three distinct stages of the infectious cycle of alphaherpesviruses: acute infection, latency and reactivation. This is the first report describing the expression of GZMs in bovine neural tissue and the first analysis of GZM expression in the context of bovine alphaherpesviruses neuropathogenesis. The findings revealed that PFN and GZM K are upregulated during BoAHV-1 or BoAHV-5 acute infection. In contrast to BoAHV-1, during BoAHV-5 latency a significant up-regulation of PFN, GZM K and GZM H was detected. PFN and GZM A, K and H expression was also up-regulated during BoAHV-5 reactivation. Therefore, a distinct pattern of PFN and GZM expression is evident along the infectious cycle of each alphaherpesvirus and this might contribute to the differences in BoAHV-1 and BoAHV-5 neuropathogenesis.
Collapse
Affiliation(s)
- Lucía Martínez Cuesta
- Centro de Investigación Veterinaria de Tandil (CIVETAN)-CONICET, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, Paraje Arroyo Seco S/N, Tandil 7000, Argentina
| | - Andrea Verna
- Área de Producción Animal, Instituto de Innovación para la Producción Agropecuaria y Desarrollo Sostenible (IPADS, CONICET-INTA), Ruta Nacional 226 km 73,5, Balcarce, 7620 Buenos Aires, Argentina
| | - Sandra Pérez
- Centro de Investigación Veterinaria de Tandil (CIVETAN)-CONICET, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, Paraje Arroyo Seco S/N, Tandil 7000, Argentina.
| |
Collapse
|
32
|
Diogo P, Martins G, Simão M, Marreiros A, Eufrásio AC, Cabrita E, Gavaia PJ. Type I Diabetes in Zebrafish Reduces Sperm Quality and Increases Insulin and Glucose Transporter Transcripts. Int J Mol Sci 2023; 24:ijms24087035. [PMID: 37108202 PMCID: PMC10138585 DOI: 10.3390/ijms24087035] [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: 03/10/2023] [Revised: 04/03/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Type I diabetes is a prominent human pathology with increasing incidence in the population; however, its cause is still unknown. This disease promotes detrimental effects on reproduction, such as lower sperm motility and DNA integrity. Hence, the investigation of the underlying mechanisms of this metabolic disturbance in reproduction and its transgenerational consequences is of the utmost importance. The zebrafish is a useful model for this research considering its high homology with human genes as well as its fast generation and regeneration abilities. Therefore, we aimed to investigate sperm quality and genes relevant to diabetes in the spermatozoa of Tg(ins:nfsb-mCherry) zebrafish, a model for type I diabetes. Diabetic Tg(ins:nfsb-mCherry) males showed significantly higher expression of transcripts for insulin a (insa) and glucose transporter (slc2a2) compared to controls. Sperm obtained from the same treatment group showed significantly lower sperm motility, plasma membrane viability, and DNA integrity compared to that from the control group. Upon sperm cryopreservation, sperm freezability was reduced, which could be a consequence of poor initial sperm quality. Altogether, the data showed similar detrimental effects related to type I diabetes in zebrafish spermatozoa at the cellular and molecular levels. Therefore, our study validates the zebrafish model for type I diabetes research in germ cells.
Collapse
Affiliation(s)
- Patrícia Diogo
- Faculty of Sciences and Technology (FCT), University of Algarve, 8005-139 Faro, Portugal
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
- Necton-Companhia Portuguesa de Culturas Marinhas S.A, Belamandil s/n, 8700-152 Olhão, Portugal
| | - Gil Martins
- Faculty of Sciences and Technology (FCT), University of Algarve, 8005-139 Faro, Portugal
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139 Faro, Portugal
| | - Márcio Simão
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139 Faro, Portugal
| | - Ana Marreiros
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, 8005-139 Faro, Portugal
| | - Ana Catarina Eufrásio
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), 4200-135 Porto, Portugal
| | - Elsa Cabrita
- Faculty of Sciences and Technology (FCT), University of Algarve, 8005-139 Faro, Portugal
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
| | - Paulo Jorge Gavaia
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139 Faro, Portugal
| |
Collapse
|
33
|
McKenzie B, Valitutti S. Resisting T cell attack: tumor-cell-intrinsic defense and reparation mechanisms. Trends Cancer 2023; 9:198-211. [PMID: 36593148 DOI: 10.1016/j.trecan.2022.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/01/2022] [Accepted: 12/07/2022] [Indexed: 01/02/2023]
Abstract
Cytotoxic T lymphocytes (CTLs) are antigen-specific killer cells equipped to identify and eliminate host cells that have been altered through infection or transformation. Both chimeric antigen-receptor (CAR) T cell therapies and immune checkpoint blockade (ICB) therapies are based on successful elimination of tumor cells by cytotoxic effectors. In this opinion article, we outline cell-intrinsic mechanisms by which tumor cells defend against CTLs, highlighting pathways that confer resistance and proposing opportunities for combination therapies. We discuss how exogenous killing entities [e.g., supramolecular attack particles (SMAPs)] offer a novel strategy to circumvent cellular resistance mechanisms. Our opinion article highlights the importance of identifying, quantifying, and targeting tumor defense mechanisms at the interface between tumor cells and CTLs as a critical consideration in the development of immunotherapy approaches.
Collapse
Affiliation(s)
- Brienne McKenzie
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, 31057 Toulouse, France.
| | - Salvatore Valitutti
- INSERM U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, 31057 Toulouse, France; Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, 31059 Toulouse, France.
| |
Collapse
|
34
|
Zhang L, Li L, Huang L, Li X, Xu C, Hu W, Sun Y, Liu F, Li Y. Voltage-dependent anion channel 2 (VDAC2) facilitates the accumulation of rice stripe virus in the vector Laodelphax striatellus. Virus Res 2023; 324:199019. [PMID: 36496034 DOI: 10.1016/j.virusres.2022.199019] [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: 10/15/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
Rice stripe virus (RSV) causes enormous losses in rice production and is transmitted by the small brown planthopper, Laodelphax striatellus, in a persistent-propagative manner. RSV accumulation within the gut lumen of the vector is indispensable for the successful transmission to rice and insects. In this study, we obtained a 1464 bp full-length cDNA of a voltage-dependent anion channel 2 from L. striatellus (LsVDAC2), which encodes a 283 amino acid protein. RSV infection increased the expression of LsVDAC2 in the midguts and ovaries of L. striatellus by 260% and 228%, respectively. Silencing of LsVDAC2 resulted in a 88% reduction of RSV loads at 24 h after RNAi, indicating that LsVDAC2 facilitates RSV accumulation in the vector. Yeast two-hybrid and GST pulldown assays demonstrated that LsVDAC2 interacted with RSV RNA-dependent RNA polymerase, RdRp. Furthermore, experiments in vivo and in vitro showed that LsVDAC2 induced the apoptotic response in RSV-infected insects and tissues. Silencing of LsVDAC2 via RNAi significantly reduced the expression of genes for apoptosis-related caspases 1a and 1c by 62% and 78%, respectively, in RSV-infected vectors. Whether LsVDAC2-induced RSV accumulation is related to RSV RdRp and LsVDAC2-induced cell apoptosis deserves further investigation.
Collapse
Affiliation(s)
- Lu Zhang
- College of Horticulture and Plant Protection, Yangzhou University, Yangzhou 225009, China
| | - Linying Li
- College of Horticulture and Plant Protection, Yangzhou University, Yangzhou 225009, China
| | - Lijun Huang
- College of Horticulture and Plant Protection, Yangzhou University, Yangzhou 225009, China
| | - Xinyi Li
- College of Horticulture and Plant Protection, Yangzhou University, Yangzhou 225009, China
| | - Chengzhu Xu
- College of Horticulture and Plant Protection, Yangzhou University, Yangzhou 225009, China
| | - Wenxing Hu
- College of Horticulture and Plant Protection, Yangzhou University, Yangzhou 225009, China
| | - Yixuan Sun
- College of Horticulture and Plant Protection, Yangzhou University, Yangzhou 225009, China
| | - Fang Liu
- College of Horticulture and Plant Protection, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-Innovation Center for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou 225009, China.
| | - Yao Li
- College of Horticulture and Plant Protection, Yangzhou University, Yangzhou 225009, China; Jiangsu Co-Innovation Center for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
35
|
Alshiraihi I, Kato TA. Apoptosis Detection Assays. Methods Mol Biol 2023; 2519:53-63. [PMID: 36066709 DOI: 10.1007/978-1-0716-2433-3_6] [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] [Indexed: 06/15/2023]
Abstract
Many apoptosis assays are available since there are many proteins regulated at multiple points and involved in apoptosis signaling cascade. To detect apoptosis accurately, two or more assays should be used since there are many overlapped features between apoptosis and necrosis. There are six major groups of available assays to detect apoptosis: membrane alteration, mitochondrial assays, cytomorphological alterations, DNA fragmentation, detection of caspase, cleaved substrate, inhibitors and regulators, and detection of apoptosis in whole mounts. Among those assay, early apoptosis could be detected through annexin V, which is based on the loss of the cellular membrane integrity. Also, there are many assays that can detect midphase of apoptosis using caspase activation and molecular processing including PARP degradation. Late phase of apoptosis could be detected with DNA fragmentation assays. Combinations of these assays allow us to identify the mechanisms of apoptosis induction after specific stimulus. This chapter will introduce three apoptosis detection assays including annexin assay, DNA/chromatin condensation assays, and TUNEL assay.
Collapse
Affiliation(s)
- Ilham Alshiraihi
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Takamitsu A Kato
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
36
|
Nüssing S, Sutton VR, Trapani JA, Parish IA. Beyond target cell death - Granzyme serine proteases in health and disease. Mol Aspects Med 2022; 88:101152. [PMID: 36368281 DOI: 10.1016/j.mam.2022.101152] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 10/06/2022] [Accepted: 10/22/2022] [Indexed: 11/09/2022]
Abstract
Granzymes are a family of small (∼32 kDa) serine proteases with a range of substrate specificities that are stored in, and released from, the cytoplasmic secretory vesicles ('granules') of cytotoxic T lymphocytes and natural killer cells. Granzymes are not digestive proteases but finely tuned processing enzymes that target their substrates in specific ways to activate various signalling pathways, or to inactivate viral proteins and other targets. Great emphasis has been placed on studying the pro-apoptotic functions of granzymes, which largely depend on their synergy with the pore-forming protein perforin, on which they rely for penetration into the target cell cytosol to access their substrates. While a critical role for granzyme B in target cell apoptosis is undisputed, both it and the remaining granzymes also influence a variety of other biological processes (including important immunoregulatory functions), which are discussed in this review. This includes the targeting of many extracellular as well as intracellular substrates, and can also lead to deleterious outcomes for the host if granzyme expression or function are dysregulated or abrogated. A final important consideration is that granzyme repertoire, biochemistry and function vary considerably across species, probably resulting from the pressures applied by viruses and other pathogens across evolutionary time. This has implications for the interpretation of granzyme function in preclinical models of disease.
Collapse
Affiliation(s)
- Simone Nüssing
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Vivien R Sutton
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Joseph A Trapani
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, 3052, Australia.
| | - Ian A Parish
- Peter MacCallum Cancer Centre, Melbourne, Victoria, 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, 3052, Australia; John Curtin School of Medical Research, ANU, ACT, Australia.
| |
Collapse
|
37
|
Weigelin B, Friedl P. T cell-mediated additive cytotoxicity - death by multiple bullets. Trends Cancer 2022; 8:980-987. [PMID: 35965200 DOI: 10.1016/j.trecan.2022.07.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 12/24/2022]
Abstract
Immune effector cells, including cytotoxic T cells (CTLs), induce apoptosis and eliminate target cells by direct cell-cell contacts. In vivo, CTLs fail to efficiently kill solid tumor cells by individual contacts but rely upon multihit interactions by many CTLs (swarming). Recent evidence has indicated that multihit interactions by CTLs induce a series of sublethal damage events in target cells, including perforin-mediated membrane damage, induction of reactive oxygen species (ROS), nuclear envelope rupture, and DNA damage. Individual damage can be repaired, but when induced in rapid sequence, sublethal damage can accumulate and induce target cell death. Here, we summarize the sublethal damage and additive cytotoxicity concepts for CTL-induced and other cell stresses and discuss the implications for improving immunotherapy and multitargeted anticancer therapies.
Collapse
Affiliation(s)
- Bettina Weigelin
- Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany.
| | - Peter Friedl
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, The Netherlands; David H. Koch Center for Applied Research of Genitourinary Cancers, Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Cancer Genomics Centre Netherlands (CGC.nl), Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
38
|
de Souza-Silva TG, Gollob KJ, Dutra WO. T-cell receptor variable region usage in Chagas disease: A systematic review of experimental and human studies. PLoS Negl Trop Dis 2022; 16:e0010546. [PMID: 36107855 PMCID: PMC9477334 DOI: 10.1371/journal.pntd.0010546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
T cells recognize their ligand, the peptide major histocompatibility complex (MHC), via the T-cell receptor (TCR), which is composed of covalently linked α and β or γ and δ chains. This recognition is critical for T-cell ontogeny and controls the selection, activation, and function of T lymphocytes. Specific TCR αβ variable regions have been associated with immunopathogenesis of Chagas disease. Here, we present a systematic review that compiles experimental in vivo and human data regarding the preferential expression of variable alpha (Vα) and variable beta (Vβ) chain regions in Trypanosoma cruzi infection. The original studies indexed in PubMed/Medline, Scopus, and Web of Science databases were screened according to the PRISMA strategy. The analysis showed that expression of TCR Vα subfamilies were evaluated in one human study, and, unlike TCR Vβ, TCR Vα presented a more restricted usage. Despite the great variability in the usage of TCR Vβ regions in human Chagas disease, a down-regulation of TCR Vβ5 expression by T cells from patients in the acute phase of the disease was shown. Opposingly, this TCR region was found overly expressed in CD4+ T cells from chronic Chagas patients. It was also demonstrated that murine Vβ9+ T cells derived from nonlymphoid organs of T. cruzi-infected animals had a modulatory profile, while splenic Vβ9+ T cells produced inflammatory cytokines, indicating that although they display the same TCR Vβ region usage, these cells are functionally distinct. Despite the limitations of few papers and year of publication of the studies, compiling the data derived from them reveals that further investigation of TCR usage will point to their potential role in protective or pathogenic responses, as biomarkers of disease progression, and in the search for dominant peptides potentially useful for the development of vaccines or therapies. Chagas disease is a neglected tropical disease, caused by infection with Trypanosoma cruzi. Differential expression of certain T-cell receptor (TCR) variable regions has been associated with the immunopathogenesis of Chagas disease. Here, we present a systematic review that compiled experimental in vivo and human data regarding the preferential expression of TCR alpha and beta chain variable regions in Chagas disease. The original studies indexed in the PubMed/Medline, Scopus, and Web of Science databases were screened according to the PRISMA strategy. Despite the great variability in the use of TCR Vβ in T. cruzi infection, the outcomes indicate that there is a down-regulation of TCR Vβ5 expression in T cells from patients in the acute phase of Chagas disease. However, this region is preferentially expressed by CD4+ T cells from chronic Chagas patients. Additionally, it has been demonstrated that murine Vβ9+ T cells derived from nonlymphoid organs displayed a modulatory profile, while splenic Vβ9+ T cells produced inflammatory cytokines, indicating that although they express the same TCR Vβ region, these cells are functionally distinct. Information on TCR expression, specificity and function have critical impact on vaccine design.
Collapse
Affiliation(s)
- Thaiany Goulart de Souza-Silva
- Institute of Biological Sciences, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Kenneth J. Gollob
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Belo Horizonte, Minas Gerais, Brazil
| | - Walderez O. Dutra
- Institute of Biological Sciences, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais, Belo Horizonte, Minas Gerais, Brazil
- * E-mail:
| |
Collapse
|
39
|
Kaymak E, Öztürk E, Akİn AT, Karabulut D, Yakan B. Thymoquinone alleviates doxorubicin induced acute kidney injury by decreasing endoplasmic reticulum stress, inflammation and apoptosis. Biotech Histochem 2022; 97:622-634. [PMID: 35989671 DOI: 10.1080/10520295.2022.2111465] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Doxorubicin (DOX) is used as an anticancer drug despite its many side effects. Thymoquinone (THQ) is a plant-derived substance that exhibits antioxidant and anti-inflammatory properties. We investigated the protective effects of THQ on DOX induced nephrotoxicity in rats. Rats were divided into five groups of eight: group 1, untreated control; group 2, olive oil group given olive oil intraperitoneally (i.p.) for 14 days; group 3, THQ group given 10 mg/kg THQ i.p. for 14 days; group 4, DOX group given a single dose of 15 mg/kg DOX i.p. on day 7 of experiment; group 5, DOX + THQ given 10 mg/kg THQ i.p. for 14 days and 15 mg/kg DOX i.p. on day 7. Kidney tissues were evaluated for histopathology. Caspase-3, IL-17, GRP78 and TNF-α immunostaining was used to determine the expression levels of these proteins among the groups. The TUNEL method was used to determine the apoptotic index. Total antioxidant status (TAS), total oxidant status (TOS), and TNF-α and TGF-β1 levels in kidney tissue were measured using ELISA assay. Histopathologic damage, caspase-3, IL-17, GRP78 and TNF-α immunoreactivity, TUNEL positive cells, TOS, TNF-α and TGF-β1 levels were increased in group 4 compared to group 1. The TAS of group 4 decreased compared to group 1. We found decreased caspase-3, IL-17, GRP78 and TNF-α expressions and TUNEL positive cells in group 5 compared to group 4. In rats given DOX, THQ reduced kidney damage by suppressing endoplasmic reticulum stress, inflammation and apoptosis pathways.
Collapse
|
40
|
Xu X, Zhang T, Xia X, Yin Y, Yang S, Ai D, Qin H, Zhou M, Song J. Pyroptosisin periodontitis: From the intricate interaction with apoptosis, NETosis, and necroptosis to the therapeutic prospects. Front Cell Infect Microbiol 2022; 12:953277. [PMID: 36093182 PMCID: PMC9450806 DOI: 10.3389/fcimb.2022.953277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/18/2022] [Indexed: 11/20/2022] Open
Abstract
Periodontitis is highly prevalent worldwide. It is characterized by periodontal attachment and alveolar bone destruction, which not only leads to tooth loss but also results in the exacerbation of systematic diseases. As such, periodontitis has a significant negative impact on the daily lives of patients. Detailed exploration of the molecular mechanisms underlying the physiopathology of periodontitis may contribute to the development of new therapeutic strategies for periodontitis and the associated systematic diseases. Pyroptosis, as one of the inflammatory programmed cell death pathways, is implicated in the pathogenesis of periodontitis. Progress in the field of pyroptosis has greatly enhanced our understanding of its role in inflammatory diseases. This review first summarizes the mechanisms underlying the activation of pyroptosis in periodontitis and the pathological role of pyroptosis in the progression of periodontitis. Then, the crosstalk between pyroptosis with apoptosis, necroptosis, and NETosis in periodontitis is discussed. Moreover, pyroptosis, as a novel link that connects periodontitis with systemic disease, is also reviewed. Finally, the current challenges associated with pyroptosis as a potential therapeutic target for periodontitis are highlighted.
Collapse
Affiliation(s)
- Xiaohui Xu
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Tingwei Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Xuyun Xia
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yuanyuan Yin
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Sihan Yang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Dongqing Ai
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Han Qin
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Mengjiao Zhou
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
- *Correspondence: Jinlin Song,
| |
Collapse
|
41
|
Yin M, Wen W, Wang H, Zhao Q, Zhu H, Chen H, Li X, Qian P. Porcine Sapelovirus 3C pro Inhibits the Production of Type I Interferon. Front Cell Infect Microbiol 2022; 12:852473. [PMID: 35782136 PMCID: PMC9240219 DOI: 10.3389/fcimb.2022.852473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/28/2022] [Indexed: 11/29/2022] Open
Abstract
Porcine sapelovirus (PSV) is the causative pathogen of reproductive obstacles, acute diarrhea, respiratory distress, or severe polioencephalomyelitis in swine. Nevertheless, the pathogenicity and pathogenic mechanism of PSV infection are not fully understood, which hinders disease prevention and control. In this study, we found that PSV was sensitive to type I interferon (IFN-β). However, PSV could not activate the IFN-β promoter and induce IFN-β mRNA expression, indicating that PSV has evolved an effective mechanism to block IFN-β production. Further study showed that PSV inhibited the production of IFN-β by cleaving mitochondrial antiviral signaling (MAVS) and degrading melanoma differentiation-associated gene 5 (MDA5) and TANK-binding kinase 1 (TBK1) through viral 3Cpro. In addition, our study demonstrated that PSV 3Cpro degrades MDA5 and TBK1 through its protease activity and cleaves MAVS through the caspase pathway. Collectively, our results revealed that PSV inhibits the production of type I interferon to escape host antiviral immunity through cleaving and degrading the adaptor molecules.
Collapse
Affiliation(s)
- Mengge Yin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Wei Wen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Haoyuan Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Qiongqiong Zhao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Hechao Zhu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic ProductsAgriculture of the People’s Republic of China, Ministry of Agriculture of the People’s Republic of China, Wuhan, China
- International Research Center for Animal DiseaseTechnology of the People’s Republic of China, Ministry of Science and Technology of the People’s Republic of China, Wuhan, China
| | - Xiangmin Li
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic ProductsAgriculture of the People’s Republic of China, Ministry of Agriculture of the People’s Republic of China, Wuhan, China
- International Research Center for Animal DiseaseTechnology of the People’s Republic of China, Ministry of Science and Technology of the People’s Republic of China, Wuhan, China
| | - Ping Qian
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic ProductsAgriculture of the People’s Republic of China, Ministry of Agriculture of the People’s Republic of China, Wuhan, China
- International Research Center for Animal DiseaseTechnology of the People’s Republic of China, Ministry of Science and Technology of the People’s Republic of China, Wuhan, China
| |
Collapse
|
42
|
McKenzie B, Khazen R, Valitutti S. Greek Fire, Poison Arrows, and Scorpion Bombs: How Tumor Cells Defend Against the Siege Weapons of Cytotoxic T Lymphocytes. Front Immunol 2022; 13:894306. [PMID: 35592329 PMCID: PMC9110820 DOI: 10.3389/fimmu.2022.894306] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/07/2022] [Indexed: 01/05/2023] Open
Abstract
CD8+ cytotoxic T lymphocytes (CTLs) are the main cellular effectors of the adaptive immune response against cancer cells, which in turn have evolved sophisticated cellular defense mechanisms to withstand CTL attack. Herein we provide a critical review of the pertinent literature on early and late attack/defense events taking place at the CTL/target cell lytic synapse. We examine the earliest steps of CTL-mediated cytotoxicity (“the poison arrows”) elicited within seconds of CTL/target cell encounter, which face commensurately rapid synaptic repair mechanisms on the tumor cell side, providing the first formidable barrier to CTL attack. We examine how breach of this first defensive barrier unleashes the inextinguishable “Greek fire” in the form of granzymes whose broad cytotoxic potential is linked to activation of cell death executioners, injury of vital organelles, and destruction of intracellular homeostasis. Herein tumor cells deploy slower but no less sophisticated defensive mechanisms in the form of enhanced autophagy, increased reparative capacity, and dysregulation of cell death pathways. We discuss how the newly discovered supra-molecular attack particles (SMAPs, the “scorpion bombs”), seek to overcome the robust defensive mechanisms that confer tumor cell resistance. Finally, we discuss the implications of the aforementioned attack/defense mechanisms on the induction of regulated cell death (RCD), and how different contemporary RCD modalities (including apoptosis, pyroptosis, and ferroptosis) may have profound implications for immunotherapy. Thus, we propose that understanding and targeting multiple steps of the attack/defense process will be instrumental to enhance the efficacy of CTL anti-tumor activity and meet the outstanding challenges in clinical immunotherapy.
Collapse
Affiliation(s)
- Brienne McKenzie
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Roxana Khazen
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France
| | - Salvatore Valitutti
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, Toulouse, France.,Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, Toulouse, France
| |
Collapse
|
43
|
Osacka J, Kiss A, Pirnik Z. Possible involvement of apoptosis in the antipsychotics side effects: A minireview. Clin Exp Pharmacol Physiol 2022; 49:836-847. [PMID: 35575958 DOI: 10.1111/1440-1681.13671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 01/04/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022]
Abstract
Antipsychotics are used in the treatment of schizophrenia and other psychiatric disorders. Generally they are divided into typical and atypical ones, according to the fact that atypical antipychotics induce less side effects and are more effective in terms of social and cognitive improvements. Their pharmacological effects are mediated via broad range of receptors that consequently influence different cellular signaling pathways. Antipsychotics produce udesirable side effects that range from relatively minor to life-threatening ones. In vitro and in vivo studies have pointed to neurotoxic effect exerted by some antipsychotics and have shown that apoptosis might play role in some side effects induced by antipsyschotics, including tardive dyskinesia, weight gain, agranulocytosis, osteoporosis, myocarditis, etc. Although cumulative data have suggested safety of atypical antipsychotics use during pregnancy some of them have been shown to induce apoptotic neurodegenerative and structural changes in fetal brains with long-lasting impact on cognitive impairment of offsprings. Typical antipsychotics seem to be more cytotoxic than atypical ones. Recently, epidemiological studies have shown lower incidence of cancer in schizophrenic patients what suggest ability of antipsychotics to suppress risk of cancer development. Some antipsychotics have been reported to inhibit cancer cell proliferation and induce their apoptosis. Thus, antipsychotics apoptotic effect may be used as a tool in the treatmnet of some types of cancer, especially in combinatorial therapies. In this minireview, we focused on pro- and anti-apototic or "Dr. Jekyll and Mr. Hyde" effects of antipsychotics, which can be involved in their side effects, as well as their promising therapeutical indications. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jana Osacka
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Experimental Endocrinology, Dubravska cesta 9, Bratislava, Slovakia
| | - Alexander Kiss
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Experimental Endocrinology, Dubravska cesta 9, Bratislava, Slovakia
| | - Zdenko Pirnik
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Experimental Endocrinology, Dubravska cesta 9, Bratislava, Slovakia.,Institute of Physiology, Faculty of Medicine Comenius University in Bratislava, Sasinkova 2, Bratislava, Slovakia.,Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam., 2Prague, Czech Republic
| |
Collapse
|
44
|
Billah MM, Khatiwada S, Morris MJ, Maloney CA. Effects of paternal overnutrition and interventions on future generations. Int J Obes (Lond) 2022; 46:901-917. [PMID: 35022547 PMCID: PMC9050512 DOI: 10.1038/s41366-021-01042-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/17/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023]
Abstract
In the last two decades, evidence from human and animal studies suggests that paternal obesity around the time of conception can have adverse effects on offspring health through developmental programming. This may make significant contributions to the current epidemic of obesity and related metabolic and reproductive complications like diabetes, cardiovascular disease, and subfertility/infertility. To date, changes in seminal fluid composition, sperm DNA methylation, histone composition, small non-coding RNAs, and sperm DNA damage have been proposed as potential underpinning mechanism to program offspring health. In this review, we discuss current human and rodent evidence on the impact of paternal obesity/overnutrition on offspring health, followed by the proposed mechanisms, with a focus on sperm DNA damage underpinning paternal programming. We also summarize the different intervention strategies implemented to minimize effects of paternal obesity. Upon critical review of literature, we find that obesity-induced altered sperm quality in father is linked with compromised offspring health. Paternal exercise intervention before conception has been shown to improve metabolic health. Further work to explore the mechanisms underlying benefits of paternal exercise on offspring are warranted. Conversion to healthy diets and micronutrient supplementation during pre-conception have shown some positive impacts towards minimizing the impact of paternal obesity on offspring. Pharmacological approaches e.g., metformin are also being applied. Thus, interventions in the obese father may ameliorate the potential detrimental impacts of paternal obesity on offspring.
Collapse
Affiliation(s)
| | - Saroj Khatiwada
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Margaret J Morris
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | | |
Collapse
|
45
|
Edaravone Attenuated Particulate Matter-Induced Lung Inflammation by Inhibiting ROS-NF-κB Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6908884. [PMID: 35502210 PMCID: PMC9056219 DOI: 10.1155/2022/6908884] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 03/05/2022] [Accepted: 03/30/2022] [Indexed: 01/31/2023]
Abstract
Background Particulate matter (PM) exposure is related to mitochondria dysfunction and airway inflammation. Antioxidant drug edaravone (EDA) is reported to improve the occurrence and development of oxidative stress-related diseases. At present, there is no data on whether EDA can alleviate lung inflammation caused by PM. Methods The anti-inflammatory effects of EDA were investigated in urban PM-induced human bronchial epithelial cells (HBECs) and C57/BL6J mouse models. In vitro, its effects on the production of intracellular reactive oxygen species (ROS), mitochondrial membrane potential (MMP), and inflammatory cytokines were assessed by DCFH-DA staining, JC-1 assay, and real-time PCR, respectively. In vivo, the oxidant stress in lung tissues was assessed by dihydroethidium (DHE) staining and malondialdehyde (MDA) activity, and inflammatory cytokines in bronchoalveolar lavage fluid (BALF) were assessed by ELISA, respectively. Furthermore, the potential signaling pathways were studied by siRNA transfection and western blot. Results PM increased the expression of inflammatory cytokines and protein, including IL-6, IL-1α, IL-1β, and COX-2, while these alternations were significantly alleviated following EDA treatment in a dose-dependent manner. EDA treatment also alleviated the inflammatory responses in lung tissues of PM-exposed mice. We further showed mitochondrial dysfunction in PM-exposed HBECs and mice, which were reversed by EDA treatment. Moreover, the phosphorylation of NF-κB p65 in PM-exposed HBECs and mice was weakened by EDA. Transfection with NF-κB p65 siRNA further inhibited PM-induced inflammation in HBECs. Conclusion We demonstrated that EDA treatment had a protective role in PM-induced lung inflammation through maintaining mitochondrial balance and regulating the ROS-NF-κB p65 signaling pathway. This provided a new therapeutic method for PM-induced lung inflammation in the future.
Collapse
|
46
|
Abstract
Apoptosis is an evolutionarily conserved sequential process of cell death to maintain a homeostatic balance between cell formation and cell death. It is a vital process for normal eukaryotic development as it contributes to the renewal of cells and tissues. Further, it plays a crucial role in the elimination of unnecessary cells through phagocytosis and prevents undesirable immune responses. Apoptosis is regulated by a complex signaling mechanism, which is driven by interactions among several protein families such as caspases, inhibitors of apoptosis proteins, B-cell lymphoma 2 (BCL-2) family proteins, and several other proteases such as perforins and granzyme. The signaling pathway consists of both pro-apoptotic and pro-survival members, which stabilize the selection of cellular survival or death. However, any aberration in this pathway can lead to abnormal cell proliferation, ultimately leading to the development of cancer, autoimmune disorders, etc. This review aims to elaborate on apoptotic signaling pathways and mechanisms, interacting members involved in signaling, and how apoptosis is associated with carcinogenesis, along with insights into targeting apoptosis for disease resolution.
Collapse
|
47
|
Tibbs E, Cao X. Emerging Canonical and Non-Canonical Roles of Granzyme B in Health and Disease. Cancers (Basel) 2022; 14:1436. [PMID: 35326588 PMCID: PMC8946077 DOI: 10.3390/cancers14061436] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 12/23/2022] Open
Abstract
The Granzyme (Gzm) family has classically been recognized as a cytotoxic tool utilized by cytotoxic T lymphocytes (CTL) and natural killer (NK) cells to illicit cell death to infected and cancerous cells. Their importance is established based on evidence showing that deficiencies in these cell death executors result in defective immune responses. Recent findings have shown the importance of Granzyme B (GzmB) in regulatory immune cells, which may contribute to tumor growth and immune evasion during cancer development. Other studies have shown that members of the Gzm family are important for biological processes such as extracellular matrix remodeling, angiogenesis and organized vascular degradation. With this growing body of evidence, it is becoming more important to understand the broader function of Gzm's rather than a specific executor of cell death, and we should be aware of the many alternative roles that Gzm's play in physiological and pathological conditions. Therefore, we review the classical as well as novel non-canonical functions of GzmB and discuss approaches to utilize these new findings to address current gaps in our understanding of the immune system and tissue development.
Collapse
Affiliation(s)
- Ellis Tibbs
- Department of Microbiology and Immunology, School of Medicine, University of Maryland Baltimore, Baltimore, MD 21201, USA;
| | - Xuefang Cao
- Department of Microbiology and Immunology, School of Medicine, University of Maryland Baltimore, Baltimore, MD 21201, USA;
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore, Baltimore, MD 21201, USA
| |
Collapse
|
48
|
Rasi V, Hameed OA, Matthey P, Bera S, Grandgenett DP, Salentinig S, Walch M, Hoft DF. Improved Purification of Human Granzyme A/B and Granulysin Using a Mammalian Expression System. Front Immunol 2022; 13:830290. [PMID: 35300343 PMCID: PMC8921980 DOI: 10.3389/fimmu.2022.830290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/08/2022] [Indexed: 01/14/2023] Open
Abstract
Cytotoxic lymphocytes release proteins contained within the cytoplasmic cytolytic granules after recognition of infected or tumor target cells. These cytotoxic granular proteins (namely granzymes, granulysin, and perforin) are key immunological mediators within human cellular immunity. The availability of highly purified cytotoxic proteins has been fundamental for understanding their function in immunity and mechanistic involvement in sepsis and autoimmunity. Methods for recovery of native cytotoxic proteins can be problematic leading to: 1) the co-purification of additional proteins, confounding interpretation of function, and 2) low yields of highly purified proteins. Recombinant protein expression of individual cytolytic components can overcome these challenges. The use of mammalian expression systems is preferred for optimal post-translational modifications and avoidance of endotoxin contamination. Some of these proteins have been proposed for host directed human therapies (e.g. - granzyme A), or treatment of systemic infections or tumors as in granulysin. We report here a novel expression system using HEK293T cells for cost-effective purification of high yields of human granzymes (granzyme A and granzyme B) and granulysin with enhanced biological activity than previous reports. The resulting proteins are free of native contaminants, fold correctly, and remain enzymatically active. Importantly, these improvements have also led to the first purification of biologically active recombinant human granulysin in high yields from a mammalian system. This method can be used as a template for purification of many other secreted cellular proteins and may lead to advances for human medicine.
Collapse
Affiliation(s)
- Valerio Rasi
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, MO, United States,Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Owais Abdul Hameed
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland,Department of Chemistry, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Patricia Matthey
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Sibes Bera
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Duane P. Grandgenett
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, MO, United States
| | - Stefan Salentinig
- Department of Chemistry, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Michael Walch
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland,*Correspondence: Daniel F. Hoft, ; Michael Walch,
| | - Daniel F. Hoft
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, MO, United States,Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, Saint Louis University School of Medicine, Saint Louis, MO, United States,*Correspondence: Daniel F. Hoft, ; Michael Walch,
| |
Collapse
|
49
|
Kim SY, Barnes MA, Sureshchandra S, Menicucci AR, Patel JJ, Messaoudi I, Nair MG. CX3CR1-Expressing Myeloid Cells Regulate Host-Helminth Interaction and Lung Inflammation. Adv Biol (Weinh) 2022; 6:e2101078. [PMID: 35119218 PMCID: PMC8934291 DOI: 10.1002/adbi.202101078] [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: 07/12/2021] [Revised: 12/31/2021] [Indexed: 11/06/2022]
Abstract
Many helminth life cycles, including hookworm, involve a mandatory lung phase, where myeloid and granulocyte subsets interact with the helminth and respond to infection-induced lung injury. To evaluate these innate subsets in Nippostrongylus brasiliensis infection, reporter mice for myeloid cells (CX3CR1GFP ) and granulocytes (PGRPdsRED ) are employed. Nippostrongylus infection induces lung infiltration of reporter cells, including CX3CR1+ myeloid cells and PGRP+ eosinophils. Strikingly, CX3CR1GFP/GFP mice, which are deficient in CX3CR1, are protected from Nippostrongylus infection with reduced weight loss, lung leukocyte infiltration, and worm burden compared to CX3CR1+/+ mice. This protective effect is specific for CX3CR1 as CCR2-deficient mice do not exhibit reduced worm burdens. Nippostrongylus co-culture with lung Ly6C+ monocytes or CD11c+ cells demonstrates that CX3CR1GFP/GFP monocytes secrete more pro-inflammatory cytokines and actively bind the parasites causing reduced motility. RNA sequencing of Ly6C+ or CD11c+ cells shows Nippostrongylus-induced gene expression changes, particularly in monocytes, associated with inflammation, chemotaxis, and extracellular matrix remodeling pathways. Analysis reveals cytotoxic and adhesion molecules as potential effectors against the parasite, such as Gzma and Gzmb, which are elevated in CX3CR1GFP/GFP monocytes. These studies validate a dual innate cell reporter for lung helminth infection and demonstrate that CX3CR1 impairs monocyte-helminth interaction.
Collapse
Affiliation(s)
| | | | | | - Andrea R. Menicucci
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, California 92697-3900, United States
| | - Jay J. Patel
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California 92521, United States
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, California 92697-3900, United States
| | | |
Collapse
|
50
|
Ning Z, Lan J, Jiang X, Zhong G, Zhang H, Wan F, Wu S, Tang Z, Bilal RM, Hu L, Huang R. Arsenic trioxide-induced autophagy affected the antioxidant capacity and apoptosis rate of chicken hepatocytes. Chem Biol Interact 2022; 354:109821. [PMID: 35051378 DOI: 10.1016/j.cbi.2022.109821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 12/20/2021] [Accepted: 01/13/2022] [Indexed: 12/19/2022]
Abstract
Arsenic has recently received widespread attention due to its high toxicological effects on multiple animals; however, the mechanism underlying this toxicity is unclear. We investigated the damaging effects of arsenic trioxide (ATO) on hepatocytes and the effects of regulating autophagy on the hepatocyte damage induced by ATO exposure. First, we investigated the effects of ATO exposure (0, 0.6, 1.2, 2.4, and 4.8 μM) on the biochemical function and autophagy of chicken hepatocytes. The findings showed that as the concentration of ATO increased, the lactate dehydrogenase (LDH) concentration increased, more autophagosomes were observed via transmission electron microscopy (TEM), and the gene and protein expression levels of P62, LC3Ⅱ, and Beclin1 increased. Adding N-acetyl-l-cystine (NAC, 1 mM) attenuated autophagy and the hepatocyte damage induced by ATO. Then, we used rapamycin (Rapa) and 3-methylpurine (3-MA) to regulate the autophagy induced by exposure to 4.8 μM ATO and observed changes in the antioxidant capacity and apoptosis rate of chicken hepatocytes. Induction of autophagy reduced ATO-induced hepatocyte apoptosis but caused no significant effect on oxidative stress in chicken hepatocytes. Inhibition of autophagy exacerbated ATO-induced hepatocyte oxidative stress and apoptosis. These findings demonstrate that autophagy plays an important role in ATO-induced cell damage.
Collapse
Affiliation(s)
- Zhijun Ning
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Juan Lan
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Xuanxuan Jiang
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Gaolong Zhong
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Hui Zhang
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Fang Wan
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Shaofeng Wu
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Zhaoxin Tang
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Rana Muhammad Bilal
- University College of Veterinary & Animal Sciences, Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan.
| | - Lianmei Hu
- College of Veterinary Medicine, Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, 510642, China.
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|