1
|
Han L, Meng L, Liu J, Xie Y, Kang R, Klionsky DJ, Tang D, Jia Y, Dai E. Macroautophagy/autophagy promotes resistance to KRAS G12D-targeted therapy through glutathione synthesis. Cancer Lett 2024; 604:217258. [PMID: 39276914 DOI: 10.1016/j.canlet.2024.217258] [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: 07/30/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
KRASG12D mutation-driven pancreatic ductal adenocarcinoma (PDAC) represents a major challenge in medicine due to late diagnosis and treatment resistance. Here, we report that macroautophagy (hereafter autophagy), a cellular degradation and recycling process, contributes to acquired resistance against novel KRASG12D-targeted therapy. The KRASG12D protein inhibitor MRTX1133 induces autophagy in KRASG12D-mutated PDAC cells by blocking MTOR activity, and increased autophagic flux prevents apoptosis. Mechanistically, autophagy facilitates the generation of glutamic acid, cysteine, and glycine for glutathione synthesis. Increased glutathione levels reduce reactive oxygen species production, which impedes CYCS translocation from mitochondria to the cytosol, ultimately preventing the formation of the APAF1 apoptosome. Consequently, genetic interventions (utilizing ATG5 or BECN1 knockout) or pharmacological inhibition of autophagy (with chloroquine, bafilomycin A1, or spautin-1) enhance the anticancer activity of MRTX1133 in vitro and in various animal models (subcutaneous, patient-derived xenograft, and orthotopic). Moreover, the release of histones by apoptotic cells triggers an adaptive immune response when combining an autophagy inhibitor with MRTX1133 in immunocompetent mice. These findings establish a new strategy to overcome KRASG12D-targeted therapy resistance by inhibiting autophagy-dependent glutathione synthesis.
Collapse
Affiliation(s)
- Leng Han
- 2nd Ward of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China
| | - Lingjun Meng
- 2nd Ward of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China
| | - Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Yangchun Xie
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Rui Kang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Daolin Tang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Yuanyuan Jia
- 2nd Ward of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China.
| | - Enyong Dai
- 2nd Ward of Oncology and Hematology Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China.
| |
Collapse
|
2
|
Favero F, Re A, Dason MS, Gravina T, Gagliardi M, Mellai M, Corazzari M, Corà D. Characterization of gut microbiota dynamics in an Alzheimer's disease mouse model through clade-specific marker-based analysis of shotgun metagenomic data. Biol Direct 2024; 19:100. [PMID: 39478626 PMCID: PMC11524029 DOI: 10.1186/s13062-024-00541-7] [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: 08/28/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder significantly impairing cognitive faculties, memory, and physical abilities. To characterize the modulation of the gut microbiota in an in vivo AD model, we performed shotgun metagenomics sequencing on 3xTgAD mice at key time points (i.e., 2, 6, and 12 months) of AD progression. Fecal samples from both 3xTgAD and wild-type mice were collected, DNA extracted, and sequenced. Quantitative taxon abundance assessment using MetaPhlAn 4 ensured precise microbial community representation. The analysis focused on species-level genome bins (SGBs) including both known and unknown SGBs (kSGBs and uSGBs, respectively) and also comprised higher taxonomic categories such as family-level genome bins (FGBs), class-level genome bins (CGBs), and order-level genome bins (OGBs). Our bioinformatic results pinpointed the presence of extensive gut microbial diversity in AD mice and showed that the largest proportion of AD- and aging-associated microbiome changes in 3xTgAD mice concern SGBs that belong to the Bacteroidota and Firmicutes phyla, along with a large set of uncharacterized SGBs. Our findings emphasize the need for further advanced bioinformatic studies for accurate classification and functional analysis of these elusive microbial species in relation to their potential bridging role in the gut-brain axis and AD pathogenesis.
Collapse
Affiliation(s)
- Francesco Favero
- Department of Translational Medicine (DIMET), University of Piemonte Orientale, Via Solaroli 17, I-28100, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), C.so Trieste, 15/A, I-28100, Novara, Italy
| | - Angela Re
- Department of Applied Science and Technology (DISAT) - Politecnico di Torino, C.so Duca degli Abruzzi, 24, I-10129, Torino, Italy
| | - Mohammed Salim Dason
- Department of Applied Science and Technology (DISAT) - Politecnico di Torino, C.so Duca degli Abruzzi, 24, I-10129, Torino, Italy
| | - Teresa Gravina
- Department of Translational Medicine (DIMET), University of Piemonte Orientale, Via Solaroli 17, I-28100, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), C.so Trieste, 15/A, I-28100, Novara, Italy
| | - Mara Gagliardi
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), C.so Trieste, 15/A, I-28100, Novara, Italy
- Department of Health Sciences (DISS), University of Piemonte Orientale, Via Solaroli 17, I- 28100, Novara, Italy
| | - Marta Mellai
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), C.so Trieste, 15/A, I-28100, Novara, Italy
- Department of Health Sciences (DISS), University of Piemonte Orientale, Via Solaroli 17, I- 28100, Novara, Italy
| | - Marco Corazzari
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), C.so Trieste, 15/A, I-28100, Novara, Italy.
- Department of Health Sciences (DISS), University of Piemonte Orientale, Via Solaroli 17, I- 28100, Novara, Italy.
| | - Davide Corà
- Department of Translational Medicine (DIMET), University of Piemonte Orientale, Via Solaroli 17, I-28100, Novara, Italy.
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), C.so Trieste, 15/A, I-28100, Novara, Italy.
| |
Collapse
|
3
|
Leblond AL, Helmchen B, Ankavay M, Lenggenhager D, Jetzer J, Helmchen F, Yurtsever H, Parrotta R, Healy ME, Pöschel A, Markkanen E, Semmo N, Ferrié M, Cocquerel L, Seeger H, Hopfer H, Müllhaupt B, Gouttenoire J, Moradpour D, Gaspert A, Weber A. HEV ORF2 protein-antibody complex deposits are associated with glomerulonephritis in hepatitis E with reduced immune status. Nat Commun 2024; 15:8849. [PMID: 39397005 PMCID: PMC11471813 DOI: 10.1038/s41467-024-53072-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 09/27/2024] [Indexed: 10/15/2024] Open
Abstract
Hepatitis E virus (HEV) infection, one of the most common forms of hepatitis worldwide, is often associated with extrahepatic, particularly renal, manifestations. However, the underlying mechanisms are incompletely understood. Here, we report the development of a de novo immune complex-mediated glomerulonephritis (GN) in a kidney transplant recipient with chronic hepatitis E. Applying immunostaining, electron microscopy, and mass spectrometry after laser-capture microdissection, we show that GN develops in parallel with increasing glomerular deposition of a non-infectious, genome-free and non-glycosylated HEV open reading frame 2 (ORF2) capsid protein. No productive HEV infection of kidney cells is detected. Patients with acute hepatitis E display similar but less pronounced deposits. Our results establish a link between the production of HEV ORF2 protein and the development of hepatitis E-associated GN in the immunocompromised state. The formation of glomerular IgG-HEV ORF2 immune complexes discovered here provides a potential mechanistic explanation of how the hepatotropic HEV can cause variable renal manifestations. These findings directly provide a tool for etiology-based diagnosis of hepatitis E-associated GN as a distinct entity and suggest therapeutic implications.
Collapse
Affiliation(s)
- Anne-Laure Leblond
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Birgit Helmchen
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Maliki Ankavay
- Division of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Daniela Lenggenhager
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Jasna Jetzer
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Fritjof Helmchen
- Brain Research Institute, University of Zurich, Zurich, Switzerland
| | | | - Rossella Parrotta
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Marc E Healy
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Amiskwia Pöschel
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich - Vetsuisse Faculty, Zürich, Switzerland
| | - Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich - Vetsuisse Faculty, Zürich, Switzerland
| | - Nasser Semmo
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Martin Ferrié
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Laurence Cocquerel
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Harald Seeger
- Clinic of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Helmut Hopfer
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Beat Müllhaupt
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Jérôme Gouttenoire
- Division of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Darius Moradpour
- Division of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ariana Gaspert
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University of Zurich (UZH) and University Hospital Zurich (USZ), Zurich, Switzerland.
- Institute of Molecular Cancer Research (IMCR), University of Zurich (UZH), Zurich, Switzerland.
| |
Collapse
|
4
|
Galassi C, Chan TA, Vitale I, Galluzzi L. The hallmarks of cancer immune evasion. Cancer Cell 2024:S1535-6108(24)00358-1. [PMID: 39393356 DOI: 10.1016/j.ccell.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/27/2024] [Accepted: 09/16/2024] [Indexed: 10/13/2024]
Abstract
According to the widely accepted "three Es" model, the host immune system eliminates malignant cell precursors and contains microscopic neoplasms in a dynamic equilibrium, preventing cancer outgrowth until neoplastic cells acquire genetic or epigenetic alterations that enable immune escape. This immunoevasive phenotype originates from various mechanisms that can be classified under a novel "three Cs" conceptual framework: (1) camouflage, which hides cancer cells from immune recognition, (2) coercion, which directly or indirectly interferes with immune effector cells, and (3) cytoprotection, which shields malignant cells from immune cytotoxicity. Blocking the ability of neoplastic cells to evade the host immune system is crucial for increasing the efficacy of modern immunotherapy and conventional therapeutic strategies that ultimately activate anticancer immunosurveillance. Here, we review key hallmarks of cancer immune evasion under the "three Cs" framework and discuss promising strategies targeting such immunoevasive mechanisms.
Collapse
Affiliation(s)
- Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Timothy A Chan
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA; Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, OH, USA; National Center for Regenerative Medicine, Cleveland, OH, USA; Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Ilio Vitale
- Italian Institute for Genomic Medicine, c/o IRCSS Candiolo, Torino, Italy; Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA; Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Angelico R, Siragusa L, Blasi F, Bellato V, Mineccia M, Lolli E, Monteleone G, Sica GS. Colorectal cancer in ulcerative colitis after liver transplantation for primary sclerosing cholangitis: a systematic review and pooled analysis of oncological outcomes. Discov Oncol 2024; 15:529. [PMID: 39378005 PMCID: PMC11461386 DOI: 10.1007/s12672-024-01304-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 09/03/2024] [Indexed: 10/11/2024] Open
Abstract
INTRODUCTION Patients with ulcerative colitis (UC) receiving liver transplantation (LT) due to primary sclerosing cholangitis (PSC) have higher risk of developing colorectal cancers (CRC). Aim of this systematic review was to define the patients' features, immunosuppressive management, and oncological outcomes of LT recipients with UC-PSC developing CRC. METHODS Searches were conducted in PubMed (MEDLINE), Cochrane Library, Web of Science for all English articles published until September 2023. Inclusion criteria were original articles including patients specifying outcomes of interest. Primary endpoints comprised incidence of CRC, disease free survival (DFS), overall survival (OS) and cancer recurrence. Secondary endpoints were patient's and tumor characteristics, graft function, immunosuppressive management and PSC recurrence. PROSPERO CRD42022369190. RESULTS Fifteen studies included, 88 patients were identified. Patients (mean age: 50 years) had a long history of UC (20 years), mainly with active colitis (79%), and developed tumor within the first 3 years from LT, while receiving a double or triple immunosuppressive therapy. Cumulative incidence of tumor was 5.5%. At one, two and three years, DFS was 92%, 82% and 75%, while OS was 87%, 81% and 79% respectively. Disease progression rate was 15%. After CRC surgery, 94% of patients maintained a good graft functionality, with no reported cases of PSC recurrence. CONCLUSIONS After LT, patients with PSC and UC have an increased risk of CRC, especially in presence of long history of UC and active colitis. Surgical resection guarantees satisfactory mid-term oncological outcomes, but samples are limited, and long-term data are lacking. National and international registry are auspicial to evaluate long-term oncological outcomes and to optimize clinical management.
Collapse
Affiliation(s)
- Roberta Angelico
- HPB and Transplant Unit, Department of Surgical Sciences, University of Rome "Tor Vergata", Rome, Italy
| | - Leandro Siragusa
- Division of Colon and Rectal Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.
| | - Francesca Blasi
- Minimally Invasive and Digestive Surgery Unit, Department of Surgical Sciences, University of Rome "Tor Vergata", Rome, Italy
| | - Vittoria Bellato
- Minimally Invasive and Digestive Surgery Unit, Department of Surgical Sciences, University of Rome "Tor Vergata", Rome, Italy
| | | | - Elisabetta Lolli
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Giuseppe S Sica
- Minimally Invasive and Digestive Surgery Unit, Department of Surgical Sciences, University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
6
|
Sandoval TA, Salvagno C, Chae CS, Awasthi D, Giovanelli P, Falco MM, Hwang SM, Teran-Cabanillas E, Suominen L, Yamazaki T, Kuo HH, Moyer JE, Martin ML, Manohar J, Kim K, Sierra MA, Ramos Y, Tan C, Emmanuelli A, Song M, Morales DK, Zamarin D, Frey MK, Cantillo E, Chapman-Davis E, Holcomb K, Mason CE, Galluzzi L, Zhou ZN, Vähärautio A, Cloonan SM, Cubillos-Ruiz JR. Iron Chelation Therapy Elicits Innate Immune Control of Metastatic Ovarian Cancer. Cancer Discov 2024; 14:1901-1921. [PMID: 39073085 PMCID: PMC11452292 DOI: 10.1158/2159-8290.cd-23-1451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024]
Abstract
Iron accumulation in tumors contributes to disease progression and chemoresistance. Although targeting this process can influence various hallmarks of cancer, the immunomodulatory effects of iron chelation in the tumor microenvironment are unknown. Here, we report that treatment with deferiprone, an FDA-approved iron chelator, unleashes innate immune responses that restrain ovarian cancer. Deferiprone reprogrammed ovarian cancer cells toward an immunostimulatory state characterized by the production of type-I IFN and overexpression of molecules that activate NK cells. Mechanistically, these effects were driven by innate sensing of mitochondrial DNA in the cytosol and concomitant activation of nuclear DNA damage responses triggered upon iron chelation. Deferiprone synergized with chemotherapy and prolonged the survival of mice with ovarian cancer by bolstering type-I IFN responses that drove NK cell-dependent control of metastatic disease. Hence, iron chelation may represent an alternative immunotherapeutic strategy for malignancies that are refractory to current T-cell-centric modalities. Significance: This study uncovers that targeting dysregulated iron accumulation in ovarian tumors represents a major therapeutic opportunity. Iron chelation therapy using an FDA-approved agent causes immunogenic stress responses in ovarian cancer cells that delay metastatic disease progression and enhance the effects of first-line chemotherapy. See related commentary by Bell and Zou, p. 1771.
Collapse
Affiliation(s)
- Tito A. Sandoval
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Camilla Salvagno
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Chang-Suk Chae
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Deepika Awasthi
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Paolo Giovanelli
- Weill Cornell Graduate School of Medical Sciences. New York, NY 10065. USA
| | - Matias Marin Falco
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sung-Min Hwang
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Eli Teran-Cabanillas
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Lasse Suominen
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medicine. New York, NY 10065, USA
| | - Hui-Hsuan Kuo
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065. USA
| | - Jenna E. Moyer
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065. USA
| | - M Laura Martin
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065. USA
| | - Jyothi Manohar
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065. USA
| | - Kihwan Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine. New York, NY 10065, USA
| | - Maria A. Sierra
- Weill Cornell Graduate School of Medical Sciences. New York, NY 10065. USA
| | - Yusibeska Ramos
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
| | - Chen Tan
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Alexander Emmanuelli
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
- Weill Cornell Graduate School of Medical Sciences. New York, NY 10065. USA
| | - Minkyung Song
- Departments of Integrative Biotechnology and of Biopharmaceutical Convergence, Sungkyunkwan University. Suwon, Gyeonggi-do, Korea
| | - Diana K. Morales
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
| | - Dmitriy Zamarin
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Melissa K. Frey
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Evelyn Cantillo
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Eloise Chapman-Davis
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Kevin Holcomb
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Christopher E. Mason
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine. New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine. New York, USA
- The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine. New York, NY, USA
| | - Lorenzo Galluzzi
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
- Department of Radiation Oncology, Weill Cornell Medicine. New York, NY 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065. USA
| | - Zhen Ni Zhou
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
| | - Anna Vähärautio
- Research Program in Systems Oncology, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Foundation for the Finnish Cancer Institute, Helsinki, Finland
| | - Suzanne M. Cloonan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine. New York, NY 10065, USA
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College. Dublin, Ireland
| | - Juan R. Cubillos-Ruiz
- Department of Obstetrics and Gynecology, Weill Cornell Medicine. New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine. New York, NY 10065, USA
- Weill Cornell Graduate School of Medical Sciences. New York, NY 10065. USA
| |
Collapse
|
7
|
Xu K, Saaoud F, Shao Y, Lu Y, Yang Q, Jiang X, Wang H, Yang X. A new paradigm in intracellular immunology: Mitochondria emerging as leading immune organelles. Redox Biol 2024; 76:103331. [PMID: 39216270 PMCID: PMC11402145 DOI: 10.1016/j.redox.2024.103331] [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: 07/19/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Mitochondria, traditionally recognized as cellular 'powerhouses' due to their pivotal role in energy production, have emerged as multifunctional organelles at the intersection of bioenergetics, metabolic signaling, and immunity. However, the understanding of their exact contributions to immunity and inflammation is still developing. This review first introduces the innovative concept of intracellular immunity, emphasizing how mitochondria serve as critical immune signaling hubs. They are instrumental in recognizing and responding to pathogen and danger signals, and in modulating immune responses. We also propose mitochondria as the leading immune organelles, drawing parallels with the broader immune system in their functions of antigen presentation, immune regulation, and immune response. Our comprehensive review explores mitochondrial immune signaling pathways, their therapeutic potential in managing inflammation and chronic diseases, and discusses cutting-edge methodologies for mitochondrial research. Targeting a broad readership of both experts in mitochondrial functions and newcomers to the field, this review sets forth new directions that could transform our understanding of intracellular immunity and the integrated immune functions of intracellular organelles.
Collapse
Affiliation(s)
- Keman Xu
- Lemole Center for Integrated Lymphatics and Vascular Research, USA
| | - Fatma Saaoud
- Lemole Center for Integrated Lymphatics and Vascular Research, USA
| | - Ying Shao
- Lemole Center for Integrated Lymphatics and Vascular Research, USA
| | - Yifan Lu
- Lemole Center for Integrated Lymphatics and Vascular Research, USA
| | | | - Xiaohua Jiang
- Lemole Center for Integrated Lymphatics and Vascular Research, USA; Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Hong Wang
- Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA
| | - Xiaofeng Yang
- Lemole Center for Integrated Lymphatics and Vascular Research, USA; Metabolic Disease Research and Thrombosis Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
8
|
Chen C, Wang J, Zhang S, Zhu X, Hu J, Liu C, Liu L. Epigenetic regulation of diverse regulated cell death modalities in cardiovascular disease: Insights into necroptosis, pyroptosis, ferroptosis, and cuproptosis. Redox Biol 2024; 76:103321. [PMID: 39186883 PMCID: PMC11388786 DOI: 10.1016/j.redox.2024.103321] [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: 07/13/2024] [Revised: 08/17/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024] Open
Abstract
Cell death constitutes a critical component of the pathophysiology of cardiovascular diseases. A growing array of non-apoptotic forms of regulated cell death (RCD)-such as necroptosis, ferroptosis, pyroptosis, and cuproptosis-has been identified and is intimately linked to various cardiovascular conditions. These forms of RCD are governed by genetically programmed mechanisms within the cell, with epigenetic modifications being a common and crucial regulatory method. Such modifications include DNA methylation, RNA methylation, histone methylation, histone acetylation, and non-coding RNAs. This review recaps the roles of DNA methylation, RNA methylation, histone modifications, and non-coding RNAs in cardiovascular diseases, as well as the mechanisms by which epigenetic modifications regulate key proteins involved in cell death. Furthermore, we systematically catalog the existing epigenetic pharmacological agents targeting novel forms of RCD and their mechanisms of action in cardiovascular diseases. This article aims to underscore the pivotal role of epigenetic modifications in precisely regulating specific pathways of novel RCD in cardiovascular diseases, thus offering potential new therapeutic avenues that may prove more effective and safer than traditional treatments.
Collapse
Affiliation(s)
- Cong Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China
| | - Jie Wang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China.
| | - Shan Zhang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Xueying Zhu
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jun Hu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China
| | - Chao Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China
| | - Lanchun Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing, 100053, China
| |
Collapse
|
9
|
Nandi S, Sikder R, Nag A, Khatua S, Sen S, Chakraborty N, Naskar A, Zhakipbekov K, Acharya K, Habtemariam S, Arslan Ateşşahin D, Goloshvili T, Ahmed Aldahish A, Sharifi‐Rad J, Calina D. Updated aspects of alpha-Solanine as a potential anticancer agent: Mechanistic insights and future directions. Food Sci Nutr 2024; 12:7088-7107. [PMID: 39479710 PMCID: PMC11521658 DOI: 10.1002/fsn3.4221] [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: 01/12/2024] [Revised: 04/11/2024] [Accepted: 05/01/2024] [Indexed: 11/02/2024] Open
Abstract
Cancer remains a critical global health challenge, with limited progress in reducing mortality despite advancements in diagnosis and treatment. The growing resistance of tumors to existing chemotherapy exacerbates this burden. In response, the search for new anticancer compounds from plants has intensified, given their historical success in yielding effective treatments. This review focuses on α-solanine, a glycoalkaloid primarily derived from potato tubers and nightshade family plants, recognized for its diverse biological activities, including anti-allergic, antipyretic, anti-inflammatory, anti-diabetic, and antibiotic properties. Recently, α-solanine has gained attention as a potential anticancer agent. Utilizing resources like PubMed/MedLine, ScienceDirect, Web of Science, Scopus, the American Chemical Society, Google Scholar, Springer Link, Wiley, and various commercial websites, this review consolidates two decades of research on α-solanine's anticancer effects and mechanisms against nine different cancers, highlighting its role in modulating various signaling pathways. It also discusses α-solanine's potential as a lead compound in cancer therapy. The abundant availability of potato peel, often discarded as waste or sold cheaply, is suggested as a sustainable source for large-scale α-solanine extraction. The study concludes that α-solanine holds promise as a standalone or adjunctive cancer treatment. However, further research is necessary to optimize this lead compound and mitigate its toxicity through various strategies.
Collapse
Affiliation(s)
- Sudeshna Nandi
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of BotanyUniversity of CalcuttaKolkataIndia
| | - Rimpa Sikder
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of BotanyUniversity of CalcuttaKolkataIndia
| | - Anish Nag
- Department of Life SciencesCHRIST (Deemed to be University)BangaloreKarnatakaIndia
| | - Somanjana Khatua
- Department of Botany, Faculty of ScienceUniversity of AllahabadPrayagrajUttar PradeshIndia
| | - Surjit Sen
- Department of BotanyFakir Chand CollegeKolkataIndia
| | | | - Arghya Naskar
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of BotanyUniversity of CalcuttaKolkataIndia
| | - Kairat Zhakipbekov
- Department of Organization and Management and Economics of Pharmacy and Clinical PharmacyAsfendiyarov Kazakh National Medical UniversityAlmatyKazakhstan
| | - Krishnendu Acharya
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of BotanyUniversity of CalcuttaKolkataIndia
| | | | - Dilek Arslan Ateşşahin
- Department of Plant and Animal Production, Baskil Vocational SchoolFırat UniversityElazıgTurkey
| | - Tamar Goloshvili
- Department of Plant Physiology and Genetic ResourcesInstitute of Botany, Ilia State UniversityTbilisiGeorgia
| | - Afaf Ahmed Aldahish
- Department of Pharmacology, College of PharmacyKing Khalid UniversityAbhaKingdom of Saudi Arabia
| | - Javad Sharifi‐Rad
- Department of Biomedical SciencesCollege of Medicine, Korea UniversitySeoulRepublic of Korea
| | - Daniela Calina
- Department of Clinical PharmacyUniversity of Medicine and Pharmacy of CraiovaCraiovaRomania
| |
Collapse
|
10
|
Zhang Y, Gong Y, Liang Z, Wu W, Chen J, Li Y, Chen R, Mei J, Huang Z, Sun J. Mitochondria- and endoplasmic reticulum-localizing iridium(III) complexes induce immunogenic cell death of 143B cells. J Inorg Biochem 2024; 259:112655. [PMID: 38943844 DOI: 10.1016/j.jinorgbio.2024.112655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Recent breakthroughs in cancer immunology have propelled immunotherapy to the forefront of cancer research as a promising treatment approach that harnesses the body's immune system to effectively identify and eliminate cancer cells. In this study, three novel cyclometalated Ir(III) complexes, Ir1, Ir2, and Ir3, were designed, synthesized, and assessed in vitro for cytotoxic activity against several tumor-derived cell lines. Among these, Ir1 exhibited the highest cytotoxic activity, with an IC50 value of 0.4 ± 0.1 μM showcasing its significant anticancer potential. Detailed mechanistic analysis revealed that co-incubation of Ir1 with 143B cells led to Ir1 accumulation within mitochondria and the endoplasmic reticulum (ER). Furthermore, Ir1 induced G0/G1 phase cell cycle arrest, while also diminishing mitochondrial membrane potential, disrupting mitochondrial function, and triggering ER stress. Intriguingly, in mice the Ir1-induced ER stress response disrupted calcium homeostasis to thereby trigger immunogenic cell death (ICD), which subsequently activated the host antitumor immune response while concurrently dampening the in vivo tumor-induced inflammatory response.
Collapse
Affiliation(s)
- Yuqing Zhang
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yao Gong
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zhijun Liang
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Wei Wu
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Jiaxi Chen
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Yuling Li
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Rui Chen
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Jun Mei
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zunnan Huang
- Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Medical University, Dongguan 523808, China.
| | - Jing Sun
- The First Dongguan Affiliated Hospital, School of Pharmacy, Guangdong Medical University, Dongguan 523808, China; Key Laboratory of Computer-Aided Drug Design of Dongguan City, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
11
|
Scimeca M, Palumbo V, Giacobbi E, Servadei F, Casciardi S, Cornella E, Cerbara F, Rotondaro G, Seghetti C, Scioli MP, Montanaro M, Barillà F, Sisto R, Melino G, Mauriello A, Bonfiglio R. Impact of the environmental pollution on cardiovascular diseases: From epidemiological to molecular evidence. Heliyon 2024; 10:e38047. [PMID: 39328571 PMCID: PMC11425171 DOI: 10.1016/j.heliyon.2024.e38047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Environmental pollution poses a significant threat to human health, particularly concerning its impact on cardiovascular diseases (CVDs). This review synthesizes epidemiological and molecular evidence to elucidate the intricate relationship between environmental pollutants and CVDs. Epidemiological studies highlight the association between exposure to air, water, and soil pollutants and increased CVD risk, including hypertension, coronary artery disease, and stroke. Furthermore, molecular investigations unravel the underlying mechanisms linking pollutant exposure to CVD pathogenesis, such as oxidative stress, inflammation, endothelial dysfunction, and autonomic imbalance. Understanding these molecular pathways is crucial for developing targeted interventions and policy strategies to mitigate the adverse effects of environmental pollution on cardiovascular health. By integrating epidemiological and molecular evidence, this review provides insights into the complex interplay between environmental factors and CVDs, emphasizing the urgent need for comprehensive preventive measures and environmental policies to safeguard public health.
Collapse
Affiliation(s)
- Manuel Scimeca
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133, Rome, Italy
| | - Valeria Palumbo
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133, Rome, Italy
| | - Erica Giacobbi
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133, Rome, Italy
| | - Francesca Servadei
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133, Rome, Italy
| | - Stefano Casciardi
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, INAIL Research, Monte Porzio Catone, Rome, 00078, Italy
| | - Elena Cornella
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133, Rome, Italy
| | - Federica Cerbara
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133, Rome, Italy
| | - Gabriele Rotondaro
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133, Rome, Italy
| | - Christian Seghetti
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133, Rome, Italy
| | - Maria Paola Scioli
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133, Rome, Italy
| | - Manuela Montanaro
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133, Rome, Italy
| | - Francesco Barillà
- Department of Systems Medicine, Tor Vergata University, 00133, Rome, Italy
| | - Renata Sisto
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, INAIL Research, Monte Porzio Catone, Rome, 00078, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133, Rome, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133, Rome, Italy
| | - Rita Bonfiglio
- Department of Experimental Medicine, TOR, University of Rome “Tor Vergata”, 00133, Rome, Italy
| |
Collapse
|
12
|
Guo D, Liu Z, Zhou J, Ke C, Li D. Significance of Programmed Cell Death Pathways in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:9947. [PMID: 39337436 PMCID: PMC11432010 DOI: 10.3390/ijms25189947] [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: 07/27/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Programmed cell death (PCD) is a form of cell death distinct from accidental cell death (ACD) and is also referred to as regulated cell death (RCD). Typically, PCD signaling events are precisely regulated by various biomolecules in both spatial and temporal contexts to promote neuronal development, establish neural architecture, and shape the central nervous system (CNS), although the role of PCD extends beyond the CNS. Abnormalities in PCD signaling cascades contribute to the irreversible loss of neuronal cells and function, leading to the onset and progression of neurodegenerative diseases. In this review, we summarize the molecular processes and features of different modalities of PCD, including apoptosis, necroptosis, pyroptosis, ferroptosis, cuproptosis, and other novel forms of PCD, and their effects on the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), multiple sclerosis (MS), traumatic brain injury (TBI), and stroke. Additionally, we examine the key factors involved in these PCD signaling pathways and discuss the potential for their development as therapeutic targets and strategies. Therefore, therapeutic strategies targeting the inhibition or facilitation of PCD signaling pathways offer a promising approach for clinical applications in treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Dong Guo
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Zhihao Liu
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Jinglin Zhou
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Chongrong Ke
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Daliang Li
- College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| |
Collapse
|
13
|
Zheng Y, Yan F, He S, Luo L. Targeting ferroptosis in autoimmune diseases: Mechanisms and therapeutic prospects. Autoimmun Rev 2024:103640. [PMID: 39278299 DOI: 10.1016/j.autrev.2024.103640] [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: 06/25/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Ferroptosis is a form of regulated cell death that relies on iron and exhibits unique characteristics, including disrupted iron balance, reduced antioxidant defenses, and abnormal lipid peroxidation. Recent research suggests that ferroptosis is associated with the onset and progression of autoimmune disorders such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), inflammatory bowel disease (IBD), and multiple sclerosis (MS). However, the precise effects and molecular mechanisms remain incompletely understood. This article presents an overview of how ferroptosis mechanisms contribute to the development and advancement of autoimmune diseases, as well as the involvement of various immune cells in linking ferroptosis to autoimmune conditions. It also explores potential drug targets within the ferroptosis pathway and recent advancements in therapeutic approaches aimed at preventing and treating autoimmune diseases by targeting ferroptosis. Lastly, the article discusses the challenges and opportunities in utilizing ferroptosis as a potential therapeutic avenue for autoimmune disorders.
Collapse
Affiliation(s)
- Yingzi Zheng
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Fangfang Yan
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, Guangdong, China
| | - Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Chinese Medicine, Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing, China.
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, China.
| |
Collapse
|
14
|
Nixon RA. Autophagy-lysosomal-associated neuronal death in neurodegenerative disease. Acta Neuropathol 2024; 148:42. [PMID: 39259382 PMCID: PMC11418399 DOI: 10.1007/s00401-024-02799-7] [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: 03/19/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/13/2024]
Abstract
Autophagy, the major lysosomal pathway for degrading damaged or obsolete constituents, protects neurons by eliminating toxic organelles and peptides, restoring nutrient and energy homeostasis, and inhibiting apoptosis. These functions are especially vital in neurons, which are postmitotic and must survive for many decades while confronting mounting challenges of cell aging. Autophagy failure, especially related to the declining lysosomal ("phagy") functions, heightens the neuron's vulnerability to genetic and environmental factors underlying Alzheimer's disease (AD) and other late-age onset neurodegenerative diseases. Components of the global autophagy-lysosomal pathway and the closely integrated endolysosomal system are increasingly implicated as primary targets of these disorders. In AD, an imbalance between heightened autophagy induction and diminished lysosomal function in highly vulnerable pyramidal neuron populations yields an intracellular lysosomal build-up of undegraded substrates, including APP-βCTF, an inhibitor of lysosomal acidification, and membrane-damaging Aβ peptide. In the most compromised of these neurons, β-amyloid accumulates intraneuronally in plaque-like aggregates that become extracellular senile plaques when these neurons die, reflecting an "inside-out" origin of amyloid plaques seen in human AD brain and in mouse models of AD pathology. In this review, the author describes the importance of lysosomal-dependent neuronal cell death in AD associated with uniquely extreme autophagy pathology (PANTHOS) which is described as triggered by lysosomal membrane permeability during the earliest "intraneuronal" stage of AD. Effectors of other cell death cascades, notably calcium-activated calpains and protein kinases, contribute to lysosomal injury that induces leakage of cathepsins and activation of additional death cascades. Subsequent events in AD, such as microglial invasion and neuroinflammation, induce further cytotoxicity. In major neurodegenerative disease models, neuronal death and ensuing neuropathologies are substantially remediable by reversing underlying primary lysosomal deficits, thus implicating lysosomal failure and autophagy dysfunction as primary triggers of lysosomal-dependent cell death and AD pathogenesis and as promising therapeutic targets.
Collapse
Affiliation(s)
- Ralph A Nixon
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, 10962, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, 10016, USA.
- Neuroscience Institute, New York University, New York, NY, 10012, USA.
| |
Collapse
|
15
|
Lambrecht R, Jansen J, Rudolf F, El-Mesery M, Caporali S, Amelio I, Stengel F, Brunner T. Drug-induced oxidative stress actively prevents caspase activation and hepatocyte apoptosis. Cell Death Dis 2024; 15:659. [PMID: 39245717 PMCID: PMC11381522 DOI: 10.1038/s41419-024-06998-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/30/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024]
Abstract
Cell death is a fundamental process in health and disease. Emerging research shows the existence of numerous distinct cell death modalities with similar and intertwined signaling pathways, but resulting in different cellular outcomes, raising the need to understand the decision-making steps during cell death signaling. Paracetamol (Acetaminophen, APAP)-induced hepatocyte death includes several apoptotic processes but eventually is executed by oncotic necrosis without any caspase activation. Here, we studied this paradoxical form of cell death and revealed that APAP not only fails to activate caspases but also strongly impedes their activation upon classical apoptosis induction, thereby shifting apoptosis to necrosis. While APAP intoxication results in massive drop in mitochondrial respiration, low cellular ATP levels could be excluded as an underlying cause of missing apoptosome formation and caspase activation. In contrast, we identified oxidative stress as a key factor in APAP-induced caspase inhibition. Importantly, caspase inhibition and the associated switch from apoptotic to necrotic cell death was reversible through the administration of antioxidants. Thus, exemplified by APAP-induced cell death, our study stresses that cellular redox status is a critical component in the decision-making between apoptotic and necrotic cell death, as it directly affects caspase activity.
Collapse
Affiliation(s)
- Rebekka Lambrecht
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
- Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Jasmin Jansen
- Biochemistry and Mass Spectrometry, Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Franziska Rudolf
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
- Collaborative Research Center TRR 353, Konstanz, Germany
| | - Mohamed El-Mesery
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany
- Collaborative Research Center TRR 353, Konstanz, Germany
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Sabrina Caporali
- Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Ivano Amelio
- Collaborative Research Center TRR 353, Konstanz, Germany
- Systems Toxicology, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Florian Stengel
- Biochemistry and Mass Spectrometry, Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
- Collaborative Research Center TRR 353, Konstanz, Germany
| | - Thomas Brunner
- Biochemical Pharmacology, Department of Biology, University of Konstanz, Konstanz, Germany.
- Collaborative Research Center TRR 353, Konstanz, Germany.
| |
Collapse
|
16
|
Mirzayans R, Murray D. Amitotic Cell Division, Malignancy, and Resistance to Anticancer Agents: A Tribute to Drs. Walen and Rajaraman. Cancers (Basel) 2024; 16:3106. [PMID: 39272964 PMCID: PMC11394378 DOI: 10.3390/cancers16173106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
Cell division is crucial for the survival of living organisms. Human cells undergo three types of cell division: mitosis, meiosis, and amitosis. The former two types occur in somatic cells and germ cells, respectively. Amitosis involves nuclear budding and occurs in cells that exhibit abnormal nuclear morphology (e.g., polyploidy) with increased cell size. In the early 2000s, Kirsten Walen and Rengaswami Rajaraman and his associates independently reported that polyploid human cells are capable of producing progeny via amitotic cell division, and that a subset of emerging daughter cells proliferate rapidly, exhibit stem cell-like properties, and can contribute to tumorigenesis. Polyploid cells that arise in solid tumors/tumor-derived cell lines are referred to as polyploid giant cancer cells (PGCCs) and are known to contribute to therapy resistance and disease recurrence following anticancer treatment. This commentary provides an update on some of these intriguing discoveries as a tribute to Drs. Walen and Rajaraman.
Collapse
Affiliation(s)
- Razmik Mirzayans
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - David Murray
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| |
Collapse
|
17
|
An G, Hui J, Zhang W, Fan A, Zhou Y, Zhao X, Lu Y, Wang X. A novel lncRNA associated with the prognosis of patients with colorectal cancer resists apoptosis through the LYN/BCL-2 pathway. Biochem Biophys Res Commun 2024; 723:150177. [PMID: 38810320 DOI: 10.1016/j.bbrc.2024.150177] [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: 03/22/2024] [Revised: 05/14/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024]
Abstract
PURPOSE We found a novel lncRNA named lncAC138150.2 related to the overall survival and staging of patients with colorectal cancer (CRC) by bioinformatic analysis using data from the Cancer Genome Atlas (TCGA), and the study aimed to elucidate the function of lncAC138150.2 and underlying mechanisms. METHODS Target molecules were knocked down by transfection with antisense oligonucleotides (ASOs), siRNAs, or lentiviruses and overexpressed by transfection with plasmids. The function of lncAC138150.2 was determined using histological, cytological, and molecular biology methods. The underlying mechanism of lncAC138150.2 function was investigated using RNA-seq, bioinformatics analysis, and molecular biology methods. RESULTS The expression of lncAC138150.2 was increased in colorectal tissues compared with paired normal tissues. The lncAC138150.2 knockdown increased apoptosis but did not change the cell proliferation, cell cycle distribution, or cell migration ability of CRC cells, while lncAC138150.2 overexpression decreased CRC apoptosis. lncAC138150.2 was mainly located in the cell nucleus, and each lncAC138150.2 transcript knockdown increased CRC apoptosis. BCL-2 pathway was significantly altered in apoptosis induced by lncAC138150.2 knockdown, which was alleviated by BAX knockdown. The expression of LYN was significantly decreased with lncAC138150.2 knockdown, LYN knockdown increased CRC apoptosis, and its overexpression completely alleviated CRC apoptosis induced by lncAC138150.2 knockdown. CONCLUSION lncAC138150.2 significantly inhibited CRC apoptosis and affected the prognosis of patients with CRC, through the LYN/BCL-2 pathway.
Collapse
Affiliation(s)
- Guangzhou An
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China; Department of Radiation Protection Medicine, Ministry of Education Key Laboratory of Hazard Assessment and Control in Special Operational Environment, Faculty of Preventive Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Juan Hui
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Wenyao Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, China
| | - Ahui Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, China
| | - Yun Zhou
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Xiaodi Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, China
| | - Yuanyuan Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi'an, 710032, China.
| | - Xin Wang
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China.
| |
Collapse
|
18
|
De Martino M, Rathmell JC, Galluzzi L, Vanpouille-Box C. Cancer cell metabolism and antitumour immunity. Nat Rev Immunol 2024; 24:654-669. [PMID: 38649722 PMCID: PMC11365797 DOI: 10.1038/s41577-024-01026-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/25/2024]
Abstract
Accumulating evidence suggests that metabolic rewiring in malignant cells supports tumour progression not only by providing cancer cells with increased proliferative potential and an improved ability to adapt to adverse microenvironmental conditions but also by favouring the evasion of natural and therapy-driven antitumour immune responses. Here, we review cancer cell-intrinsic and cancer cell-extrinsic mechanisms through which alterations of metabolism in malignant cells interfere with innate and adaptive immune functions in support of accelerated disease progression. Further, we discuss the potential of targeting such alterations to enhance anticancer immunity for therapeutic purposes.
Collapse
Affiliation(s)
- Mara De Martino
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Jeffrey C Rathmell
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| | - Claire Vanpouille-Box
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
| |
Collapse
|
19
|
Sarkar R, Choudhury SM, Kanneganti TD. Classical apoptotic stimulus, staurosporine, induces lytic inflammatory cell death, PANoptosis. J Biol Chem 2024; 300:107676. [PMID: 39151726 PMCID: PMC11418131 DOI: 10.1016/j.jbc.2024.107676] [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: 03/09/2024] [Revised: 07/27/2024] [Accepted: 08/03/2024] [Indexed: 08/19/2024] Open
Abstract
Innate immunity is the body's first line of defense against disease, and regulated cell death is a central component of this response that balances pathogen clearance and inflammation. Cell death pathways are generally categorized as non-lytic and lytic. While non-lytic apoptosis has been extensively studied in health and disease, lytic cell death pathways are also increasingly implicated in infectious and inflammatory diseases and cancers. Staurosporine (STS) is a well-known inducer of non-lytic apoptosis. However, in this study, we observed that STS also induces lytic cell death at later timepoints. Using biochemical assessments with genetic knockouts, pharmacological inhibitors, and gene silencing, we identified that STS triggered PANoptosis via the caspase-8/RIPK3 axis, which was mediated by RIPK1. PANoptosis is a lytic, innate immune cell death pathway initiated by innate immune sensors and driven by caspases and RIPKs through PANoptosome complexes. Deletion of caspase-8 and RIPK3, core components of the PANoptosome complex, protected against STS-induced lytic cell death. Overall, our study identifies STS as a time-dependent inducer of lytic cell death, PANoptosis. These findings emphasize the importance of understanding trigger- and time-specific activation of distinct cell death pathways to advance our understanding of the molecular mechanisms of innate immunity and cell death for clinical translation.
Collapse
Affiliation(s)
- Roman Sarkar
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Sk Mohiuddin Choudhury
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | |
Collapse
|
20
|
Maremonti F, Tonnus W, Gavali S, Bornstein S, Shah A, Giacca M, Linkermann A. Ferroptosis-based advanced therapies as treatment approaches for metabolic and cardiovascular diseases. Cell Death Differ 2024; 31:1104-1112. [PMID: 39068204 PMCID: PMC11369293 DOI: 10.1038/s41418-024-01350-1] [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: 05/12/2024] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024] Open
Abstract
Ferroptosis has attracted attention throughout the last decade because of its tremendous clinical importance. Here, we review the rapidly growing body of literature on how inhibition of ferroptosis may be harnessed for the treatment of common diseases, and we focus on metabolic and cardiovascular unmet medical needs. We introduce four classes of preclinically established ferroptosis inhibitors (ferrostatins) such as iron chelators, radical trapping agents that function in the cytoplasmic compartment, lipophilic radical trapping antioxidants and ninjurin-1 (NINJ1) specific monoclonal antibodies. In contrast to ferroptosis inducers that cause serious untoward effects such as acute kidney tubular necrosis, the side effect profile of ferrostatins appears to be limited. We also consider ferroptosis as a potential side effect itself when several advanced therapies harnessing small-interfering RNA (siRNA)-based treatment approaches are tested. Importantly, clinical trial design is impeded by the lack of an appropriate biomarker for ferroptosis detection in serum samples or tissue biopsies. However, we discuss favorable clinical scenarios suited for the design of anti-ferroptosis clinical trials to test such first-in-class compounds. We conclude that targeting ferroptosis exhibits outstanding treatment options for metabolic and cardiovascular diseases, but we have only begun to translate this knowledge into clinically relevant applications.
Collapse
Affiliation(s)
- Francesca Maremonti
- Division of Nephrology, Medical Clinic III, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
- Department of Medicine V, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Wulf Tonnus
- Division of Nephrology, Medical Clinic III, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Shubhangi Gavali
- Division of Nephrology, Medical Clinic III, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany
- Department of Medicine V, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Stefan Bornstein
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Diabetes and Nutritional Sciences, King's College London, London, UK
- Center for Regenerative Therapies, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of TU Dresden Faculty of Medicine, Dresden, Germany
| | - Ajay Shah
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Mauro Giacca
- King's College London British Heart Foundation Centre, School of Cardiovascular & Metabolic Medicine and Sciences, London, UK
| | - Andreas Linkermann
- Division of Nephrology, Medical Clinic III, University Hospital Dresden, Technische Universität Dresden, Dresden, Germany.
- Department of Medicine V, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany.
- Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
21
|
Li C, Hu J, Jiang X, Tan H, Mao Y. Identification and validation of an immune-derived multiple programmed cell death index for predicting clinical outcomes, molecular subtyping, and drug sensitivity in lung adenocarcinoma. Clin Transl Oncol 2024; 26:2274-2295. [PMID: 38563847 DOI: 10.1007/s12094-024-03439-y] [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: 01/23/2024] [Accepted: 03/01/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES Comprehensive cross-interaction of multiple programmed cell death (PCD) patterns in the patients with lung adenocarcinoma (LUAD) have not yet been thoroughly investigated. METHODS Here, we collected 19 different PCD patterns, including 1911 PCD-related genes, and developed an immune-derived multiple programmed cell death index (MPCDI) based on machine learning methods. RESULTS Using the median MPCDI scores, we categorized the LUAD patients into two groups: low-MPCDI and high-MPCDI. Our analysis of the TCGA-LUAD training cohort and three external GEO cohorts (GSE37745, GSE30219, and GSE68465) revealed that patients with high-MPCDI experienced a more unfavorable prognosis, whereas those with low-MPCDI had a better prognosis. Furthermore, the results of both univariate and multivariate Cox regression analyses further confirmed that MPCDI serves as a novel independent risk factor. By combining clinical characteristics with the MPCDI, we constructed a nomogram that provides an accurate and reliable quantitative tool for personalized clinical management of LUAD patients. The findings obtained from the analysis of C-index and the decision curve revealed that the nomogram outperformed various clinical variables in terms of net clinical benefit. Encouragingly, the low-MPCDI patients are more sensitive to commonly used chemotherapy drugs, which suggests that MPCDI scores have a guiding role in chemotherapy for LUAD patients. CONCLUSION Therefore, MPCDI can be used as a novel clinical diagnostic classifier, providing valuable insights into the clinical management and clinical decision-making for LUAD patients.
Collapse
Affiliation(s)
- Chunhong Li
- Central Laboratory, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, Guangxi, China.
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, Guangxi, China.
- Central Laboratory, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, Guangxi, China.
| | - Jiahua Hu
- Central Laboratory, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, Guangxi, China
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Xiling Jiang
- School of Medical Laboratory Medicine, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Haiyin Tan
- School of Medical Laboratory Medicine, Guilin Medical University, Guilin, 541004, Guangxi, China
| | - Yiming Mao
- Department of Thoracic Surgery, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215028, China.
| |
Collapse
|
22
|
Liu Z, Hou P, Fang J, Shao C, Shi Y, Melino G, Peschiaroli A. Hyaluronic acid metabolism and chemotherapy resistance: recent advances and therapeutic potential. Mol Oncol 2024; 18:2087-2106. [PMID: 37953485 PMCID: PMC11467803 DOI: 10.1002/1878-0261.13551] [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: 08/03/2023] [Revised: 10/04/2023] [Accepted: 11/10/2023] [Indexed: 11/14/2023] Open
Abstract
Hyaluronic acid (HA) is a major component of the extracellular matrix, providing essential mechanical scaffolding for cells and, at the same time, mediating essential biochemical signals required for tissue homeostasis. Many solid tumors are characterized by dysregulated HA metabolism, resulting in increased HA levels in cancer tissues. HA interacts with several cell surface receptors, such as cluster of differentiation 44 and receptor for hyaluronan-mediated motility, thus co-regulating important signaling pathways in cancer development and progression. In this review, we describe the enzymes controlling HA metabolism and its intracellular effectors emphasizing their impact on cancer chemotherapy resistance. We will also explore the current and future prospects of HA-based therapy, highlighting the opportunities and challenges in the field.
Collapse
Affiliation(s)
- Zhanhong Liu
- Department of Experimental MedicineUniversity of Rome Tor VergataRomeItaly
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and ProtectionThe First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow UniversityChina
| | - Pengbo Hou
- Department of Experimental MedicineUniversity of Rome Tor VergataRomeItaly
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and ProtectionThe First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow UniversityChina
| | - Jiankai Fang
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and ProtectionThe First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow UniversityChina
| | - Changshun Shao
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and ProtectionThe First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow UniversityChina
| | - Yufang Shi
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and ProtectionThe First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow UniversityChina
| | - Gerry Melino
- Department of Experimental MedicineUniversity of Rome Tor VergataRomeItaly
| | - Angelo Peschiaroli
- Institute of Translational Pharmacology (IFT), National Research Council (CNR)RomeItaly
| |
Collapse
|
23
|
Stefanes NM, de Oliveira Silva L, Walter LO, Steimbach JV, Markendorf E, Ribeiro AAB, Feuser PE, Cordeiro AP, Santos-Silva MC. Sodium diethyldithiocarbamate trihydrate: an effective and selective compound for hematological malignancies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03399-8. [PMID: 39186189 DOI: 10.1007/s00210-024-03399-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
Myeloid leukemias and lymphomas are among the most common and well-studied hematological malignancies. However, due to the aggressiveness and rapid progression of certain subtypes, treating these diseases remains a challenge. Considering the promising results of diethyldithiocarbamates in preclinical and clinical oncology trials, this study aimed to investigate the potential of sodium diethyldithiocarbamate trihydrate (DETC) as a prototype for developing new drugs to treat hematological malignancies. In silico analysis using SwissADME was conducted to evaluate the physicochemical characteristics and pharmacokinetic properties of DETC. An in vitro investigation utilizing the MTT assay assessed the cytotoxic effects of DETC on neoplastic and non-neoplastic cell lines. Selectivity was determined using a selectivity index and a hemolysis assay, while the mechanism of cell death in neoplastic cell lines was examined through flow cytometry analysis of pro-apoptotic and anti-apoptotic protein levels. The results demonstrated that the physicochemical characteristics of DETC are suitable for oral administration. Furthermore, the compound showed promising cytotoxic activity against human myeloid leukemia (K562) and Burkitt's lymphoma (Daudi) cell lines, with high selectivity for neoplastic cells over non-neoplastic cells of the bone marrow microenvironment (HS-5 cell line). Moreover, hemolysis was observed only at very high concentrations. The cytotoxicity mechanism of DETC against both neoplastic cell lines involved cell cycle arrest and the production of reactive oxygen species. In K562 cells, cell death was induced via apoptosis. Additional experiments are needed to confirm the exact mechanism of cell death in Daudi Burkitt's lymphoma cells.
Collapse
Affiliation(s)
- Natália Marcéli Stefanes
- Experimental Oncology and Hemopathies Laboratory, Department of Clinical Analysis, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
- Graduate Program in Pharmacy, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Lisandra de Oliveira Silva
- Experimental Oncology and Hemopathies Laboratory, Department of Clinical Analysis, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
- Graduate Program in Pharmacy, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Laura Otto Walter
- Experimental Oncology and Hemopathies Laboratory, Department of Clinical Analysis, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
- Graduate Program in Pharmacy, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - João Vitor Steimbach
- Experimental Oncology and Hemopathies Laboratory, Department of Clinical Analysis, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
- Graduate Program in Pharmacy, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Emanueli Markendorf
- Experimental Oncology and Hemopathies Laboratory, Department of Clinical Analysis, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Amanda Abdalla Biasi Ribeiro
- Experimental Oncology and Hemopathies Laboratory, Department of Clinical Analysis, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
- Graduate Program in Pharmacy, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Paulo Emílio Feuser
- Graduate Program in Chemical Engineering, Technological Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Arthur Poester Cordeiro
- Graduate Program in Chemical Engineering, Technological Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Maria Cláudia Santos-Silva
- Experimental Oncology and Hemopathies Laboratory, Department of Clinical Analysis, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil.
- Graduate Program in Pharmacy, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil.
| |
Collapse
|
24
|
Marković T, Popović S, Matić S, Mitrović M, Anđić M, Kočović A, Vukić M, Petrović V, Branković J, Vuković N, Todorović D, Kačániová M, Baskić D. Insights into Molecular Mechanisms of Anticancer Activity of Juniperus communis Essential Oil in HeLa and HCT 116 Cells. PLANTS (BASEL, SWITZERLAND) 2024; 13:2351. [PMID: 39273835 PMCID: PMC11397105 DOI: 10.3390/plants13172351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024]
Abstract
As cancer remains a significant global health challenge, there is an increasing need for novel therapeutic approaches. We investigated the antitumor potential of Juniperus communis berry essential oil on cervical cancer HeLa and colorectal HCT 116 cells. Cytotoxicity was evaluated through the MTT assay, revealing concentration-dependent reductions in cell viability. A clonogenic assay demonstrated long-term cytotoxic effects. Apoptosis markers were assessed via flow cytometric analysis and showed an induction of the intrinsic pathway in both cell lines, demonstrated by the elevated levels of cleaved caspase-3, Bax/Bcl-2 ratio, JC-10 monomer formation, and cytochrome C migration to the cytosol. The treatment inhibited cell-survival pathways in HCT 116 cells and arrested HeLa cells in the S phase. An extensive molecular docking screening provided insight into the binding affinity and interaction patterns of the essential oil components with NADH ubiquinone oxidoreductase and superoxide dismutase enzymes, further confirming the induction of the intrinsic pathway of apoptosis. The obtained in silico and in vitro results indicated the anticancer potential of J. communis berry essential oil as it interferes with cancer cell molecular mechanisms. Our findings highlight J. communis berry essential oil as a promising natural agent with anticancer potential.
Collapse
Affiliation(s)
- Tijana Marković
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića, 69, 34000 Kragujevac, Serbia
| | - Suzana Popović
- Centre for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića, 69, 34000 Kragujevac, Serbia
| | - Sanja Matić
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića, 69, 34000 Kragujevac, Serbia
| | - Marina Mitrović
- Department of Medical Biochemistry, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića, 69, 34000 Kragujevac, Serbia
| | - Marijana Anđić
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića, 69, 34000 Kragujevac, Serbia
| | - Aleksandar Kočović
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića, 69, 34000 Kragujevac, Serbia
| | - Milena Vukić
- Department of Chemistry, Faculty of Science, University of Kragujevac, Radoja Domanovića, 12, 34000 Kragujevac, Serbia
| | - Vladimir Petrović
- Department of Chemistry, Faculty of Science, University of Kragujevac, Radoja Domanovića, 12, 34000 Kragujevac, Serbia
| | - Jovica Branković
- Department of Chemistry, Faculty of Science, University of Kragujevac, Radoja Domanovića, 12, 34000 Kragujevac, Serbia
| | - Nenad Vuković
- Department of Chemistry, Faculty of Science, University of Kragujevac, Radoja Domanovića, 12, 34000 Kragujevac, Serbia
| | - Danijela Todorović
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića, 69, 34000 Kragujevac, Serbia
| | - Miroslava Kačániová
- Institute of Horticulture, Faculty of Horticulture and Landscape Engineering, Slovak University of Agriculture, Tr. A. Hlinku 2, 94976 Nitra, Slovakia
- School of Medical & Health Sciences, University of Economics and Human Sciences in Warsaw, Okopowa 59, 01 043 Warsaw, Poland
| | - Dejan Baskić
- Centre for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića, 69, 34000 Kragujevac, Serbia
- Institute of Public Health Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
25
|
Lian X, Tang X. Immune infiltration analysis based on pyroptosis-related gene in metabolic dysfunction-associated fatty liver disease. Heliyon 2024; 10:e34348. [PMID: 39145004 PMCID: PMC11320144 DOI: 10.1016/j.heliyon.2024.e34348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024] Open
Abstract
Introduction Metabolic dysfunction-associated fatty liver disease (MAFLD) is a prevalent chronic disease that can involve pyroptosis. The primary objective of this study was to conduct a thorough and comprehensive analysis the pyroptosis-related genes in MAFLD. Methods We identified pyroptosis-related differentially expressed genes (PRDEGs) in both healthy individuals and MAFLD patients. Using various bioinformatic approaches, we conducted an immune infiltration analysis from multiple perspectives. Results A total of 20 pyroptosis-related LASSO genes were obtained, and 10 hub genes were used to do immune infiltration analysis. The hub genes were utilized in the construction of interaction networks between mRNA-miRNA and mRNA-TF. Immune characteristics analysis revealed multiple immune cell types significantly related to PRDEG expression, particularly genes HSP90AA1, TSLP, CDK9, and BRD4. Conclusion Pyroptosis-related immune infiltration might be a mechanism of MAFLD progression and offers a research direction for potential treatment techniques.
Collapse
Affiliation(s)
- Xin Lian
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Xulei Tang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, 730000, China
| |
Collapse
|
26
|
Chakraborty S, Mishra A, Choudhuri A, Bhaumik T, Sengupta R. Leveraging the redundancy of S-denitrosylases in response to S-nitrosylation of caspases: Experimental strategies and beyond. Nitric Oxide 2024; 149:18-31. [PMID: 38823434 DOI: 10.1016/j.niox.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
Redox-based protein posttranslational modifications, such as S-nitrosylation of critical, active site cysteine thiols have garnered significant clinical attention and research interest, reasoning for one of the crucial biological implications of reactive messenger molecule, nitric oxide in the cellular repertoire. The stringency of the S-(de)nitrosylation-based redox switch governs the activity and contribution of several susceptible enzymes in signal transduction processes and diverse pathophysiological settings, thus establishing it as a transient yet reasonable, and regulated mechanism of NO adduction and release. Notably, endogenous proteases like cytosolic and mitochondrial caspases with a molecular weight ranging from 33 to 55 kDa are susceptible to performing this biochemistry in the presence of major oxidoreductases, which further unveils the enormous redox-mediated regulational control of caspases in the etiology of diseases. In addition to advancing the progress of the current state of understanding of 'redox biochemistry' in the field of medicine and enriching the existing dynamic S-nitrosoproteome, this review stands as a testament to an unprecedented shift in the underpinnings for redundancy and redox relay between the major redoxin/antioxidant systems, fine-tuning of which can command the apoptotic control of caspases at the face of nitro-oxidative stress. These intricate functional overlaps and cellular backups, supported rationally by kinetically favorable reaction mechanisms suggest the physiological relevance of identifying and involving such cognate substrates for cellular S-denitrosylases that can shed light on the bigger picture of extensively proposing targeted therapies and redox-based drug designing to potentially alleviate the side effects of NOx/ROS in disease pathogenesis.
Collapse
Affiliation(s)
- Surupa Chakraborty
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Akansha Mishra
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Ankita Choudhuri
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Tamal Bhaumik
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Rajib Sengupta
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India.
| |
Collapse
|
27
|
Chiu FC, Kuo HM, Yu CL, Selvam P, Su IL, Tseng CC, Yuan CH, Wen ZH. Marine-derived antimicrobial peptide piscidin-1 triggers extrinsic and intrinsic apoptosis in oral squamous cell carcinoma through reactive oxygen species production and inhibits angiogenesis. Free Radic Biol Med 2024; 220:28-42. [PMID: 38679300 DOI: 10.1016/j.freeradbiomed.2024.04.235] [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: 03/30/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
Cancer of the head and neck encompasses a wide range of cancers, including oral and oropharyngeal cancers. Oral cancer is often diagnosed at advanced stages and has a dismal prognosis. Piscidin-1, a marine antimicrobial peptide (AMP) containing approximately 22 amino acids, also exhibits significant anticancer properties. We investigated the possible anti-oral cancer effects of piscidin-1 and clarified the mechanisms underlying these effects. We treated the oral squamous cell carcinoma cell lines OC2 and SCC4 with piscidin-1. Cell viability and the expression of different hallmark apoptotic molecules, including reactive oxygen species (ROS), were tested using the appropriate MTT assay, flow cytometry and western blotting assays, and human umbilical vein endothelial cell (HUVEC) wound healing, migration, and tube formation (angiogenesis) assays. Piscidin-1 increases cleaved caspase 3 levels to induce apoptosis. Piscidin-1 also increases ROS levels and intensifies oxidative stress in the endoplasmic reticulum and mitochondria, causing mitochondrial dysfunction. Additionally, it decreases the oxygen consumption rates and activity of mitochondrial complexes I-V. As expected, the antioxidants MitoTEMPOL and N-acetylcysteine reduce piscidin-1-induced ROS generation and intracellular calcium accumulation. Piscidin-1 also inhibits matrix metalloproteinase (MMP)-2/-9 expression in HUVECs, affecting migration and tube formation angiogenesis. We demonstrated that piscidin-1 can promote apoptosis via both intrinsic and extrinsic apoptotic pathways and findings indicate that piscidin-1 has anti-proliferative and anti-angiogenic properties in oral cancer treatment. Our study on piscidin-1 thus provides a basis for future translational anti-oral cancer drug research and a new theoretical approach for anti-oral cancer clinical research.
Collapse
Affiliation(s)
- Fu-Ching Chiu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
| | - Hsiao-Mei Kuo
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan; Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 833301, Taiwan
| | - Chen-Ling Yu
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
| | - Padhmavathi Selvam
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
| | - I-Li Su
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Antai Medical Care Corporation, Antai Tian-Sheng Memorial Hospital, Pingtung, 92842, Taiwan
| | - Chung-Chih Tseng
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan; Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung, 80284, Taiwan
| | - Chien-Han Yuan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan; Department of Otolaryngology, Kaohsiung Armed Forces General Hospital, Kaohsiung, 80284, Taiwan; Department of Otolaryngology, National Defense Medical Center, Taipei 11490, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan.
| |
Collapse
|
28
|
Chen F, Lin J, Kang R, Tang D, Liu J. Alkaliptosis induction counteracts paclitaxel-resistant ovarian cancer cells via ATP6V0D1-mediated ABCB1 inhibition. Mol Carcinog 2024; 63:1515-1527. [PMID: 38751020 DOI: 10.1002/mc.23741] [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: 01/18/2024] [Revised: 03/23/2024] [Accepted: 05/04/2024] [Indexed: 07/10/2024]
Abstract
Paclitaxel serves as the cornerstone chemotherapy for ovarian cancer, yet its prolonged administration frequently culminates in drug resistance, presenting a substantial challenge. Here we reported that inducing alkaliptosis, rather than apoptosis or ferroptosis, effectively overcomes paclitaxel resistance. Mechanistically, ATPase H+ transporting V0 subunit D1 (ATP6V0D1), a key regulator of alkaliptosis, plays a pivotal role by mediating the downregulation of ATP-binding cassette subfamily B member 1 (ABCB1), a multidrug resistance protein. Both ATP6V0D1 overexpression through gene transfection and pharmacological enhancement of ATP6V0D1 protein stability using JTC801 effectively inhibit ABCB1 upregulation, resulting in growth inhibition in drug-resistant cells. Additionally, increasing intracellular pH to alkaline (pH 8.5) via sodium hydroxide application suppresses ABCB1 expression, whereas reducing the pH to acidic conditions (pH 6.5) with hydrochloric acid amplifies ABCB1 expression in drug-resistant cells. Collectively, these results indicate a potentially effective therapeutic strategy for targeting paclitaxel-resistant ovarian cancer by inducing ATP6V0D1-dependent alkaliptosis.
Collapse
Affiliation(s)
- Fangquan Chen
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Junhao Lin
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
29
|
Yin J, Yu Y, Huang X, Chan FKM. Necroptosis in immunity, tissue homeostasis, and cancer. Curr Opin Immunol 2024; 89:102455. [PMID: 39167896 DOI: 10.1016/j.coi.2024.102455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 08/06/2024] [Indexed: 08/23/2024]
Abstract
Immune and tissue homeostasis is achieved through balancing signals that regulate cell survival, proliferation, and cell death. Recent studies indicate that certain cell death programs can stimulate inflammation and are often referred as 'immunogenic cell death' (ICD). ICD is a double-edged sword that can confer protection against pathogen infection but also cause tissue damage. Necroptosis is a key ICD module that has been shown to participate in host defense against pathogen infection, tissue homeostasis, and cancer response to immunotherapy. Here, we will review recent findings on the regulation of necroptosis signaling and its role in pathogen infection, tissue homeostasis, and cancer.
Collapse
Affiliation(s)
| | - Yuqiang Yu
- Department of Cardiology of the Second Affiliated Hospital, China; State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, China
| | | | - Francis K-M Chan
- Department of Cardiology of the Second Affiliated Hospital, China; State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, China; Liangzhu Laboratory, China; Zhejiang University School of Medicine, 1369 West Wenyi Road, Hangzhou 311121, China.
| |
Collapse
|
30
|
Ge A, Chan C, Yang X. Exploring the Dark Matter of Human Proteome: The Emerging Role of Non-Canonical Open Reading Frame (ncORF) in Cancer Diagnosis, Biology, and Therapy. Cancers (Basel) 2024; 16:2660. [PMID: 39123386 PMCID: PMC11311765 DOI: 10.3390/cancers16152660] [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: 06/26/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Cancer develops from abnormal cell growth in the body, causing significant mortalities every year. To date, potent therapeutic approaches have been developed to eradicate tumor cells, but intolerable toxicity and drug resistance can occur in treated patients, limiting the efficiency of existing treatment strategies. Therefore, searching for novel genes critical for cancer progression and therapeutic response is urgently needed for successful cancer therapy. Recent advances in bioinformatics and proteomic techniques have allowed the identification of a novel category of peptides encoded by non-canonical open reading frames (ncORFs) from historically non-coding genomic regions. Surprisingly, many ncORFs express functional microproteins that play a vital role in human cancers. In this review, we provide a comprehensive description of different ncORF types with coding capacity and technological methods in discovering ncORFs among human genomes. We also summarize the carcinogenic role of ncORFs such as pTINCR and HOXB-AS3 in regulating hallmarks of cancer, as well as the roles of ncORFs such as HOXB-AS3 and CIP2A-BP in cancer diagnosis and prognosis. We also discuss how ncORFs such as AKT-174aa and DDUP are involved in anti-cancer drug response and the underestimated potential of ncORFs as therapeutic targets.
Collapse
Affiliation(s)
| | | | - Xiaolong Yang
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada; (A.G.); (C.C.)
| |
Collapse
|
31
|
Eid N, Bhatnagar P. Natural isothiocyanates of the genus Capparis as potential agonists of apoptosis and antitumor agents: Mechanisms and implications. World J Pharmacol 2024; 13:96477. [DOI: 10.5497/wjp.v13.i1.96477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024] Open
Abstract
In this editorial, we comment on a recent publication, which highlights the important findings from the study, including the antitumor and anti-inflammatory effects of isothiocyanates, their underlying mechanisms, and implications. Additionally, a related perspective is discussed.
Collapse
Affiliation(s)
- Nabil Eid
- Department of Anatomy, Division of Human Biology, School of Medicine, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Payal Bhatnagar
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
32
|
Xu J, Zhu M, Luo P, Gong Y. Machine Learning Screening and Validation of PANoptosis-Related Gene Signatures in Sepsis. J Inflamm Res 2024; 17:4765-4780. [PMID: 39051056 PMCID: PMC11268777 DOI: 10.2147/jir.s461809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024] Open
Abstract
Background Sepsis is a syndrome marked by life-threatening organ dysfunction and a disrupted host immune response to infection. PANoptosis is a recent conceptual development, which emphasises the interconnectedness among multiple programmed cell deaths in various diseases. Nevertheless, the role of PANoptosis in sepsis is still unclear. Methods We utilized the GSE65682 dataset to identify PANoptosis-related genes (PRGs) and associated immune characteristics in sepsis, classified sepsis samples based on PRGs using the ConsensusClusterPlus method and applied the Weighted Gene Co-Expression Network Analysis (WGCNA) algorithm to identify cluster-specific hub genes. Based on PANoptosis -specific DEGs, we compared results from machine learning models and the best-performing model was selected. Predictive efficiency was validated through external dataset, nomogram, survival analysis, quantitative real-time PCR, and western blot. Results The expression levels of PRGs were generally dysregulated in sepsis patients compared with normal samples, and higher PRGs expression correlated with increased immune cell infiltration. In addition, two distinct PANoptosis-related clusters were defined, and functional analysis indicated that DEGs associated with these clusters were primarily linked to immune-related pathways. The SVM model was selected as best-performing model, with lower residuals and the highest area under the curve (AUC = 0.967), which was then validated in an external dataset (AUC = 0.989) and through in vivo experiments. Additional validation through nomogram and survival analysis further confirmed its substantial predictive efficacy. Conclusion Our findings exposed the intricate association between PANoptosis and sepsis, offering important insights on sepsis diagnosis and potential therapeutic targets.
Collapse
Affiliation(s)
- Jingjing Xu
- Department of Intensive Care Unit, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Mingyu Zhu
- Department of Intensive Care Unit, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Pengxiang Luo
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Yuanqi Gong
- Department of Intensive Care Unit, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People’s Republic of China
| |
Collapse
|
33
|
Jannus F, Sainz J, Reyes-Zurita FJ. Principal Bioactive Properties of Oleanolic Acid, Its Derivatives, and Analogues. Molecules 2024; 29:3291. [PMID: 39064870 PMCID: PMC11279785 DOI: 10.3390/molecules29143291] [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: 06/22/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Natural products have always played an important role in pharmacotherapy, helping to control pathophysiological processes associated with human disease. Thus, natural products such as oleanolic acid (OA), a pentacyclic triterpene that has demonstrated important activities in several disease models, are in high demand. The relevant properties of this compound have motivated re-searchers to search for new analogues and derivatives using the OA as a scaffold to which new functional groups have been added or modifications have been realized. OA and its derivatives have been shown to be effective in the treatment of inflammatory processes, triggered by chronic diseases or bacterial and viral infections. OA and its derivatives have also been found to be effective in diabetic disorders, a group of common endocrine diseases characterized by hyperglycemia that can affect several organs, including the liver and brain. This group of compounds has been reported to exhibit significant bioactivity against cancer processes in vitro and in vivo. In this review, we summarize the bioactive properties of OA and its derivatives as anti-inflammatory, anti-bacterial, antiviral, anti-diabetic, hepatoprotective, neuroprotective, and anticancer agents.
Collapse
Affiliation(s)
- Fatin Jannus
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva, 18071 Granada, Spain;
| | - Juan Sainz
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva, 18071 Granada, Spain;
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Av. de la Ilustración, 114, PTS, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria IBs.Granada, 18010 Granada, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), University of Barcelona, 08908 Barcelona, Spain
| | - Fernando J. Reyes-Zurita
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, Av. Fuentenueva, 18071 Granada, Spain;
- Instituto de Investigación Biosanitaria IBs.Granada, 18010 Granada, Spain
| |
Collapse
|
34
|
Akiyama M, Kanayama M, Umezawa Y, Nagao T, Izumi Y, Yamamoto M, Ohteki T. An early regulatory mechanism of hyperinflammation by restricting monocyte contribution. Front Immunol 2024; 15:1398153. [PMID: 39040105 PMCID: PMC11260625 DOI: 10.3389/fimmu.2024.1398153] [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: 03/09/2024] [Accepted: 06/06/2024] [Indexed: 07/24/2024] Open
Abstract
Innate immune cells play a key role in inflammation as a source of pro-inflammatory cytokines. However, it remains unclear how innate immunity-mediated inflammation is fine-tuned to minimize tissue damage and assure the host's survival at the early phase of systemic inflammation. The results of this study with mouse models demonstrate that the supply of monocytes is restricted depending on the magnitude of inflammation. During the acute phase of severe inflammation, monocytes, but not neutrophils, were substantially reduced by apoptosis and the remaining monocytes were dysfunctional in the bone marrow. Monocyte-specific ablation of Casp3/7 prevented monocyte apoptosis but promoted monocyte necrosis in the bone marrow, leading to elevated levels of pro-inflammatory cytokines and the increased mortality of mice during systemic inflammation. Importantly, the limitation of monocyte supply was dependent on pro-inflammatory cytokines in vivo. Consistently, a reduction of monocytes was observed in the peripheral blood during cytokine-release syndrome (CRS) patients, a pathogen-unrelated systemic inflammation induced by chimeric antigen receptor-T cell (CAR-T cell) therapy. Thus, monocytes act as a safety valve to alleviate tissue damage caused by inflammation and ensure host survival, which may be responsible for a primitive immune-control mechanism that does not require intervention by acquired immunity.
Collapse
Affiliation(s)
- Megumi Akiyama
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Department of Hematology, Graduate School of Medical and Dental Sciences, TMDU, Tokyo, Japan
| | - Masashi Kanayama
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yoshihiro Umezawa
- Department of Hematology, Graduate School of Medical and Dental Sciences, TMDU, Tokyo, Japan
| | - Toshikage Nagao
- Department of Hematology, Graduate School of Medical and Dental Sciences, TMDU, Tokyo, Japan
| | - Yuta Izumi
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masahide Yamamoto
- Department of Hematology, Graduate School of Medical and Dental Sciences, TMDU, Tokyo, Japan
| | - Toshiaki Ohteki
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
35
|
Guilbaud E, Galluzzi L. A mitochondrial checkpoint to NF-κB signaling. Cell Death Dis 2024; 15:477. [PMID: 38961079 PMCID: PMC11222492 DOI: 10.1038/s41419-024-06868-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/17/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
Mitochondrial dysfunction can elicit multiple inflammatory pathways, especially when apoptotic caspases are inhibited. Such an inflammatory program is negatively regulated by the autophagic disposal of permeabilized mitochondria. Recent data demonstrate that the ubiquitination of mitochondrial proteins is essential for NEMO-driven NF-kB activation downstream of mitochondrial permeabilization.
Collapse
Affiliation(s)
- Emma Guilbaud
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
36
|
Jin X, Jin W, Tong L, Zhao J, Zhang L, Lin N. Therapeutic strategies of targeting non-apoptotic regulated cell death (RCD) with small-molecule compounds in cancer. Acta Pharm Sin B 2024; 14:2815-2853. [PMID: 39027232 PMCID: PMC11252466 DOI: 10.1016/j.apsb.2024.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/29/2024] [Accepted: 03/18/2024] [Indexed: 07/20/2024] Open
Abstract
Regulated cell death (RCD) is a controlled form of cell death orchestrated by one or more cascading signaling pathways, making it amenable to pharmacological intervention. RCD subroutines can be categorized as apoptotic or non-apoptotic and play essential roles in maintaining homeostasis, facilitating development, and modulating immunity. Accumulating evidence has recently revealed that RCD evasion is frequently the primary cause of tumor survival. Several non-apoptotic RCD subroutines have garnered attention as promising cancer therapies due to their ability to induce tumor regression and prevent relapse, comparable to apoptosis. Moreover, they offer potential solutions for overcoming the acquired resistance of tumors toward apoptotic drugs. With an increasing understanding of the underlying mechanisms governing these non-apoptotic RCD subroutines, a growing number of small-molecule compounds targeting single or multiple pathways have been discovered, providing novel strategies for current cancer therapy. In this review, we comprehensively summarized the current regulatory mechanisms of the emerging non-apoptotic RCD subroutines, mainly including autophagy-dependent cell death, ferroptosis, cuproptosis, disulfidptosis, necroptosis, pyroptosis, alkaliptosis, oxeiptosis, parthanatos, mitochondrial permeability transition (MPT)-driven necrosis, entotic cell death, NETotic cell death, lysosome-dependent cell death, and immunogenic cell death (ICD). Furthermore, we focused on discussing the pharmacological regulatory mechanisms of related small-molecule compounds. In brief, these insightful findings may provide valuable guidance for investigating individual or collaborative targeting approaches towards different RCD subroutines, ultimately driving the discovery of novel small-molecule compounds that target RCD and significantly enhance future cancer therapeutics.
Collapse
Affiliation(s)
- Xin Jin
- Department of Ultrasound, Department of Medical Oncology and Department of Hematology, the First Hospital of China Medical University, China Medical University, Shenyang 110001, China
| | - Wenke Jin
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Linlin Tong
- Department of Ultrasound, Department of Medical Oncology and Department of Hematology, the First Hospital of China Medical University, China Medical University, Shenyang 110001, China
| | - Jia Zhao
- Department of Ultrasound, Department of Medical Oncology and Department of Hematology, the First Hospital of China Medical University, China Medical University, Shenyang 110001, China
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Na Lin
- Department of Ultrasound, Department of Medical Oncology and Department of Hematology, the First Hospital of China Medical University, China Medical University, Shenyang 110001, China
| |
Collapse
|
37
|
Hu S, Wang F, Mao L, Jiang X, Luo Y, Qin X, Zou Z, Chen C, Yu C, Zhang J. NBR1-mediated autophagic degradation of caspase 8 protects vascular endothelial cells against arsenite-induced apoptotic cell death. Biochem Biophys Res Commun 2024; 715:150006. [PMID: 38678786 DOI: 10.1016/j.bbrc.2024.150006] [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: 01/25/2024] [Revised: 04/06/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
Vascular endothelial cells play a critical role in maintaining the health of blood vessels, but dysfunction can lead to cardiovascular diseases. The impact of arsenite exposure on cardiovascular health is a significant concern due to its potential adverse effects. This study aims to explore how NBR1-mediated autophagy in vascular endothelial cells can protect against oxidative stress and apoptosis induced by arsenite. Initially, our observations revealed that arsenite exposure increased oxidative stress and triggered apoptotic cell death in human umbilical vein endothelial cells (HUVECs). However, treatment with the apoptosis inhibitor Z-VAD-FMK notably reduced arsenite-induced apoptosis. Additionally, arsenite activated the autophagy pathway and enhanced autophagic flux in HUVECs. Interestingly, inhibition of autophagy exacerbated arsenite-induced apoptotic cell death. Our findings also demonstrated the importance of autophagy receptor NBR1 in arsenite-induced cytotoxicity, as it facilitated the recruitment of caspase 8 to autophagosomes for degradation. The protective effect of NBR1 against arsenite-induced apoptosis was compromised when autophagy was inhibited using pharmacological inhibitors or through genetic knockdown of essential autophagy genes. Conversely, overexpression of NBR1 facilitated caspase 8 degradation and reduced apoptotic cell death in arsenite-treated HUVECs. In conclusion, our study highlights the vital role of NBR1-mediated autophagic degradation of caspase 8 in safeguarding vascular endothelial cells from arsenite-induced oxidative stress and apoptotic cell death. Targeting this pathway could offer a promising therapeutic approach to mitigate cardiovascular diseases associated with arsenite exposure.
Collapse
Affiliation(s)
- Siyao Hu
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, 400016, Chongqing, People's Republic of China
| | - Fu Wang
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, 400016, Chongqing, People's Republic of China
| | - Lejiao Mao
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, 400016, Chongqing, People's Republic of China
| | - Xuejun Jiang
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, 400016, Chongqing, People's Republic of China
| | - Yilin Luo
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, 400016, Chongqing, People's Republic of China
| | - Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, 400016, Chongqing, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, 400016, Chongqing, People's Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, 400016, Chongqing, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, 400016, Chongqing, People's Republic of China.
| | - Chao Yu
- Chongqing Key Laboratory for Pharmaceutical Metabolism Research, College of Pharmacy, Chongqing Medical University, 400016, Chongqing, People's Republic of China.
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, Institute of Life Sciences, Chongqing Medical University, 400016, Chongqing, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, 400016, Chongqing, People's Republic of China.
| |
Collapse
|
38
|
Thangameeran SIM, Tsai ST, Liew HK, Pang CY. Examining Transcriptomic Alterations in Rat Models of Intracerebral Hemorrhage and Severe Intracerebral Hemorrhage. Biomolecules 2024; 14:678. [PMID: 38927081 PMCID: PMC11202056 DOI: 10.3390/biom14060678] [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: 04/28/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Intracerebral hemorrhage (ICH) is a life-threatening condition associated with significant morbidity and mortality. This study investigates transcriptomic alterations in rodent models of ICH and severe ICH to shed light on the genetic pathways involved in hemorrhagic brain injury. We performed principal component analysis, revealing distinct principal component segments of normal rats compared to ICH and severe ICH rats. We employed heatmaps and volcano plots to identify differentially expressed genes and utilized bar plots and KEGG pathway analysis to elucidate the molecular pathways involved. We identified a multitude of differentially expressed genes in both the ICH and severe ICH models. Our results revealed 5679 common genes among the normal, ICH, and severe ICH groups in the upregulated genes group, and 1196 common genes in the downregulated genes, respectively. A volcano plot comparing these groups further highlighted common genes, including PDPN, TIMP1, SERPINE1, TUBB6, and CD44. These findings underscore the complex interplay of genes involved in inflammation, oxidative stress, and neuronal damage. Furthermore, pathway enrichment analysis uncovered key signaling pathways, including the TNF signaling pathway, protein processing in the endoplasmic reticulum, MAPK signaling pathway, and Fc gamma R-mediated phagocytosis, implicated in the pathogenesis of ICH.
Collapse
Affiliation(s)
| | - Sheng-Tzung Tsai
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan; (S.I.M.T.); (S.-T.T.)
- Neuro-Medical Scientific Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Hock-Kean Liew
- Neuro-Medical Scientific Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
- PhD Program in Pharmacology and Toxicology, Tzu Chi University, Hualien 97004, Taiwan
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Cheng-Yoong Pang
- Institute of Medical Sciences, Tzu Chi University, Hualien 97004, Taiwan; (S.I.M.T.); (S.-T.T.)
- Neuro-Medical Scientific Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| |
Collapse
|
39
|
Beltrán-Visiedo M, Serrano-Del Valle A, Jiménez-Aldúan N, Soler-Agesta R, Naval J, Galluzzi L, Marzo I. Cytofluorometric assessment of calreticulin exposure on CD38 + plasma cells from the human bone marrow. Methods Cell Biol 2024; 189:189-206. [PMID: 39393883 DOI: 10.1016/bs.mcb.2024.05.009] [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: 10/13/2024]
Abstract
Exposure of the endoplasmic reticulum chaperone calreticulin (CALR) on the surface of stressed and dying cells is paramount for their effective engulfment by professional antigen-presenting cells such as dendritic cells (DCs). Importantly, this is required (but not sufficient) for DCs to initiate an adaptive immune response that culminates with an effector phase as well as with the establishment of immunological memory. Conversely, the early exposure of phosphatidylserine (PS) on the outer layer of the plasma membrane is generally associated with the rapid engulfment of stressed and dying cells by tolerogenic macrophages. Supporting the clinical relevance of the CALR exposure pathway, the spontaneous or therapy-driven translocation of CALR to the surface of malignant cells, as well as intracellular biomarkers thereof, have been associated with improved disease outcome in patients affected by a variety of neoplasms, with the notable exception of multiple myeloma (MM). Here, we describe an optimized protocol for the flow cytometry-assisted quantification of surface-exposed CALR and PS on CD38+ plasma cells from the bone marrow of patients with MM. With some variations, we expect this method to be straightforwardly adaptable to the detection of CALR and PS on the surface of cancer cells isolated from patients with neoplasms other than MM.
Collapse
Affiliation(s)
- Manuel Beltrán-Visiedo
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | | | - Nelia Jiménez-Aldúan
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Zaragoza, Spain
| | - Ruth Soler-Agesta
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Zaragoza, Spain
| | - Javier Naval
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Zaragoza, Spain
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| | - Isabel Marzo
- Apoptosis, Immunity & Cancer Group, IIS Aragón, University of Zaragoza, Zaragoza, Spain.
| |
Collapse
|
40
|
Soler-Agesta R, Ripollés-Yuba C, Marco-Brualla J, Moreno-Loshuertos R, Sato A, Beltrán-Visiedo M, Galluzzi L, Anel A. Generation of transmitochondrial cybrids in cancer cells. Methods Cell Biol 2024; 189:23-40. [PMID: 39393884 DOI: 10.1016/bs.mcb.2024.05.010] [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: 10/13/2024]
Abstract
At odds with historical views suggesting that mitochondrial functions are largely dispensable for cancer cells, it is now clear that mitochondria have a major impact on malignant transformation, tumor progression and response to treatment. Mitochondria are indeed critical for neoplastic cells not only as an abundant source of ATP and other metabolic intermediates, but also as gatekeepers of apoptotic cell death and inflammation. Interestingly, while mitochondrial components are mostly encoded by nuclear genes, mitochondria contain a small, circular genome that codes for a few mitochondrial proteins, ribosomal RNAs and transfer RNAs. Here, we describe a straightforward method to generate transmitochondrial cybrids, i.e., cancer cells depleted of their mitochondrial DNA and reconstituted with intact mitochondria from another cellular source. Once established, transmitochondrial cybrids can be stably propagated and are valuable to dissect the specific impact of the mitochondrial genome on cancer cell functions.
Collapse
Affiliation(s)
- Ruth Soler-Agesta
- University of Zaragoza/Aragón Health Research Institute, Biochemistry and Molecular and Cell Biology, Zaragoza, Spain; Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Cristina Ripollés-Yuba
- University of Zaragoza/Aragón Health Research Institute, Biochemistry and Molecular and Cell Biology, Zaragoza, Spain
| | - Joaquín Marco-Brualla
- University of Zaragoza/Aragón Health Research Institute, Biochemistry and Molecular and Cell Biology, Zaragoza, Spain
| | - Raquel Moreno-Loshuertos
- University of Zaragoza/Aragón Health Research Institute, Biochemistry and Molecular and Cell Biology, Zaragoza, Spain
| | - Ai Sato
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Manuel Beltrán-Visiedo
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| | - Alberto Anel
- University of Zaragoza/Aragón Health Research Institute, Biochemistry and Molecular and Cell Biology, Zaragoza, Spain.
| |
Collapse
|
41
|
Mitrović M, Djukić MB, Vukić M, Nikolić I, Radovanović MD, Luković J, Filipović IP, Matić S, Marković T, Klisurić OR, Popović S, Matović ZD, Ristić MS. Search for new biologically active compounds: in vitro studies of antitumor and antimicrobial activity of dirhodium(II,II) paddlewheel complexes. Dalton Trans 2024; 53:9330-9349. [PMID: 38747564 DOI: 10.1039/d4dt01082e] [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: 06/05/2024]
Abstract
Four neutral Rh1-Rh4 complexes of the general formula [Rh2(CH3COO)4L2], where L is an N-alkylimidazole ligand, were synthesized and characterized using various spectroscopic techniques, and in the case of Rh4 the crystal structure was confirmed. Investigation of the interactions of these complexes with HSA by fluorescence spectroscopy revealed that the binding constants Kb are moderately strong (∼104 M-1), and site-marker competition experiments showed that the complexes bind to Heme site III (subdomain IB). Competitive binding studies for CT DNA using EB and HOE showed that the complexes bind to the minor groove, which was also confirmed by viscosity experiments. Molecular docking confirmed the experimental data for HSA and CT DNA. Antimicrobial tests showed that the Rh2-Rh4 complexes exerted a strong inhibitory effect on G+ bacteria B. cereus and G- bacteria V. parahaemolyticus as well as on the yeast C. tropicalis, which showed a higher sensitivity compared to fluconazole. The cytotoxic activity of Rh1-Rh4 complexes tested on three cancer cell lines (HeLa, HCT116 and MDA-MB-231) and on healthy MRC-5 cells showed that all investigated complexes elicited more efficient cytotoxicity on all tested tumor cells than on control cells. Investigation of the mechanism of action revealed that the Rh1-Rh4 complexes inhibit cell proliferation via different mechanisms of action, namely apoptosis (increase in expression of the pro-apoptotic Bax protein and caspase-3 protein in HeLa and HCT116 cells; changes in mitochondrial potential and mitochondrial damage; release of cytochrome c from the mitochondria; cell cycle arrest in G2/M phase in both HeLa and HCT116 cells together with a decrease in the expression of cyclin A and cyclin B) and autophagy (reduction in the expression of the protein p62 in HeLa and HCT116 cells).
Collapse
Affiliation(s)
- Marina Mitrović
- University of Kragujevac, Faculty of Medical Sciences, Department of Medical Biochemistry, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Maja B Djukić
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovića 12, 34000 Kragujevac, Serbia.
| | - Milena Vukić
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovića 12, 34000 Kragujevac, Serbia.
| | - Ivana Nikolić
- University of Kragujevac, Faculty of Medical Sciences, Department of Medical Biochemistry, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Marko D Radovanović
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovića 12, 34000 Kragujevac, Serbia.
| | - Jovan Luković
- University of Kragujevac, Faculty of Medical Sciences, Department of Medical Biochemistry, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Ignjat P Filipović
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovića 12, 34000 Kragujevac, Serbia.
| | - Sanja Matić
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Tijana Marković
- University of Kragujevac, Faculty of Medical Sciences, Department of Pharmacy, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Olivera R Klisurić
- University of Novi Sad, Faculty of Sciences, Department of Physics, Trg Dositeja Obradovića 4, 21000 Novi Sad, Serbia
| | - Suzana Popović
- University of Kragujevac, Faculty of Medical Sciences, Centre for Molecular Medicine and Stem Cell Research, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Zoran D Matović
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovića 12, 34000 Kragujevac, Serbia.
| | - Marija S Ristić
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovića 12, 34000 Kragujevac, Serbia.
| |
Collapse
|
42
|
Dadsena S, Cuevas Arenas R, Vieira G, Brodesser S, Melo MN, García-Sáez AJ. Lipid unsaturation promotes BAX and BAK pore activity during apoptosis. Nat Commun 2024; 15:4700. [PMID: 38830851 PMCID: PMC11148036 DOI: 10.1038/s41467-024-49067-6] [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: 07/07/2023] [Accepted: 05/22/2024] [Indexed: 06/05/2024] Open
Abstract
BAX and BAK are proapoptotic members of the BCL2 family that directly mediate mitochondrial outer membrane permeabilition (MOMP), a central step in apoptosis execution. However, the molecular architecture of the mitochondrial apoptotic pore remains a key open question and especially little is known about the contribution of lipids to MOMP. By performing a comparative lipidomics analysis of the proximal membrane environment of BAK isolated in lipid nanodiscs, we find a significant enrichment of unsaturated species nearby BAK and BAX in apoptotic conditions. We then demonstrate that unsaturated lipids promote BAX pore activity in model membranes, isolated mitochondria and cellular systems, which is further supported by molecular dynamics simulations. Accordingly, the fatty acid desaturase FADS2 not only enhances apoptosis sensitivity, but also the activation of the cGAS/STING pathway downstream mtDNA release. The correlation of FADS2 levels with the sensitization to apoptosis of different lung and kidney cancer cell lines by co-treatment with unsaturated fatty acids supports the relevance of our findings. Altogether, our work provides an insight on how local lipid environment affects BAX and BAK function during apoptosis.
Collapse
Affiliation(s)
- Shashank Dadsena
- Institute for Genetics, CECAD Research Center, University of Cologne, Cologne, Germany
| | - Rodrigo Cuevas Arenas
- Structural Biochemistry, Bijvoet Center for Biomolecular Research, Utrecht University, 3584CG, Utrecht, The Netherlands
| | - Gonçalo Vieira
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Susanne Brodesser
- Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Manuel N Melo
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ana J García-Sáez
- Institute for Genetics, CECAD Research Center, University of Cologne, Cologne, Germany.
- Department of Membrane Dynamics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
| |
Collapse
|
43
|
Wu L, Xue X, He C, Lai Y, Tong L. Cell death‑related molecules and targets in the progression of urolithiasis (Review). Int J Mol Med 2024; 53:52. [PMID: 38666544 PMCID: PMC11090264 DOI: 10.3892/ijmm.2024.5376] [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: 03/05/2024] [Accepted: 04/02/2024] [Indexed: 05/04/2024] Open
Abstract
Urolithiasis is a high‑incidence disease caused by calcium oxalate (mainly), uric acid, calcium phosphate, struvite, apatite, cystine and other stones. The development of kidney stones is closely related to renal tubule cell damage and crystal adhesion and aggregation. Cell death, comprising the core steps of cell damage, can be classified into various types (i.e., apoptosis, ferroptosis, necroptosis and pyroptosis). Different crystal types, concentrations, morphologies and sizes cause tubular cell damage via the regulation of different forms of cell death. Oxidative stress caused by high oxalate or crystal concentrations is considered to be a precursor to a variety of types of cell death. In addition, complex crosstalk exists among numerous signaling pathways and their key molecules in various types of cell death. Urolithiasis is considered a metabolic disorder, and tricarboxylic acid cycle‑related molecules, such as citrate and succinate, are closely related to cell death and the inhibition of stone development. However, a literature review of the associations between kidney stone development, metabolism and various types of cell death is currently lacking, at least to the best of our knowledge. Thus, the present review summarizes the major advances in the understanding of regulated cell death and urolithiasis progression.
Collapse
Affiliation(s)
- Liping Wu
- Department of Pharmacy, Ganzhou People's Hospital, Ganzhou, Jiangxi 341099, P.R. China
| | - Xiaoyan Xue
- Department of Pharmacy, Ganzhou People's Hospital, Ganzhou, Jiangxi 341099, P.R. China
| | - Chengwu He
- Department of Urology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, P.R. China
| | - Yongchang Lai
- Department of Urology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518033, P.R. China
- Department of Pharmaceutical Management, School of Medical Business, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510006, P.R. China
| | - Lingfei Tong
- Department of Pharmacy, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
44
|
Chen X, Tsvetkov AS, Shen HM, Isidoro C, Ktistakis NT, Linkermann A, Koopman WJ, Simon HU, Galluzzi L, Luo S, Xu D, Gu W, Peulen O, Cai Q, Rubinsztein DC, Chi JT, Zhang DD, Li C, Toyokuni S, Liu J, Roh JL, Dai E, Juhasz G, Liu W, Zhang J, Yang M, Liu J, Zhu LQ, Zou W, Piacentini M, Ding WX, Yue Z, Xie Y, Petersen M, Gewirtz DA, Mandell MA, Chu CT, Sinha D, Eftekharpour E, Zhivotovsky B, Besteiro S, Gabrilovich DI, Kim DH, Kagan VE, Bayir H, Chen GC, Ayton S, Lünemann JD, Komatsu M, Krautwald S, Loos B, Baehrecke EH, Wang J, Lane JD, Sadoshima J, Yang WS, Gao M, Münz C, Thumm M, Kampmann M, Yu D, Lipinski MM, Jones JW, Jiang X, Zeh HJ, Kang R, Klionsky DJ, Kroemer G, Tang D. International consensus guidelines for the definition, detection, and interpretation of autophagy-dependent ferroptosis. Autophagy 2024; 20:1213-1246. [PMID: 38442890 PMCID: PMC11210914 DOI: 10.1080/15548627.2024.2319901] [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: 08/25/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 03/07/2024] Open
Abstract
Macroautophagy/autophagy is a complex degradation process with a dual role in cell death that is influenced by the cell types that are involved and the stressors they are exposed to. Ferroptosis is an iron-dependent oxidative form of cell death characterized by unrestricted lipid peroxidation in the context of heterogeneous and plastic mechanisms. Recent studies have shed light on the involvement of specific types of autophagy (e.g. ferritinophagy, lipophagy, and clockophagy) in initiating or executing ferroptotic cell death through the selective degradation of anti-injury proteins or organelles. Conversely, other forms of selective autophagy (e.g. reticulophagy and lysophagy) enhance the cellular defense against ferroptotic damage. Dysregulated autophagy-dependent ferroptosis has implications for a diverse range of pathological conditions. This review aims to present an updated definition of autophagy-dependent ferroptosis, discuss influential substrates and receptors, outline experimental methods, and propose guidelines for interpreting the results.Abbreviation: 3-MA:3-methyladenine; 4HNE: 4-hydroxynonenal; ACD: accidentalcell death; ADF: autophagy-dependentferroptosis; ARE: antioxidant response element; BH2:dihydrobiopterin; BH4: tetrahydrobiopterin; BMDMs: bonemarrow-derived macrophages; CMA: chaperone-mediated autophagy; CQ:chloroquine; DAMPs: danger/damage-associated molecular patterns; EMT,epithelial-mesenchymal transition; EPR: electronparamagnetic resonance; ER, endoplasmic reticulum; FRET: Försterresonance energy transfer; GFP: green fluorescent protein;GSH: glutathione;IF: immunofluorescence; IHC: immunohistochemistry; IOP, intraocularpressure; IRI: ischemia-reperfusion injury; LAA: linoleamide alkyne;MDA: malondialdehyde; PGSK: Phen Green™ SK;RCD: regulatedcell death; PUFAs: polyunsaturated fatty acids; RFP: red fluorescentprotein;ROS: reactive oxygen species; TBA: thiobarbituricacid; TBARS: thiobarbituric acid reactive substances; TEM:transmission electron microscopy.
Collapse
Affiliation(s)
- Xin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Andrey S. Tsvetkov
- Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Han-Ming Shen
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macau, China
| | - Ciro Isidoro
- Department of Health Sciences, University of Piemonte Orientale, Novara, Italy
| | | | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Werner J.H. Koopman
- Department of Pediatrics, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Shouqing Luo
- Peninsula Medical School, University of Plymouth, Plymouth, UK
| | - Daqian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Gu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA Cancer-University of Liège, Liège, Belgium
| | - Qian Cai
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - David C. Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge, UK
| | - Jen-Tsan Chi
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - Donna D. Zhang
- Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shinya Toyokuni
- Department of Pathology and Biological Response, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan
| | - Jinbao Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Enyong Dai
- The Second Department of Hematology and Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Gabor Juhasz
- Biological Research Center, Institute of Genetics, Szeged, Hungary
- Department of Anatomy, Cell and Developmental Biology, Eotvos Lorand University, Budapest, Hungary
| | - Wei Liu
- Department of Orthopedics, Changzheng Hospital, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Minghua Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Research Center of Pediatric Cancer, Changsha, China
| | - Jiao Liu
- DAMP Laboratory, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiping Zou
- Departments of Surgery and Pathology, University of Michigan Medical School, Ann Arbor, USA
| | - Mauro Piacentini
- Department of Biology, University of Rome “Tor Vergata”, Rome, Italy
- National Institute for Infectious Diseases IRCCS “Lazzaro Spallanzani”, Rome, Italy
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Zhenyu Yue
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yangchun Xie
- Department of Oncology, Central South University, Changsha, Hunan, China
| | - Morten Petersen
- Functional genomics, Department of Biology, Copenhagen University, Denmark
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA, USA
| | - Michael A. Mandell
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, USA
| | - Charleen T. Chu
- Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA; Wilmer Eye lnstitute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eftekhar Eftekharpour
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer, Villejuif, France; Gustave Roussy Cancer, Villejuif, France
| | - Boris Zhivotovsky
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden, Europe
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Engelhardt Institute of Molecular Biology, Moscow, Russia
| | - Sébastien Besteiro
- LPHI, University Montpellier, CNRS, Montpellier, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | | | - Do-Hyung Kim
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Valerian E. Kagan
- Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hülya Bayir
- Department of Pediatrics, Columbia University, New York, USA
| | - Guang-Chao Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Scott Ayton
- Florey Institute, University of Melbourne, Parkville, Australia
| | - Jan D. Lünemann
- Department of Neurology with Institute of Translational Neurology, University of Münster, Münster, Germany
| | - Masaaki Komatsu
- Department of Physiology, Juntendo University School of Medicine, Bunkyo-ku Tokyo, Japan
| | - Stefan Krautwald
- Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Eric H. Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jiayi Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Medical Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jon D. Lane
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Junichi Sadoshima
- Rutgers New Jersey Medical School, Department of Cell Biology and Molecular Medicine, Newark, USA
| | - Wan Seok Yang
- Department of Biological Sciences, St. John’s University, New York City, NY, USA
| | - Minghui Gao
- The HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Christian Münz
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Michael Thumm
- Department of Cellular Biochemistry, University Medical Center Goettingen, Goettingen, Germany
| | - Martin Kampmann
- Department of Biochemistry & Biophysics, University of California, San Francisco, USA
- Institute for Neurodegenerative Diseases, University of California, San Francisco, USA
| | - Di Yu
- Faculty of Medicine, Frazer Institute, University of Queensland, Brisbane, Australia
- Faculty of Medicine, Ian Frazer Centre for Children’s Immunotherapy Research, Child Health Research Centre, University of Queensland, Brisbane, Australia
| | - Marta M. Lipinski
- Department of Anesthesiology & Department of Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jace W. Jones
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Herbert J. Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer, Villejuif, France; Gustave Roussy Cancer, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
45
|
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
|
46
|
Galluzzi L, Guilbaud E, Schmidt D, Kroemer G, Marincola FM. Targeting immunogenic cell stress and death for cancer therapy. Nat Rev Drug Discov 2024; 23:445-460. [PMID: 38622310 PMCID: PMC11153000 DOI: 10.1038/s41573-024-00920-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 04/17/2024]
Abstract
Immunogenic cell death (ICD), which results from insufficient cellular adaptation to specific stressors, occupies a central position in the development of novel anticancer treatments. Several therapeutic strategies to elicit ICD - either as standalone approaches or as means to convert immunologically cold tumours that are insensitive to immunotherapy into hot and immunotherapy-sensitive lesions - are being actively pursued. However, the development of ICD-inducing treatments is hindered by various obstacles. Some of these relate to the intrinsic complexity of cancer cell biology, whereas others arise from the use of conventional therapeutic strategies that were developed according to immune-agnostic principles. Moreover, current discovery platforms for the development of novel ICD inducers suffer from limitations that must be addressed to improve bench-to-bedside translational efforts. An improved appreciation of the conceptual difference between key factors that discriminate distinct forms of cell death will assist the design of clinically viable ICD inducers.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| | - Emma Guilbaud
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | | | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | | |
Collapse
|
47
|
Fan L, Hu H. Involvement of multiple forms of cell death in patulin-induced toxicities. Toxicon 2024; 244:107768. [PMID: 38768831 DOI: 10.1016/j.toxicon.2024.107768] [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: 01/22/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
Patulin (PAT) is the most common mycotoxin found in moldy fruits and their derived products, and is reported to cause diverse toxic effects, including hepatotoxicity, nephrotoxicity, cardiotoxicity, neurotoxicity, immunotoxicity, gastrointestinal toxicity and dermal toxicity. The cell death induction by PAT is suggested to be a key cellular mechanism involved in PAT-induced toxicities. Accumulating evidence indicates that the multiple forms of cell death are induced in response to PAT exposure, including apoptosis, autophagic cell death, pyroptosis and ferroptosis. Mechanistically, the cell death induction by PAT is associated the oxidative stress induction via reducing the antioxidant capacity or inducing pro-oxidant NADPH oxidase, the activation of mitochondrial pathway via regulating BCL-2 family proteins, the disruption of iron metabolism through ferritinophagy-mediated ferritin degradation, and the induction of the NOD-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome/caspase-1/gasdermin D (GSDMD) pathway. In this review article, we summarize the present understanding of the cell death induction by PAT, discuss the potential signaling pathways underlying PAT-induced cell death, and propose the issues that need to be addressed to promote the development of cell death-based approach to counteract PAT-induced toxicities.
Collapse
Affiliation(s)
- Lihong Fan
- College of Veterinary Medicine, China Agricultural University, No.2 Yuanmingyuan West Road, Haidian District, Beijing, 100193, China.
| | - Hongbo Hu
- College of Food Science and Nutritional Engineering, China Agricultural University, No.17 Qinghua East Road, Haidian District, Beijing, 100083, China
| |
Collapse
|
48
|
Zhang Z, Hou L, Liu D, Luan S, Huang M, Zhao L. Directly targeting BAX for drug discovery: Therapeutic opportunities and challenges. Acta Pharm Sin B 2024; 14:2378-2401. [PMID: 38828138 PMCID: PMC11143528 DOI: 10.1016/j.apsb.2024.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/25/2024] [Accepted: 02/04/2024] [Indexed: 06/05/2024] Open
Abstract
For over two decades, the development of B-cell lymphoma-2 (Bcl-2) family therapeutics has primarily focused on anti-apoptotic proteins, resulting in the first-in-class drugs called BH3 mimetics, especially for Bcl-2 inhibitor Venetoclax. The pro-apoptotic protein Bcl-2-associated X protein (BAX) plays a crucial role as the executioner protein of the mitochondrial regulated cell death, contributing to organismal development, tissue homeostasis, and immunity. The dysregulation of BAX is closely associated with the onset and progression of diseases characterized by pathologic cell survival or death, such as cancer, neurodegeneration, and heart failure. In addition to conducting thorough investigations into the physiological modulation of BAX, research on the regulatory mechanisms of small molecules identified through biochemical screening approaches has prompted the identification of functional and potentially druggable binding sites on BAX, as well as diverse all-molecule BAX modulators. This review presents recent advancements in elucidating the physiological and pharmacological modulation of BAX and in identifying potentially druggable binding sites on BAX. Furthermore, it highlights the structural and mechanistic insights into small-molecule modulators targeting diverse binding surfaces or conformations of BAX, offering a promising avenue for developing next-generation apoptosis modulators to treat a wide range of diseases associated with dysregulated cell death by directly targeting BAX.
Collapse
Affiliation(s)
- Zhenwei Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linghui Hou
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shenglin Luan
- China Resources Sanjiu Medical & Pharmaceutical Co., Ltd., Shenzhen 518000, China
| | - Min Huang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linxiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
49
|
Mirzayans R. Changing the Landscape of Solid Tumor Therapy from Apoptosis-Promoting to Apoptosis-Inhibiting Strategies. Curr Issues Mol Biol 2024; 46:5379-5396. [PMID: 38920994 PMCID: PMC11202608 DOI: 10.3390/cimb46060322] [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: 04/17/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024] Open
Abstract
The many limitations of implementing anticancer strategies under the term "precision oncology" have been extensively discussed. While some authors propose promising future directions, others are less optimistic and use phrases such as illusion, hype, and false hypotheses. The reality is revealed by practicing clinicians and cancer patients in various online publications, one of which has stated that "in the quest for the next cancer cure, few researchers bother to look back at the graveyard of failed medicines to figure out what went wrong". The message is clear: Novel therapeutic strategies with catchy names (e.g., synthetic "lethality") have not fulfilled their promises despite decades of extensive research and clinical trials. The main purpose of this review is to discuss key challenges in solid tumor therapy that surprisingly continue to be overlooked by the Nomenclature Committee on Cell Death (NCCD) and numerous other authors. These challenges include: The impact of chemotherapy-induced genome chaos (e.g., multinucleation) on resistance and relapse, oncogenic function of caspase 3, cancer cell anastasis (recovery from late stages of apoptosis), and pitfalls of ubiquitously used preclinical chemosensitivity assays (e.g., cell "viability" and tumor growth delay studies in live animals) that score such pro-survival responses as "lethal" events. The studies outlined herein underscore the need for new directions in the management of solid tumors.
Collapse
Affiliation(s)
- Razmik Mirzayans
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| |
Collapse
|
50
|
Franzese O, Ancona P, Bianchi N, Aguiari G. Apoptosis, a Metabolic "Head-to-Head" between Tumor and T Cells: Implications for Immunotherapy. Cells 2024; 13:924. [PMID: 38891056 PMCID: PMC11171541 DOI: 10.3390/cells13110924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/18/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
Induction of apoptosis represents a promising therapeutic approach to drive tumor cells to death. However, this poses challenges due to the intricate nature of cancer biology and the mechanisms employed by cancer cells to survive and escape immune surveillance. Furthermore, molecules released from apoptotic cells and phagocytes in the tumor microenvironment (TME) can facilitate cancer progression and immune evasion. Apoptosis is also a pivotal mechanism in modulating the strength and duration of anti-tumor T-cell responses. Combined strategies including molecular targeting of apoptosis, promoting immunogenic cell death, modulating immunosuppressive cells, and affecting energy pathways can potentially overcome resistance and enhance therapeutic outcomes. Thus, an effective approach for targeting apoptosis within the TME should delicately balance the selective induction of apoptosis in tumor cells, while safeguarding survival, metabolic changes, and functionality of T cells targeting crucial molecular pathways involved in T-cell apoptosis regulation. Enhancing the persistence and effectiveness of T cells may bolster a more resilient and enduring anti-tumor immune response, ultimately advancing therapeutic outcomes in cancer treatment. This review delves into the pivotal topics of this multifaceted issue and suggests drugs and druggable targets for possible combined therapies.
Collapse
Affiliation(s)
- Ornella Franzese
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| | - Pietro Ancona
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy;
| | - Nicoletta Bianchi
- Department of Translational Medicine, University of Ferrara, Via Fossato di Mortara 70, 44121 Ferrara, Italy;
| | - Gianluca Aguiari
- Department of Neuroscience and Rehabilitation, University of Ferrara, Via F. Mortara 74, 44121 Ferrara, Italy;
| |
Collapse
|