1
|
Chen W, Sun M, Sun Y, Yang Q, Gao H, Li L, Fu R, Dong N. Proteasome inhibition induces apoptosis through simultaneous inactivation of MCL-1/BCL-XL by NOXA independent of CHOP and JNK pathways. Toxicology 2024; 508:153906. [PMID: 39117261 DOI: 10.1016/j.tox.2024.153906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Proteasome inhibitors have been employed in the treatment of relapsed multiple myeloma and mantle cell lymphoma. The observed toxicity caused by proteasome inhibitors is a universal phenotype in numerous cancer cells with different sensitivity. In this study, we investigate the conserved mechanisms underlying the toxicity of the proteasome inhibitor bortezomib using gene editing approaches. Our findings utilizing different caspase knocking out cells reveal that bortezomib induces classic intrinsic apoptosis by activating caspase-9 and caspase-3/7, leading to pore-forming protein GSDME cleavage and subsequent lytic cell death or called secondary necrosis, a phenotype also observed in many apoptosis triggers like TNFα plus CHX, DTT and tunicamycin treatment in HeLa cells. Furthermore, through knocking out of nearly all BH3-only proteins including BIM, BAD, BID, BMF and PUMA, we demonstrate that NOXA is the sole BH3-only protein responsible for bortezomib-induced apoptosis. Of note, NOXA is well known for selectively binding to MCL-1 and A1, but our studies utilizing different BH3 mimetics as well as immunoprecipitation assays indicate that, except for the constitutive interaction of NOXA with MCL-1, the accumulation of NOXA after bortezomib treatment allows it to interact with BCL-XL, then simultaneous relieving suppression on apoptosis by both anti-apoptotic proteins BCL-XL and MCL-1. In addition, though bortezomib-induced significant ER stress and JNK activation were observed in the study, further genetic depletion experiments prove that bortezomib-induced apoptosis occurs independently of ER stress-related apoptosis factor CHOP and JNK. In summary, these results provide a solid conclusion about the critical role of NOXA in inactivation of BCL-XL except MCL-1 in bortezomib-induced apoptosis.
Collapse
Affiliation(s)
- Wenjuan Chen
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Mengning Sun
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yi Sun
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qinglan Yang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Hui Gao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Li Li
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Rongrong Fu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Na Dong
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| |
Collapse
|
2
|
Zhang X, Rameika N, Zhong L, Rendo V, Veanes M, Kundu S, Nuciforo S, Dupuis J, Al Azhar M, Tsiara I, Seeburger P, Al Nassralla S, Ljungström V, Svensson R, Stoimenov I, Artursson P, Heim MH, Globisch D, Sjöblom T. Loss of heterozygosity of CYP2D6 enhances the sensitivity of hepatocellular carcinomas to talazoparib. EBioMedicine 2024; 109:105368. [PMID: 39368455 PMCID: PMC11490764 DOI: 10.1016/j.ebiom.2024.105368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 09/10/2024] [Accepted: 09/14/2024] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Loss of heterozygosity (LOH) diminishes genetic diversity within cancer genomes. A tumour arising in an individual heterozygous for a functional and a loss-of-function (LoF) allele of a gene occasionally retain only the LoF allele. This can result in deficiency of specific protein activities in cancer cells, creating unique differences between tumour cells and normal cells of the individual. Such differences may constitute vulnerabilities that can be exploited through allele-specific therapies. METHODS To discover frequently lost genes with prevalent LoF alleles, we mined the 1000 Genomes dataset for SNVs causing protein truncation through base substitution, indels or splice site disruptions, resulting in 60 LoF variants in 60 genes. From these, the variant rs3892097 in the liver enzyme CYP2D6 was selected because it is located within a genomic region that frequently undergoes LOH in several tumor types including hepatocellular cancers. To evaluate the relationship between CYP2D6 activity and the toxicities of anticancer agents, we screened 525 compounds currently in clinical use or undergoing clinical trials using cell model systems with or without CYP2D6 activity. FINDINGS We identified 12 compounds, AZD-3463, CYC-116, etoposide, everolimus, GDC-0349, lenvatinib, MK-8776, PHA-680632, talazoparib, tyrphostin 9, VX-702, and WZ-3146, using an engineered HEK293T cell model. Of these, talazoparib and MK-8776 demonstrated consistently heightened cytotoxic effects against cells with compromised CYP2D6 activity in engineered hepatocellular cancer cell models. Moreover, talazoparib displayed CYP2D6 genotype dependent effects on primary hepatocellular carcinoma organoids. INTERPRETATION Exploiting the loss of drug-metabolizing enzyme gene activity in tumor cells following loss of heterozygosity could present a promising therapeutic strategy for targeted cancer treatment. FUNDING This work was funded by Barncancerfonden (T.S, PR2022-0099 and PR2020-0171, X.Z, TJ2021-0111), Cancerfonden (T.S, 211719Pj and D.G, 222449Pj), Vetenskapsrådet (T.S, 2020-02371 and D.G, 2020-04707), and the Erling Persson Foundation (T.S, 2020-0037 and T.S, 2023-0113).
Collapse
Affiliation(s)
- Xiaonan Zhang
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Natallia Rameika
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Lei Zhong
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Verónica Rendo
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Margus Veanes
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Snehangshu Kundu
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Sandro Nuciforo
- Department of Biomedicine, University Hospital and University of Basel, CH-4031, Basel, Switzerland
| | - Jordan Dupuis
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Muhammad Al Azhar
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Ioanna Tsiara
- Department of Chemistry-BMC, Science for Life Laboratory, Uppsala University, SE-751 23, Uppsala, Sweden
| | - Pauline Seeburger
- Department of Chemistry-BMC, Science for Life Laboratory, Uppsala University, SE-751 23, Uppsala, Sweden
| | - Shahed Al Nassralla
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Viktor Ljungström
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Richard Svensson
- Uppsala Drug Optimization and Pharmaceutical Profiling Facility (UDOPP), SciLifeLab Chemical Biology Consortium Sweden (CBCS), Department of Pharmacy, Uppsala University, 751 23, Uppsala, Sweden; Department of Pharmacy, Uppsala University, Husargatan 3, 751 23, Uppsala, Sweden
| | - Ivaylo Stoimenov
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Husargatan 3, 751 23, Uppsala, Sweden
| | - Markus H Heim
- Department of Biomedicine, University Hospital and University of Basel, CH-4031, Basel, Switzerland; Clarunis University Center for Gastrointestinal and Liver Diseases, CH-4002, Basel, Switzerland
| | - Daniel Globisch
- Department of Chemistry-BMC, Science for Life Laboratory, Uppsala University, SE-751 23, Uppsala, Sweden
| | - Tobias Sjöblom
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden.
| |
Collapse
|
3
|
Li JR, Parthasarathy AK, Kannappan AS, Arsang-Jang S, Dong J, Cheng C. Characterization of driver mutations identifies gene signatures predictive of prognosis and treatment sensitivity in multiple myeloma. Oncologist 2024:oyae244. [PMID: 39250742 DOI: 10.1093/oncolo/oyae244] [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: 04/22/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
In multiple myeloma (MM), while frequent mutations in driver genes are crucial for disease progression, they traditionally offer limited insights into patient prognosis. This study aims to enhance prognostic understanding in MM by analyzing pathway dysregulations in key cancer driver genes, thereby identifying actionable gene signatures. We conducted a detailed quantification of mutations and pathway dysregulations in 10 frequently mutated cancer driver genes in MM to characterize their comprehensive mutational impacts on the whole transcriptome. This was followed by a systematic survival analysis to identify significant gene signatures with enhanced prognostic value. Our systematic analysis highlighted 2 significant signatures, TP53 and LRP1B, which notably outperformed mere mutation status in prognostic predictions. These gene signatures remained prognostically valuable even when accounting for clinical factors, including cytogenetic abnormalities, the International Staging System (ISS), and its revised version (R-ISS). The LRP1B signature effectively distinguished high-risk patients within low/intermediate-risk categories and correlated with significant changes in the tumor immune microenvironment. Additionally, the LRP1B signature showed a strong association with proteasome inhibitor pathways, notably predicting patient responses to bortezomib and the progression from monoclonal gammopathy of unknown significance to MM. Through a rigorous analysis, this study underscores the potential of specific gene signatures in revolutionizing the prognostic landscape of MM, providing novel clinical insights that could influence future translational oncology research.
Collapse
Affiliation(s)
- Jian-Rong Li
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, United States
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX 77030, United States
| | | | | | - Shahram Arsang-Jang
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Jing Dong
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, United States
- Medical College of Wisconsin Cancer Center, Milwaukee, WI 53226, United States
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, United States
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX 77030, United States
- The Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, United States
| |
Collapse
|
4
|
Deng Y, Liu T, Scifo E, Li T, Xie K, Taschler B, Morsy S, Schaaf K, Ehninger A, Bano D, Ehninger D. Analysis of the senescence-associated cell surfaceome reveals potential senotherapeutic targets. Aging Cell 2024:e14312. [PMID: 39228130 DOI: 10.1111/acel.14312] [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: 02/13/2024] [Revised: 07/22/2024] [Accepted: 07/27/2024] [Indexed: 09/05/2024] Open
Abstract
The accumulation of senescent cells is thought to play a crucial role in aging-associated physiological decline and the pathogenesis of various age-related pathologies. Targeting senescence-associated cell surface molecules through immunotherapy emerges as a promising avenue for the selective removal of these cells. Despite its potential, a thorough characterization of senescence-specific surface proteins remains to be achieved. Our study addresses this gap by conducting an extensive analysis of the cell surface proteome, or "surfaceome", in senescent cells, spanning various senescence induction regimes and encompassing both murine and human cell types. Utilizing quantitative mass spectrometry, we investigated enriched cell surface proteins across eight distinct models of senescence. Our results uncover significant changes in surfaceome expression profiles during senescence, highlighting extensive modifications in cell mechanics and extracellular matrix remodeling. Our research also reveals substantive heterogeneity of senescence, predominantly influenced by cell type and senescence inducer. A key discovery of our study is the identification of four unique cell surface proteins with extracellular epitopes. These proteins are expressed in senescent cells, absent or present at low levels in their proliferating counterparts, and notably upregulated in tissues from aged mice and an Alzheimer's disease mouse model. These proteins stand out as promising candidates for senotherapeutic targeting, offering potential pathways for the detection and strategic targeting of senescent cell populations in aging and age-related diseases.
Collapse
Affiliation(s)
- Yushuang Deng
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ting Liu
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Enzo Scifo
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Tao Li
- Department of Neurodegenerative Disease and Geriatric Psychiatry/Neurology, University of Bonn Medical Center, Bonn, Germany
| | - Kan Xie
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Sarah Morsy
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- AvenCell Europe GmbH, Dresden, Germany
| | - Kristina Schaaf
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Daniele Bano
- Aging and Neurodegeneration Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Dan Ehninger
- Translational Biogerontology Lab, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| |
Collapse
|
5
|
Shaaban A, Scott SS, Greenlee AN, Binda N, Noor A, Webb A, Guo S, Purdy N, Pennza N, Habib A, Mohammad SJ, Smith SA. Atrial fibrillation in cancer, anticancer therapies, and underlying mechanisms. J Mol Cell Cardiol 2024; 194:118-132. [PMID: 38897563 PMCID: PMC11500699 DOI: 10.1016/j.yjmcc.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Atrial fibrillation (AF) is a common arrhythmic complication in cancer patients and can be exacerbated by traditional cytotoxic and targeted anticancer therapies. Increased incidence of AF in cancer patients is independent of confounding factors, including preexisting myocardial arrhythmogenic substrates, type of cancer, or cancer stage. Mechanistically, AF is characterized by fast unsynchronized atrial contractions with rapid ventricular response, which impairs ventricular filling and results in various symptoms such as fatigue, chest pain, and shortness of breath. Due to increased blood stasis, a consequence of both cancer and AF, concern for stroke increases in this patient population. To compound matters, cardiotoxic anticancer therapies themselves promote AF; thereby exacerbating AF morbidity and mortality in cancer patients. In this review, we examine the relationship between AF, cancer, and cardiotoxic anticancer therapies with a focus on the shared molecular and electrophysiological mechanisms linking these disease processes. We also explore the potential role of sodium-glucose co-transporter 2 inhibitors (SGLT2i) in the management of anticancer-therapy-induced AF.
Collapse
Affiliation(s)
- Adnan Shaaban
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA
| | - Shane S Scott
- Medical Scientist Training Program, Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Ashley N Greenlee
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Nkongho Binda
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA
| | - Ali Noor
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Averie Webb
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Shuliang Guo
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Najhee Purdy
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Nicholas Pennza
- Ohio University Heritage College of Osteopathic Medicine, Athens, OH 45701, USA
| | - Alma Habib
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA
| | - Somayya J Mohammad
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Sakima A Smith
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
6
|
Xu P, Yang JC, Chen B, Ning S, Zhang X, Wang L, Nip C, Shen Y, Johnson OT, Grigorean G, Phinney B, Liu L, Wei Q, Corey E, Tepper CG, Chen HW, Evans CP, Dall'Era MA, Gao AC, Gestwicki JE, Liu C. Proteostasis perturbation of N-Myc leveraging HSP70 mediated protein turnover improves treatment of neuroendocrine prostate cancer. Nat Commun 2024; 15:6626. [PMID: 39103353 PMCID: PMC11300456 DOI: 10.1038/s41467-024-50459-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 07/11/2024] [Indexed: 08/07/2024] Open
Abstract
N-Myc is a key driver of neuroblastoma and neuroendocrine prostate cancer (NEPC). One potential way to circumvent the challenge of undruggable N-Myc is to target the protein homeostasis (proteostasis) system that maintains N-Myc levels. Here, we identify heat shock protein 70 (HSP70) as a top partner of N-Myc, which binds a conserved "SELILKR" motif and prevents the access of E3 ubiquitin ligase, STIP1 homology and U-box containing protein 1 (STUB1), possibly through steric hindrance. When HSP70's dwell time on N-Myc is increased by treatment with the HSP70 allosteric inhibitor, STUB1 is in close proximity with N-Myc and becomes functional to promote N-Myc ubiquitination on the K416 and K419 sites and forms polyubiquitination chains linked by the K11 and K63 sites. Notably, HSP70 inhibition significantly suppressed NEPC tumor growth, increased the efficacy of aurora kinase A (AURKA) inhibitors, and limited the expression of neuroendocrine-related pathways.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Joy C Yang
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Bo Chen
- Department of Urologic Surgery, University of California, Davis, CA, USA
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Shu Ning
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Xiong Zhang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA
| | - Leyi Wang
- Department of Urologic Surgery, University of California, Davis, CA, USA
- Graduate Group in Integrative Pathobiology, University of California, Davis, CA, USA
| | - Christopher Nip
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Yuqiu Shen
- Department of Urologic Surgery, University of California, Davis, CA, USA
| | - Oleta T Johnson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | | | - Brett Phinney
- Proteomics Core Facility, University of California, Davis, CA, USA
| | - Liangren Liu
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Qiang Wei
- Department of Urology, West China Hospital, Sichuan University, Sichuan, China
| | - Eva Corey
- Department of Urology, University of Washington, Washington, WA, USA
| | - Clifford G Tepper
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Hong-Wu Chen
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Christopher P Evans
- Department of Urologic Surgery, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Marc A Dall'Era
- Department of Urologic Surgery, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Allen C Gao
- Department of Urologic Surgery, University of California, Davis, CA, USA
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Chengfei Liu
- Department of Urologic Surgery, University of California, Davis, CA, USA.
- Graduate Group in Integrative Pathobiology, University of California, Davis, CA, USA.
- University of California, Davis Comprehensive Cancer Center, Sacramento, CA, USA.
| |
Collapse
|
7
|
INANAGA S, SHIMODA H, IGASE M, KONDO H, KOIZUMI I, MIZUNO T. Establishment of a histiocytic sarcoma cell line and anti-tumor effect of bortezomib in the African pygmy hedgehog (Atelerix albiventris). J Vet Med Sci 2024; 86:833-840. [PMID: 38880614 PMCID: PMC11300126 DOI: 10.1292/jvms.23-0426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 06/02/2024] [Indexed: 06/18/2024] Open
Abstract
The African pygmy hedgehog (Atelerix albiventris) is known to have a high incidence of tumor. However, investigating the tumors of this species has been constrained by the limited availability of research materials such as cell lines and genome information. In this study, we successfully established a novel cell line from a histiocytic sarcoma (HS) of an African pygmy hedgehog, allowing us to conduct a drug screening. We investigated using FDA-approved drug library screening to determine which anticancer drug this tumor cell line is sensitive to, and as a result of apoptosis experiments, bortezomib among the three proteasome inhibitors was found to induce cell death of cancer cells by significantly increasing caspase-3 cleavage (P<0.01). Thus, we elucidated that the proteasome inhibitors, particularly bortezomib, exhibit anti-tumor effects on a cell line derived from an HS in an African pygmy hedgehog through a mechanism comparable to that described in human tumors. This study reports the first characterized cell line from the African pygmy hedgehog and also highlights the potential utility of bortezomib as an anti-tumor treatment for HS in this species.
Collapse
Affiliation(s)
- Sakuya INANAGA
- Laboratory of Molecular Diagnostics and Therapeutics, The
United Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi,
Japan
| | - Hiroshi SHIMODA
- Laboratory of Veterinary Microbiology, The United Graduate
School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Masaya IGASE
- Laboratory of Molecular Diagnostics and Therapeutics, The
United Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi,
Japan
| | - Hirotaka KONDO
- Laboratory of Veterinary Pathology, Department of Veterinary
Medicine, Nihon University, Kanagawa, Japan
| | - Iori KOIZUMI
- Laboratory of Veterinary Microbiology, The United Graduate
School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
- Koizumi Nest Animal Hospital, Fukuoka, Japan
| | - Takuya MIZUNO
- Laboratory of Molecular Diagnostics and Therapeutics, The
United Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi,
Japan
| |
Collapse
|
8
|
Sun K, Yang L, Wang F, Liu Y, Xu N, Shi ZY, Chen WM, Li K, Qin YZ. PRAME promotes proliferation of multiple myeloma cells through CTMP/Akt/p21/CCND3 axis by ubiquitinating CTMP and p21. Heliyon 2024; 10:e34094. [PMID: 39071619 PMCID: PMC11283035 DOI: 10.1016/j.heliyon.2024.e34094] [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: 10/24/2023] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
Multiple myeloma (MM) is a Ubiquitin Proteasome System (UPS)-dysfunction disease. We previously reported that high PRAME transcript levels associated with unfavorable progression free survival (PFS) in patients with no bortezomib therapy, and bortezomib-containing regimen significantly improved PFS in patients with high PRAME transcript levels, which indicated that PRAME expression was prognostic for MM patients, and was related to proteasome inhibitor treatment. However, molecular mechanisms underlying the above clinical performance remain unclear. In the present study, MM cell models with PRAME knockdown and overexpression were established, and PRAME was identified to play the role of promoting proliferation in MM cells. P-Akt signaling was found to be activated as PRAME overexpressed. As a substrate recognizing subunit (SRS) of the E3 ubiquitin ligase, PRAME targets substrate proteins and mediates their degradation. CTMP and p21 were found to be the novel targets of PRAME in the Cul2-dependent substrate recognition process. PRAME interacted with and mediated ubiquitination and degradation of CTMP and p21, which led to accumulation of p-Akt and CCND3 proteins, and thus promoted cell proliferation and increased bortezomib sensitivity in MM cells.
Collapse
Affiliation(s)
- Kai Sun
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, PR China
| | - Lu Yang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, PR China
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Feng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Ying Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Nan Xu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, PR China
| | - Zong-Yan Shi
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, PR China
| | - Wen-Min Chen
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, PR China
| | - Ke Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, PR China
| | - Ya-Zhen Qin
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, 100044, PR China
| |
Collapse
|
9
|
Jagtap YA, Kumar P, Dubey AR, Kinger S, Choudhary A, Karmakar S, Lal G, Kumar A, Kumar A, Prasad A, Mishra A. Acetaminophen induces mitochondrial apoptosis through proteasome dysfunctions. Life Sci 2024; 349:122732. [PMID: 38768775 DOI: 10.1016/j.lfs.2024.122732] [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: 11/15/2023] [Revised: 03/12/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
Acetaminophen is a known antipyretic and non-opioid analgesic for mild pain and fever. Numerous studies uncover their hidden chemotherapeutics applications, including chronic cancer pain management. Acetaminophen also represents an anti-proliferative effect in some cancer cells. Few studies also suggest that the use of Acetaminophen can trigger apoptosis and impede cellular growth. However, Acetaminophen's molecular potential and precise mechanism against improper cellular proliferation and use as an effective anti-proliferative agent still need to be better understood. Here, our current findings show that Acetaminophen induces proteasomal dysfunctions, resulting in aberrant protein accumulation and mitochondrial abnormalities, and consequently induces cell apoptosis. We observed that the Acetaminophen treatment leads to improper aggregation of ubiquitylated expanded polyglutamine proteins, which may be due to the dysfunctions of proteasome activities. Our in-silico analysis suggests the interaction of Acetaminophen and proteasome. Furthermore, we demonstrated the accumulation of proteasome substrates and the depletion of proteasome activities after treating Acetaminophen in cells. Acetaminophen induces proteasome dysfunctions and mitochondrial abnormalities, leading to pro-apoptotic morphological changes and apoptosis successively. These results suggest that Acetaminophen can induce cell death and may retain a promising anti-proliferative effect. These observations can open new possible molecular strategies in the near future for developing and designing specific and effective proteasome inhibitors, which can be helpful in conjugation with other anti-tumor drugs for their better efficiency.
Collapse
Affiliation(s)
- Yuvraj Anandrao Jagtap
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, 342037, India
| | - Prashant Kumar
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, 342037, India
| | - Ankur Rakesh Dubey
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, 342037, India
| | - Sumit Kinger
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, 342037, India
| | - Akash Choudhary
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, 342037, India
| | - Surojit Karmakar
- National Centre for Cell Science (NCCS), Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Girdhari Lal
- National Centre for Cell Science (NCCS), Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, Chhattisgarh, 492010, India
| | - Amit Kumar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Amit Prasad
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, 175005, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, 342037, India.
| |
Collapse
|
10
|
He H, Zhang X, Wang J, Liu Q, Zhang L, Chen L, Yuan Y, Zhao Z, Li H, Chen Z. Development of Degraders of Cyclin-Dependent Kinases 4 and 6 Based on Rational Drug Design. J Med Chem 2024; 67:11354-11364. [PMID: 38943626 DOI: 10.1021/acs.jmedchem.4c00965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2024]
Abstract
Degradation of target proteins has been considered to be a promising therapeutic approach, but the rational design of compounds for degradation remains a challenge. In this study, we reasonably designed and synthesized only 10 compounds to discover effective CDK4/6 protein degraders. Among the newly synthesized compounds, 7f achieved dual degradation of CDK4/6 protein, with DC50 values of 10.5 and 2.5 nM, respectively. Compound 7f also exhibited inhibitory proliferative activity against Jurkat cells with an IC50 value of 0.18 μM. Furthermore, 7f induced cell apoptosis and G1 phase cell cycle arrest in a dose-dependent manner in Jurkat cells. In conclusion, these findings demonstrate the potential of 7f as a CDK4/6 degrader and a potential therapeutic strategy against cancer, thereby expanding the potential of CDK4/6 dual PROTACs.
Collapse
Affiliation(s)
- Huan He
- Innovation Center for AI and Drug Discovery (ICAIDD), East China Normal University, Shanghai 200062, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xingsen Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jie Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Qi Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - LeiHao Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lu Chen
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yuan Yuan
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zhenjiang Zhao
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Honglin Li
- Innovation Center for AI and Drug Discovery (ICAIDD), East China Normal University, Shanghai 200062, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Lingang Laboratory, Shanghai 200031, China
| | - Zhuo Chen
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
11
|
Guo Q, Jin Y, Lin M, Zeng C, Zhang J. NF-κB signaling in therapy resistance of breast cancer: Mechanisms, approaches, and challenges. Life Sci 2024; 348:122684. [PMID: 38710275 DOI: 10.1016/j.lfs.2024.122684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024]
Abstract
Breast cancer is the most common type of cancer and is the second leading cause of cancer-related mortality in women. Chemotherapy, targeted therapy, endocrine therapy, and radiotherapy are all effective in destroying tumor cells, but they also activate the defense and protection systems of cancer cells, leading to treatment resistance. Breast cancer is characterized by a highly inflammatory tumor microenvironment. The NF-κB pathway is essential for connecting inflammation and cancer, as well as for tumor growth and therapy resistance. An increase in NF-κB signaling boosts the growth potential of breast cancer cells and facilitates the spread of tumors to bone, lymph nodes, lungs, and liver. This review focuses on the mechanisms by which chemotherapy, targeted therapy, endocrine therapy, and radiotherapy induce breast cancer resistance through NF-κB signaling. Additionally, we investigate therapeutic regimens, including single agents or in combination with target inhibitors, plant extracts, nanomedicines, and miRNAs, that have been reported in clinical trials, in vivo, and in vitro to reverse resistance. In particular, NF-κB inhibitors combined with tamoxifen were shown to significantly increase the sensitivity of breast cancer cells to tamoxifen. Combination therapy of miRNA-34a with doxorubicin was also found to synergistically inhibit the progression of doxorubicin-resistant breast cancer by inhibiting Notch/NF-κB signaling.
Collapse
Affiliation(s)
- Qing Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yizi Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mingxi Lin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cheng Zeng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Park M, Kim JW. Updates on the mechanisms of toxicities associated with monoclonal antibodies targeting growth factor signaling and immune cells in cancer. Toxicol Res 2024; 40:335-348. [PMID: 38911540 PMCID: PMC11187026 DOI: 10.1007/s43188-024-00233-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/12/2024] [Accepted: 03/15/2024] [Indexed: 06/25/2024] Open
Abstract
Monoclonal antibody (mAb)-based immunotherapy currently is considered to be an optimal therapeutic approach to cancer treatment, either in combination with surgery, radiation, and/or chemotherapy or alone. Various solid tumors and hematological malignancies have been characterized by distinct molecular targets, which could be utilized as innovative anticancer agents. Notably, receptor tyrosine kinases, including HER2, EGFR, VEGFR, and PDGFR, which act as receptors for growth factors, serve as crucial target proteins, expanding their role in the cancer therapeutic market. In contrast to conventional anticancer agents that directly target cancer cells, the advent of immunotherapy introduces novel approaches, such as immune checkpoint blockers (ICBs) and mAbs targeting surface antigens on immune cells in hematological malignancies and lymphomas. While these immunotherapies have mitigated the acquired resistance observed in traditional targeted therapies, they also exhibit diverse toxicities. Herein, this review focuses on describing the well-established toxicities and newly proposed mechanisms of monoclonal antibody toxicity in recent studies. Understanding these molecular mechanisms is indispensable to overcoming the limitations of mAbs-based therapies, facilitating the development of innovative anticancer agents, and uncovering novel indications for cancer treatment in the future.
Collapse
Affiliation(s)
- Miso Park
- College of Pharmacy, Kangwon National University, Chuncheon, Gangwon-do Republic of Korea
| | - Ji Won Kim
- Jeju Research Institute of Pharmaceutical Sciences, College of Pharmacy, Jeju National University, Jeju-do, Republic of Korea
| |
Collapse
|
13
|
Liu X, Wang M, Wang Q, Zhang H. A ubiquitin-proteasome system-related signature to predict prognosis, immune infiltration, and therapy efficacy for breast cancer. Immunol Res 2024; 72:368-382. [PMID: 38036900 DOI: 10.1007/s12026-023-09440-x] [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: 06/21/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023]
Abstract
The ubiquitin-proteasome system (UPS) is an essential regulatory system for maintaining homeostasis, and its dysfunction may cause various diseases. The activity of proteasome and ubiquitin-conjugating enzymes has been found to be greatly increased in breast cancer (BC), indicating that the heterogeneity of UPS may be related to the progression of BC. Gene data was obtained from The Cancer Genome Atlas and Gene Expression Omnibus databases and performed in multiple algorithms to construct a UPS-related signature for BC. Patients in the UPS low-risk group had greater overall and recurrence-free survival probability than those in the UPS high-risk group. This signature was closely associated with functional enrichment. Some high metabolism-related pathways were more active in the UPS high-risk group. The UPS low-risk group had more abundant anti-tumor immune cells, while in the UPS high-risk group, immunosuppressive cells were dominant. More importantly, we found that the UPS low-risk group was more sensitive to immunotherapy, while the UPS high-risk group responded better to radiotherapy. Drug sensitivity analysis identified more effective chemotherapy drugs in different UPS-related risk groups. This UPS-related signature may serve as a novel biomarker and independent prognostic factor for BC. It can effectively predict prognosis, immune infiltration, and therapy efficacy, providing new strategies for individualized treatment.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Meihuan Wang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qian Wang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Huawei Zhang
- Department of Ultrasound, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
14
|
Sogabe K, Nakamura S, Higa Y, Miki H, Oda A, Maruhashi T, Sumitani R, Oura M, Takahashi M, Nakamura M, Maeda Y, Hara T, Yamagami H, Fujii S, Kagawa K, Ozaki S, Kurahashi K, Endo I, Aihara KI, Nakaue E, Hiasa M, Teramachi J, Harada T, Abe M. Acute accumulation of PIM2 and NRF2 and recovery of β5 subunit activity mitigate multiple myeloma cell susceptibility to proteasome inhibitors. Int J Hematol 2024; 119:303-315. [PMID: 38245883 DOI: 10.1007/s12185-023-03705-9] [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] [Received: 08/12/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
Resistance to proteasome inhibitors (PIs) has emerged as an important clinical issue. We investigated the mechanisms underlying multiple myeloma (MM) cell resistance to PIs. To mimic their pharmacokinetic/pharmacodynamic (PK/PD) profiles, MM cells were treated with bortezomib and carfilzomib for 1 h at concentrations up to 400 and 1,000 nM, respectively. Susceptibility to these PIs markedly varied among MM cell lines. Pulsatile treatments with PIs suppressed translation, as demonstrated by incorporation of puromycin at 24 h in PI-susceptible MM.1S cells, but not PI-resistant KMS-11 cells. Inhibition of β5 subunit activity decreased at 24 h in KMS-11 cells, even with the irreversible PI carfilzomib, but not under suppression of protein synthesis with cycloheximide. Furthermore, the proteasome-degradable pro-survival factors PIM2 and NRF2 acutely accumulated in MM cells subjected to pulsatile PI treatments. Accumulated NRF2 was trans-localized into the nucleus to induce the expression of its target gene, HMOX1, in MM cells. PIM and Akt inhibition restored the anti-MM effects of PIs, even against PI-resistant KMS-11 cells. Collectively, these results suggest that increased synthesis of β5 proteasome subunit and acute accumulation of PIM2 and NRF2 reduce the anti-MM effects of PIs.
Collapse
Affiliation(s)
- Kimiko Sogabe
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Shingen Nakamura
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-Cho, Tokushima, 770-8503, Japan.
| | - Yoshiki Higa
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hirokazu Miki
- Division of Transfusion Medicine and Cell Therapy, Tokushima University Hospital, Tokushima, Japan
| | - Asuka Oda
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Tomoko Maruhashi
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Ryohei Sumitani
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masahiro Oura
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Mamiko Takahashi
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masafumi Nakamura
- Department of Internal Medicine, Tokushima Prefecture Naruto Hospital, Tokushima, Japan
| | - Yusaku Maeda
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Tomoyo Hara
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Hiroki Yamagami
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Shiro Fujii
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kumiko Kagawa
- Department of Hematology, Tokushima Prefectural Central Hospital, Tokushima, Japan
| | - Shuji Ozaki
- Department of Hematology, Tokushima Prefectural Central Hospital, Tokushima, Japan
| | - Kiyoe Kurahashi
- Department of Community Medicine for Respirology, Hematology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Itsuro Endo
- Department of Bioregulatory Sciences, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Ken-Ichi Aihara
- Department of Community Medicine and Medical Science, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-Cho, Tokushima, 770-8503, Japan
| | - Emiko Nakaue
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masahiro Hiasa
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Jumpei Teramachi
- Department of Oral Function and Anatomy, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takeshi Harada
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masahiro Abe
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.
- Department of Hematology, Kawashima Hospital, 6-1 Kitasakoichiban-Cho, Tokushima, 770-0011, Japan.
| |
Collapse
|
15
|
Pakjoo M, Ahmadi SE, Zahedi M, Jaafari N, Khademi R, Amini A, Safa M. Interplay between proteasome inhibitors and NF-κB pathway in leukemia and lymphoma: a comprehensive review on challenges ahead of proteasome inhibitors. Cell Commun Signal 2024; 22:105. [PMID: 38331801 PMCID: PMC10851565 DOI: 10.1186/s12964-023-01433-5] [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: 08/13/2023] [Accepted: 12/11/2023] [Indexed: 02/10/2024] Open
Abstract
The current scientific literature has extensively explored the potential role of proteasome inhibitors (PIs) in the NF-κB pathway of leukemia and lymphoma. The ubiquitin-proteasome system (UPS) is a critical component in regulating protein degradation in eukaryotic cells. PIs, such as BTZ, are used to target the 26S proteasome in hematologic malignancies, resulting in the prevention of the degradation of tumor suppressor proteins, the activation of intrinsic mitochondrial-dependent cell death, and the inhibition of the NF-κB signaling pathway. NF-κB is a transcription factor that plays a critical role in the regulation of apoptosis, cell proliferation, differentiation, inflammation, angiogenesis, and tumor migration. Despite the successful use of PIs in various hematologic malignancies, there are limitations such as resistant to these inhibitors. Some reports suggest that PIs can induce NF-κB activation, which increases the survival of malignant cells. This article discusses the various aspects of PIs' effects on the NF-κB pathway and their limitations. Video Abstract.
Collapse
Affiliation(s)
- Mahdi Pakjoo
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- ATMP department, Breast cancer research center, Motamed cancer institute, ACECR, P.O. BOX:15179/64311, Tehran, Iran
| | - Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Zahedi
- Department of Medical Biotechnology, School of Allied Medicine, Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Jaafari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reyhane Khademi
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Amini
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Li Y, Liu X, Zhao F, Zhao Z, Li X, Wang J, Huang B, Chen A. Comprehensive analysis of PSMD family members and validation of PSMD9 as a potential therapeutic target in human glioblastoma. CNS Neurosci Ther 2024; 30:e14366. [PMID: 37485655 PMCID: PMC10848081 DOI: 10.1111/cns.14366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/24/2023] [Accepted: 07/02/2023] [Indexed: 07/25/2023] Open
Abstract
AIMS PSMD family members, as important components of the 26S proteasome, are well known to be involved in protein degradation. However, their role in glioblastoma (GBM) has not been rigorously investigated. We aimed to perform systematic analysis of the expression signature, prognostic significance and functions of PSMD family genes in GBM to reveal potential prognostic markers and new therapeutic targets among PSMD family members. METHODS In this study, we systemically analyzed PSMD family members in terms of their expression profiles, prognostic implications, DNA methylation levels, and genetic alterations; the relationships between their expression levels and immune infiltration and drug sensitivity; and their potential functional enrichment in GBM through bioinformatics assessment. Moreover, in vitro and in vivo experiments were used to validate the biological functions of PSMD9 and its targeted therapeutic effect in GBM. RESULTS The mRNA levels of PSMD5/8/9/10/11/13/14 were higher in GBM than in normal brain tissues, and the mRNA levels of PSMD1/4/5/8/9/11/12 were higher in high-grade glioma (WHO grade III & IV) than in low-grade glioma (WHO grade II). High mRNA expression of PSMD2/6/8/9/12/13/14 and low mRNA expression of PSMD7 were associated with poor overall survival (OS). Multivariate Cox regression analysis identified PSMD2/5/6/8/9/10/11/12 as independent prognostic factors for OS prediction. In addition, the protein-protein interaction network and gene set enrichment analysis results suggested that PSMD family members and their interacting molecules were involved in the regulation of the cell cycle, cell invasion and migration, and other biological processes in GBM. In addition, knockdown of PSMD9 inhibited cell proliferation, invasion and migration and induced G2/M cell cycle arrest in LN229 and A172 GBM cells. Moreover, PSMD9 promoted the malignant progression of GBM in vivo. GBM cell lines with high PSMD9 expression were more resistant to panobinostat, a potent deacetylase inhibitor, than those with low PSMD9 expression. In vitro and in vivo experiments further validated that PSMD9 overexpression rescued the GBM inhibitory effect of panobinostat. CONCLUSION This study provides new insights into the value of the PSMD family in human GBM diagnosis and prognosis evaluation, and we further identified PSMD9 as a potential therapeutic target. These findings may lead to the development of effective therapeutic strategies for GBM.
Collapse
Affiliation(s)
- Yaquan Li
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| | - Xuemeng Liu
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| | - Feihu Zhao
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| | - Zhimin Zhao
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| | - Xingang Li
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| | - Jian Wang
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
- Department of BiomedicineUniversity of BergenBergenNorway
| | - Bin Huang
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| | - Anjing Chen
- Department of NeurosurgeryQilu HospitalCheeloo College of Medicine and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Jinan Microecological Biomedicine Shandong Laboratory and Shandong Key Laboratory of Brain Function RemodelingJinanChina
| |
Collapse
|
17
|
Giovannuzzi S, Nikitjuka A, Pereira Resende BR, Smietana M, Nocentini A, Supuran CT, Winum JY. Boron-containing carbonic anhydrases inhibitors. Bioorg Chem 2024; 143:106976. [PMID: 38000350 DOI: 10.1016/j.bioorg.2023.106976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/24/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023]
Abstract
Over the last decades, the medicinal chemistry of boron-based compounds has been extensively explored, designing valuable small molecule drugs to tackle diseases and conditions, such as cancer, infections, inflammatory and neurological disorders. Notably, boron has proven to also be a valuable element for the development of inhibitors of the metalloenzymes carbonic anhydrases (CAs), a class of drug targets with significant potential in medicinal chemistry. Incorporating boron into carbonic anhydrase inhibitors (CAIs) can modulate the ligand ability to recognize the target and/or influence selectivity towards different CA isoforms, using the tail approach and boron-based tails. The electron-deficient nature of boron and its associated properties have also led to the discovery of novel zinc-binding CAIs, such as boronic acids and the benzoxaboroles, capable of inhibiting the CAs upon a Lewis acid-base mechanism of action. The present manuscript reviews the state-of-the-art of boron-based CAIs. As research in the applications of boron compounds in medicinal chemistry continues, it is anticipated that new boron-based CAIs will soon expand the current array of such compounds. However, further research is imperative to fully unlock the potential of boron-based CAIs and to advance them towards clinical applications.
Collapse
Affiliation(s)
- Simone Giovannuzzi
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France; Neurofarba Department, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| | - Anna Nikitjuka
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France; Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Bruna Rafaela Pereira Resende
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France; Neurofarba Department, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| | | | - Alessio Nocentini
- Neurofarba Department, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy.
| | - Claudiu T Supuran
- Neurofarba Department, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| | | |
Collapse
|
18
|
Ali EMH, Loy CA, Trader DJ. ByeTAC: Bypassing an E3 Ligase for Targeted Protein Degradation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.20.576376. [PMID: 38293213 PMCID: PMC10827187 DOI: 10.1101/2024.01.20.576376] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Targeted protein degradation utilizing a bifunctional molecule to initiate ubiquitination and subsequent degradation by the 26S proteasome has been shown to be a powerful therapeutic intervention. Many bifunctional molecules, including covalent and non-covalent ligands to proteins of interest, have been developed. The traditional target protein degradation methodology targets the protein of interest in both healthy and diseased cell populations, and a therapeutic window is obtained based on the overexpression of the targeted protein. We report here a series of bifunctional degraders that do not rely on interacting with an E3 ligase, but rather a 26S proteasome subunit, which we have named ByeTACs: Bypassing E3 Targeting Chimeras. Rpn-13 is a non-essential ubiquitin receptor for the 26S proteasome. Cells under significant stress or require significant ubiquitin-dependent degradation of proteins for survival, incorporate Rpn-13 in the 26S to increase protein degradation rates. The targeted protein degraders reported here are bifunctional molecules that include a ligand to Rpn-13 and BRD4, the protein of interest we wish to degrade. We synthesized a suite of degraders with varying PEG chain lengths and showed that bifunctional molecules that incorporate a Rpn-13 binder (TCL1) and a BRD4 binder (JQ1) with a PEG linker of 3 or 4 units are the most effective to induce BRD4 degradation. We also demonstrate that our new targeted protein degraders are dependent upon proteasome activity and Rpn-13 expression levels. This establishes a new mechanism of action for our ByeTACs that can be employed for the targeted degradation of a wide variety of protein substrates.
Collapse
|
19
|
Bendellaa M, Lelièvre P, Coll JL, Sancey L, Deniaud A, Busser B. Roles of zinc in cancers: From altered metabolism to therapeutic applications. Int J Cancer 2024; 154:7-20. [PMID: 37610131 DOI: 10.1002/ijc.34679] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/10/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023]
Abstract
Zinc (Zn) is a crucial trace element involved in various cellular processes, including oxidative stress, apoptosis and immune response, contributing to cellular homeostasis. Dysregulation of Zn homeostasis occurs in certain cancers. This review discusses the role of Zn in cancer and its associated components, such as Zn-related proteins, their potential as biomarkers and the use of Zn-based strategies for tumor treatment. ZIP and ZnT proteins regulate Zn metabolism under normal conditions, but their expression is aberrant in cancer. These Zn proteins can serve as prognostic or diagnostic biomarkers, aiding in early cancer detection and disease monitoring. Moreover, targeting Zn and its pathways offers potential therapeutic approaches for cancer treatment. Modulating Zn biodistribution within cells using metal-binding agents allows for the control of downstream signaling pathways. Direct utilization of zinc as a therapeutic agent, including Zn supplementation or Zn oxide nanoparticle administration, holds promise for improving the prognosis of cancer patients.
Collapse
Affiliation(s)
- Mohamed Bendellaa
- Grenoble Alpes University, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble, France
| | - Pierre Lelièvre
- Grenoble Alpes University, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble, France
| | - Jean-Luc Coll
- Grenoble Alpes University, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble, France
| | - Lucie Sancey
- Grenoble Alpes University, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble, France
| | - Aurélien Deniaud
- Grenoble Alpes University, CNRS, CEA, IRIG, Laboratoire de Chimie et Biologie des Métaux, Grenoble, France
| | - Benoit Busser
- Grenoble Alpes University, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Grenoble, France
- Department of Laboratory Medicine, Grenoble Alpes University Hospital, Grenoble, France
- Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
20
|
Chen X, Liu G, Wu B. Analysis and experimental validation of the innate immune gene PSMD1 in liver hepatocellular carcinoma and pan-cancer. Heliyon 2023; 9:e21164. [PMID: 37928041 PMCID: PMC10623288 DOI: 10.1016/j.heliyon.2023.e21164] [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: 05/23/2023] [Revised: 09/09/2023] [Accepted: 10/17/2023] [Indexed: 11/07/2023] Open
Abstract
This work intends to examine the diagnostic, prognostic, and biological roles of PSMD1 (proteasome 26S subunit, non-ATPase 1) in liver hepatocellular carcinoma (LIHC) and other malignancies, using bioinformatics techniques. PSMD1 is an innate immune gene that has been identified as a biomarker for several cancers. By analyzing TCGA data, we determined that PSMD1 has excellent diagnostic and prognostic value in LIHC. We also examined its correlation with stage-matching clinical features, particularly T staging and stage staging. Independent prognostic analysis, nomogram, and Decision Curve Analysis (DCA) analysis confirmed the predictive ability of PSMD1 on patient clinical outcomes. Our focus was on exploring the biological process, immune infiltration, and genetic variation in which PSMD1 is involved in LIHC. We found a close relationship between PSMD1 and the tumor microenvironment (TME), as well as various immune cell infiltration, immune function, and immune checkpoints. Furthermore, our results suggested that liver cancer patients with low PSMD1 expression were more actively responsive to immunotherapy according to TIDE predictions. Additionally, we observed significant differences in patient survival based on the different immune molecular types of tumors and their correlation with PSMD1 expression. The close relationship between PSMD1 and copy number variation (CNV), tumor mutational burden (TMB), and methylation was also confirmed, showing a significant impact on patient survival. Moreover, the pan-cancer analysis revealed that PSMD1 is closely related to the diagnosis and prognosis of various cancers, as well as immune infiltration across different cancer types. In summary, PSMD1 has the potential to be a useful diagnostic and prognostic biomarker for LIHC and other types of cancers. It is closely associated with indicators such as immune infiltration, CNV, TMB, and methylation. The identification of PSMD1 may offer a potential intervention target for LIHC and various cancers.
Collapse
Affiliation(s)
- Xing Chen
- Hepatobiliary Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi 046000, China
- Changzhi Medical College, Changzhi, Shanxi 046000, China
| | - Guihai Liu
- Clinical Drug Experiment Center, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi 046000, China
| | - Buqiang Wu
- Hepatobiliary Surgery, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi 046000, China
| |
Collapse
|
21
|
Terui H, Segawa Y, Asano Y. Targeting B cells for treatment of systemic sclerosis. Curr Opin Rheumatol 2023; 35:317-323. [PMID: 37540776 DOI: 10.1097/bor.0000000000000961] [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: 08/06/2023]
Abstract
PURPOSE OF REVIEW The pathogenesis of systemic sclerosis (SSc) has been linked to dysfunctional B cells as demonstrated in previous research. This review aims to show the evidence and ongoing clinical trials of B cell-targeted therapy and overview the various aspects of B cell involvement in SSc. RECENT FINDINGS We provide an overview of the current understanding and therapeutic strategies targeting B cells in SSc patients. Several molecular targets of B cells have been identified for treating SSc, including CD20, CD19, B-cell activating factor (BAFF), and proteasome. SUMMARY Many clinical trials have demonstrated that B cells play a critical role in the pathogenesis of SSc and may be a potential therapeutic target to improve disease symptoms. Although large-scale clinical studies are needed, various B cell-targeted therapies have the potential to address the unmet needs of SSc patients.
Collapse
Affiliation(s)
- Hitoshi Terui
- Department of Dermatology, Tohoku University Graduate School of Medicine, Japan
| | | | | |
Collapse
|
22
|
Zhao X, Wang Y, Xia H, Liu S, Huang Z, He R, Yu L, Meng N, Wang H, You J, Li J, Yam JWP, Xu Y, Cui Y. Roles and Molecular Mechanisms of Biomarkers in Hepatocellular Carcinoma with Microvascular Invasion: A Review. J Clin Transl Hepatol 2023; 11:1170-1183. [PMID: 37577231 PMCID: PMC10412705 DOI: 10.14218/jcth.2022.00013s] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 01/18/2023] [Accepted: 03/21/2023] [Indexed: 07/03/2023] Open
Abstract
Hepatocellular carcinoma (HCC) being a leading cause of cancer-related death, has high associated mortality and recurrence rates. It has been of great necessity and urgency to find effective HCC diagnosis and treatment measures. Studies have shown that microvascular invasion (MVI) is an independent risk factor for poor prognosis after hepatectomy. The abnormal expression of biomacromolecules such as circ-RNAs, lncRNAs, STIP1, and PD-L1 in HCC patients is strongly correlated with MVI. Deregulation of several markers mentioned in this review affects the proliferation, invasion, metastasis, EMT, and anti-apoptotic processes of HCC cells through multiple complex mechanisms. Therefore, these biomarkers may have an important clinical role and serve as promising interventional targets for HCC. In this review, we provide a comprehensive overview on the functions and regulatory mechanisms of MVI-related biomarkers in HCC.
Collapse
Affiliation(s)
- Xudong Zhao
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yudan Wang
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Haoming Xia
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shuqiang Liu
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ziyue Huang
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Risheng He
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Liang Yu
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Nanfeng Meng
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hang Wang
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Junqi You
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jinglin Li
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Judy Wai Ping Yam
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Xiamen Medical College, Xiamen, Fujian, China
- Jiangsu Province Engineering Research Center of Tumor Targeted Nano Diagnostic and Therapeutic Materials, Yancheng Teachers University, Yancheng, Jiangsu, China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang province, Hangzhou, Zhejiang, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
- Key Laboratory of Intelligent Pharmacy and Individualized Therapy of Huzhou, Department of Pharmacy, Changxing People’s Hospital, Changxing, Zhejiang, China
| | - Yunfu Cui
- Department of Hepatopancreatobiliary Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
23
|
Gumkowska-Sroka O, Kotyla K, Mojs E, Palka K, Kotyla P. Novel Therapeutic Strategies in the Treatment of Systemic Sclerosis. Pharmaceuticals (Basel) 2023; 16:1066. [PMID: 37630981 PMCID: PMC10458905 DOI: 10.3390/ph16081066] [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: 05/29/2023] [Revised: 07/09/2023] [Accepted: 07/17/2023] [Indexed: 08/27/2023] Open
Abstract
Systemic sclerosis is a connective tissue disease of unknown origin and with an unpredictable course, with both cutaneous and internal organ manifestations. Despite the enormous progress in rheumatology and clinical immunology, the background of this disease is largely unknown, and no specific therapy exists. The therapeutic approach aims to treat and preserve the function of internal organs, and this approach is commonly referred to as organ-based treatment. However, in modern times, data from other branches of medicine may offer insight into how to treat disease-related complications, making it possible to find new drugs to treat this disease. In this review, we present therapeutic options aiming to stop the progression of fibrotic processes, restore the aberrant immune response, stop improper signalling from proinflammatory cytokines, and halt the production of disease-related autoantibodies.
Collapse
Affiliation(s)
- Olga Gumkowska-Sroka
- Department of Rheumatology and Clinical Immunology, Voivodeship Hospital No. 5 in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland;
- Department of Internal Medicine Rheumatology and Clinical Immunology, Medical University of Silesia, 40-055 Katowice, Poland; (K.K.); (K.P.)
| | - Kacper Kotyla
- Department of Internal Medicine Rheumatology and Clinical Immunology, Medical University of Silesia, 40-055 Katowice, Poland; (K.K.); (K.P.)
| | - Ewa Mojs
- Department of Clinical Psychology, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Klaudia Palka
- Department of Internal Medicine Rheumatology and Clinical Immunology, Medical University of Silesia, 40-055 Katowice, Poland; (K.K.); (K.P.)
| | - Przemysław Kotyla
- Department of Rheumatology and Clinical Immunology, Voivodeship Hospital No. 5 in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland;
- Department of Internal Medicine Rheumatology and Clinical Immunology, Medical University of Silesia, 40-055 Katowice, Poland; (K.K.); (K.P.)
| |
Collapse
|
24
|
Huang Z, Tan Y. The Potential of Cylindromatosis (CYLD) as a Therapeutic Target in Oxidative Stress-Associated Pathologies: A Comprehensive Evaluation. Int J Mol Sci 2023; 24:8368. [PMID: 37176077 PMCID: PMC10179184 DOI: 10.3390/ijms24098368] [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: 03/29/2023] [Revised: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Oxidative stress (OS) arises as a consequence of an imbalance between the formation of reactive oxygen species (ROS) and the capacity of antioxidant defense mechanisms to neutralize them. Excessive ROS production can lead to the damage of critical biomolecules, such as lipids, proteins, and DNA, ultimately contributing to the onset and progression of a multitude of diseases, including atherosclerosis, chronic obstructive pulmonary disease, Alzheimer's disease, and cancer. Cylindromatosis (CYLD), initially identified as a gene linked to familial cylindromatosis, has a well-established and increasingly well-characterized function in tumor inhibition and anti-inflammatory processes. Nevertheless, burgeoning evidence suggests that CYLD, as a conserved deubiquitination enzyme, also plays a pivotal role in various key signaling pathways and is implicated in the pathogenesis of numerous diseases driven by oxidative stress. In this review, we systematically examine the current research on the function and pathogenesis of CYLD in diseases instigated by oxidative stress. Therapeutic interventions targeting CYLD may hold significant promise for the treatment and management of oxidative stress-induced human diseases.
Collapse
Affiliation(s)
| | - Yanjie Tan
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China;
| |
Collapse
|
25
|
Nakaue E, Teramachi J, Tenshin H, Hiasa M, Harada T, Oda A, Inoue Y, Shimizu S, Higa Y, Sogabe K, Oura M, Hara T, Sumitani R, Maruhashi T, Yamagami H, Endo I, Tanaka E, Abe M. Mechanisms of preferential bone formation in myeloma bone lesions by proteasome inhibitors. Int J Hematol 2023:10.1007/s12185-023-03601-2. [PMID: 37039914 DOI: 10.1007/s12185-023-03601-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/04/2023] [Accepted: 04/04/2023] [Indexed: 04/12/2023]
Abstract
Proteasome inhibitors (PIs) can preferentially restore bone in bone-defective lesions of patients with multiple myeloma (MM) who respond favorably to these drugs. Most prior in vitro studies on PIs used continuous exposure to low PI concentrations, although pharmacokinetic analysis in patients has shown that serum concentrations of PIs change in a pulsatile manner. In the present study, we explored the effects of pulsatile treatment with PIs on bone metabolism to simulate in vivo PI pharmacokinetics. Pulsatile treatment with bortezomib, carfilzomib, or ixazomib induced MM cell death but only marginally affected the viability of osteoclasts (OCs) with F-actin ring formation. Pulsatile PI treatment suppressed osteoclastogenesis in OC precursors and bone resorption by mature OCs. OCs robustly enhanced osteoblastogenesis in cocultures with OCs and MC3T3-E1 pre-osteoblastic cells, indicating OC-mediated coupling to osteoblastogenesis. Importantly, pulsatile PI treatment did not impair robust OC-mediated osteoblastogenesis. These results suggest that PIs might sufficiently reduce MM cell-derived osteoblastogenesis inhibitors to permit OC-driven bone formation coupling while suppressing OC differentiation and activity in good responders to PIs. OC-mediated coupling to osteoblastogenesis appears to be a predominant mechanism for preferential occurrence of bone regeneration at sites of osteoclastic bone destruction in good responders.
Collapse
Affiliation(s)
- Emiko Nakaue
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Jumpei Teramachi
- Department of Oral Function and Anatomy, Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama University Graduate School, 2-5-1 Shikata, Okayama, 700-8525, Japan.
| | - Hirofumi Tenshin
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masahiro Hiasa
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Takeshi Harada
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Asuka Oda
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Yusuke Inoue
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - So Shimizu
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yoshiki Higa
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kimiko Sogabe
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Masahiro Oura
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Tomoyo Hara
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Ryohei Sumitani
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Tomoko Maruhashi
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Hiroki Yamagami
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Itsuro Endo
- Department of Bioregulatory Sciences, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Eiji Tanaka
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masahiro Abe
- Department of Hematology, Endocrinology and Metabolism, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan.
| |
Collapse
|
26
|
Paterek A, Oknińska M, Pilch Z, Sosnowska A, Ramji K, Mackiewicz U, Golab J, Nowis D, Mączewski M. Arginase Inhibition Mitigates Bortezomib-Exacerbated Cardiotoxicity in Multiple Myeloma. Cancers (Basel) 2023; 15:cancers15072191. [PMID: 37046852 PMCID: PMC10093116 DOI: 10.3390/cancers15072191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) is associated with increased cardiovascular morbidity and mortality, while MM therapies also result in adverse cardiac effects. Endothelial dysfunction and impaired nitric oxide (NO) pathway is their possible mediator. OBJECTIVE Since MM is associated with increased arginase expression, resulting in the consumption of ʟ-arginine, precursor for NO synthesis, our aim was to test if cardiotoxicity mediated by MM and MM therapeutic, bortezomib (a proteasome inhibitor), can be ameliorated by an arginase inhibitor through improved endothelial function. METHODS We used a mouse Vĸ*MYC model of non-light chain MM. Cardiac function was assessed by echocardiography. RESULTS MM resulted in progressive left ventricular (LV) systolic dysfunction, and bortezomib exacerbated this effect, leading to significant impairment of LV performance. An arginase inhibitor, OAT-1746, protected the heart against bortezomib- or MM-induced toxicity but did not completely prevent the effects of the MM+bortezomib combination. MM was associated with improved endothelial function (assessed as NO production) vs. healthy controls, while bortezomib did not affect it. OAT-1746 improved endothelial function only in healthy mice. NO plasma concentration was increased by OAT-1746 but was not affected by MM or bortezomib. CONCLUSIONS Bortezomib exacerbates MM-mediated LV systolic dysfunction in a mouse model of MM, while an arginase inhibitor partially prevents it. Endothelium does not mediate either these adverse or beneficial effects. This suggests that proteasome inhibitors should be used with caution in patients with advanced myeloma, where the summation of cardiotoxicity could be expected. Therapies aimed at the NO pathway, in particular arginase inhibitors, could offer promise in the prevention/treatment of cardiotoxicity in MM.
Collapse
Affiliation(s)
- Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka Street, 01-813 Warsaw, Poland
| | - Marta Oknińska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka Street, 01-813 Warsaw, Poland
| | - Zofia Pilch
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
| | - Anna Sosnowska
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
| | - Kavita Ramji
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
| | - Urszula Mackiewicz
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka Street, 01-813 Warsaw, Poland
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
- Centre of Preclinical Research, Medical University of Warsaw, 1B Banacha Street, 02-097 Warsaw, Poland
| | - Dominika Nowis
- Department of Immunology, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
- Laboratory of Experimental Medicine, Medical University of Warsaw, 5 Nielubowicza Street, 02-097 Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, 99/103 Marymoncka Street, 01-813 Warsaw, Poland
| |
Collapse
|
27
|
Biomedical importance of the ubiquitin-proteasome system in diabetes and metabolic transdifferentiation of pancreatic duct epithelial cells into β-cells. Gene 2023; 858:147191. [PMID: 36632913 DOI: 10.1016/j.gene.2023.147191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/09/2023]
Abstract
The ubiquitin-proteasome system (UPS) is a major pathway for cellular protein degradation. The molecular function of the UPS is the removal of damaged proteins, and this function is applied in many biological processes, including inflammation, proliferation, and apoptosis. Accumulating evidence also suggests that the UPS also has a key role in pancreatic β-cell transdifferentiation in diabetes and can be targeted for treatment of diabetic diseases. In this review, we summarized the mechanistic roles of the UPS in the biochemical activities of pancreatic β-cells, including the role of the UPS in insulin synthesis and secretion, as well as β-cell degradation. Also, we discuss how the UPS mediates the transdifferentiation of pancreatic duct epithelial cells into β-cells as the experimental basis for the development of new strategies for the treatment of diabetes in regenerative medicine.
Collapse
|
28
|
Bissani Gasparin C, Pilger DA. 8‐Hydroxyquinoline, Derivatives and Metal‐Complexes: A Review of Antileukemia Activities. ChemistrySelect 2023. [DOI: 10.1002/slct.202204219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Affiliation(s)
- Carolina Bissani Gasparin
- Laboratório de Análises Bioquímicas e Citológicas (LABC) Departamento de Análises Faculdade de Farmácia Universidade Federal do Rio Grande do Sul (UFRGS) Av. Ipiranga 2752, Bairro Santana CEP 90610–000 Porto Alegre RS Brazil
| | - Diogo André Pilger
- Laboratório de Análises Bioquímicas e Citológicas (LABC) Departamento de Análises Faculdade de Farmácia Universidade Federal do Rio Grande do Sul (UFRGS) Av. Ipiranga 2752, Bairro Santana CEP 90610–000 Porto Alegre RS Brazil
- Postgraduate Program in Pharmaceutical Sciences Universidade Federal do Rio Grande do Sul (UFRGS) Av. Ipiranga 2752, Bairro Santana CEP 90610–000 Porto Alegre RS Brazil
| |
Collapse
|
29
|
Zeng G, Yu Q, Zhuang R, Zhu H, Shao J, Xi J, Zhang J. Recent Advances and Future Perspectives of Noncompetitive Proteasome Inhibitors. Bioorg Chem 2023; 135:106507. [PMID: 37030106 DOI: 10.1016/j.bioorg.2023.106507] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
The proteasome regulates intracellular processes, maintains biological homeostasis, and has shown great significance in the study of various diseases, such as neurodegenerative diseases, immune-related diseases, and cancer, especially in hematologic malignancies such as multiple myeloma (MM) and mantle cell lymphoma (MCL). All clinically used proteasome inhibitors bind to the active site of the proteasome and thus exhibit a competitive mechanism. The development of resistance and intolerance during treatment drives the search for inhibitors with different mechanisms of action. In this review, we provide an overview of noncompetitive proteasome inhibitors, including their mechanisms of action, function, possible applications, and their advantages and disadvantages compared with competitive inhibitors.
Collapse
|
30
|
Long Y, Hu Z, Yang D, Wang F, Zhao C, Zhang Y, Zhang Y, Ma H, Lv H. Pharmacological inhibition of the ubiquitin-specific protease 8 effectively suppresses glioblastoma cell growth. Open Life Sci 2023; 18:20220562. [PMID: 36816802 PMCID: PMC9922063 DOI: 10.1515/biol-2022-0562] [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: 11/02/2022] [Revised: 12/08/2022] [Accepted: 01/04/2023] [Indexed: 02/11/2023] Open
Abstract
Glioblastoma (GBM) is a malignant brain tumor. The purpose of this study is to estimate the potential effects and underlying mechanisms of a ubiquitin-specific protease 8 (USP8) small-molecule inhibitor on the phenotypic characteristics of GBM cells. The growth, migration, invasion, and stemness of GBM LN229 and T98G cells were evaluated by conducting cell proliferation, colony formation, wound healing, transwell, Ki-67 staining, spheroid formation, and ionizing radiation assays, and the results collectively showed the suppressive effects of USP8 inhibition on GBM cells. Furthermore, transcriptomic profiling of GBM cells treated with the USP8 inhibitor deubiquitinase (DUB)-IN-1 revealed significantly altered mRNA expression induced by pharmacological USP8 inhibition, from which we confirmed downregulated Aurora kinase A (AURKA) protein levels using immunoblotting assays. Our findings indicated that the proliferation, invasion, and stemness of LN229 and T98G cells were markedly suppressed by USP8 inhibition. Pharmacological USP8 suppression elicits multiple tumor-inhibitory effects, likely through dysregulating various mRNA expression events, including that of the key cell cycle regulator and oncogenic protein AURKA. Therefore, our observations corroborate the GBM-supportive roles of USP8 and suggest pharmacological USP8 inhibition is a viable therapeutic approach to target GBM. The purpose of this study was to investigate the effect and mechanism of action of the USP8 inhibitor DUB-IN-1 on GBM.
Collapse
Affiliation(s)
- Yu Long
- Department of Pharmacy, The Second Affiliated Hospital, Dalian Medical University, No. 467 Zhongshan Road, Dalian 116000, China
| | - Zengchun Hu
- Department of Neurosurgery, The Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Dian Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Fuqiang Wang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Chen’ge Zhao
- Department of Pharmacy, The Second Affiliated Hospital, Dalian Medical University, No. 467 Zhongshan Road, Dalian 116000, China
| | - Yang Zhang
- Department of Pharmacy, The Second Affiliated Hospital, Dalian Medical University, No. 467 Zhongshan Road, Dalian 116000, China
| | - Yingqiu Zhang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian 116000, China
| | - Hui Ma
- Department of Pharmacy, The Second Affiliated Hospital, Dalian Medical University, No. 467 Zhongshan Road, Dalian 116000, China
| | - Huiyi Lv
- Department of Pharmacy, The Second Affiliated Hospital, Dalian Medical University, No. 467 Zhongshan Road, Dalian 116000, China,Dalian Kexiang Technology Development Co., LTD, No. 467 Zhongshan Road, Dalian 116000, China
| |
Collapse
|
31
|
Autophagy/Mitophagy Regulated by Ubiquitination: A Promising Pathway in Cancer Therapeutics. Cancers (Basel) 2023; 15:cancers15041112. [PMID: 36831455 PMCID: PMC9954143 DOI: 10.3390/cancers15041112] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Autophagy is essential for organismal development, maintenance of energy homeostasis, and quality control of organelles and proteins. As a selective form of autophagy, mitophagy is necessary for effectively eliminating dysfunctional mitochondria. Both autophagy and mitophagy are linked with tumor progression and inhibition. The regulation of mitophagy and autophagy depend upon tumor type and stage. In tumors, mitophagy has dual roles: it removes damaged mitochondria to maintain healthy mitochondria and energy production, which are necessary for tumor growth. In contrast, mitophagy has been shown to inhibit tumor growth by mitigating excessive ROS production, thus preventing mutation and chromosomal instability. Ubiquitination and deubiquitination are important modifications that regulate autophagy. Multiple E3 ubiquitin ligases and DUBs modulate the activity of the autophagy and mitophagy machinery, thereby influencing cancer progression. In this review, we summarize the mechanistic association between cancer development and autophagy/mitophagy activities regulated by the ubiquitin modification of autophagic proteins. In addition, we discuss the function of multiple proteins involved in autophagy/mitophagy in tumors that may represent potential therapeutic targets.
Collapse
|
32
|
Zhan W, Li D, Saha P, Wang R, Zhang H, Ajay AK, Deban C, Sukenick G, Azzi J, Lin G. Discovery of Highly Selective Inhibitors of the Human Constitutive Proteasome β5c Chymotryptic Subunit. J Med Chem 2023; 66:1172-1185. [PMID: 36608337 PMCID: PMC10157300 DOI: 10.1021/acs.jmedchem.2c00733] [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] [Indexed: 01/07/2023]
Abstract
We describe our discovery and development of potent and highly selective inhibitors of human constitutive proteasome chymotryptic activity (β5c). Structure-activity relationship studies of the novel class of inhibitors focused on optimization of N-cap, C-cap, and side chain of the chemophore asparagine. Compound 32 is the most potent and selective β5c inhibitor in this study. A docking study provides a structure rationale for potency and selectivity. Kinetic studies show a reversible and noncompetitive inhibition mechanism. It enters the cells to engage the proteasome target, potently and selectively kills multiple myeloma cells, and does so by synergizing with a β5i-selective inhibitor.
Collapse
Affiliation(s)
- Wenhu Zhan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| | - Daqiang Li
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| | - Priya Saha
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| | - Rong Wang
- NMR Analytical Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Hao Zhang
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| | - Amrendra K. Ajay
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christa Deban
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - George Sukenick
- NMR Analytical Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Jamil Azzi
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gang Lin
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| |
Collapse
|
33
|
Wani S, Humaira, Farooq I, Ali S, Rehman MU, Arafah A. Proteomic profiling and its applications in cancer research. Proteomics 2023. [DOI: 10.1016/b978-0-323-95072-5.00015-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
34
|
Cancer proteomics: An overview. Proteomics 2023. [DOI: 10.1016/b978-0-323-95072-5.00009-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
35
|
Firdous P, Hassan T, Farooq S, Nissar K. Applications of proteomics in cancer diagnosis. Proteomics 2023. [DOI: 10.1016/b978-0-323-95072-5.00014-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
36
|
Pereira WA, Nascimento ÉCM, Martins JBL. Electronic and structural study of T315I mutated form in DFG-out conformation of BCR-ABL inhibitors. J Biomol Struct Dyn 2022; 40:9774-9788. [PMID: 34121617 DOI: 10.1080/07391102.2021.1935320] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this work, the four main drugs for the treatment of chronic myeloid leukemia were analyzed, being imatinib, dasatinib, nilotinib and ponatinib followed by four derivative molecules of nilotinib and ponatinib. For these derivative molecules, the fluorine atoms were replaced by hydrogen and chlorine atoms in order to shade light to the structural effects on this set of inhibitors. Electronic studies were performed at density functional theory level with the B3LYP functional and 6-311+G(d,p) basis set. The frontier molecular orbitals, gap HOMO-LUMO, and NBO were analyzed and compared to docking studies for mutant T315I tyrosine kinase protein structure code 3IK3, in the DFG-out conformation. Structural similarities were pointed out, such as the presence of groups common to all inhibitors and modifications raised up on new generations of imatinib-based inhibitors. One of them is the trifluoromethyl group present in nilotinib and later included in ponatinib, in addition to the 1-methylpiperazin-1-ium group that is present in imatinib and ponatinib. The frontier molecular orbitals of imatinib and ponatinib are contributing to the same amino acid residues, and the ineffectiveness of imatinib against the T315I mutation was discussed.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Washington A Pereira
- Institute of Chemistry, Laboratory of Computational Chemistry, University of Brasília, Brasília, Federal District, Brazil
| | - Érica C M Nascimento
- Institute of Chemistry, Laboratory of Computational Chemistry, University of Brasília, Brasília, Federal District, Brazil
| | - João B L Martins
- Institute of Chemistry, Laboratory of Computational Chemistry, University of Brasília, Brasília, Federal District, Brazil
| |
Collapse
|
37
|
Oksuzoglu E, Dursun B. Patterns of the Expression of Cyclin Genes in Bortezomib-Sensitive and Resistant Cells of Multiple Myeloma. BIOL BULL+ 2022. [DOI: 10.1134/s1062359022140126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
38
|
Filali H, El Yaagoubi OM, Ayoub Lahmadi, Quessar A, Antri SE, Samaki H, Aboudkhil S. Functional and quantitative evaluation of the 20S proteasome in serum and intracellular in145 moroccan patients with hematologic malignancies. Clin Proteomics 2022; 19:41. [PMID: 36380291 PMCID: PMC9664591 DOI: 10.1186/s12014-022-09375-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/22/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Regulatory degradation of intracellular proteins plays an essential role in most biological processes, particularly in the control of cell proliferation and differentiation. In eukaryotes, intracellular proteolysis is largely provided by the Ubiquitin / Proteasome system. Alterations and dysfunction of protein degradation by the Ubiquitin / Proteasome system, such as transcription factors, cell cycle regulators or tumor suppressor proteins, have been linked to human. Pathologies, including blood cancers. Mainly localized in the nucleus and cytoplasm of cells, the proteasome can be detected in the cell culture supernatant or in the peripheral blood of patients. This study deals with the problems of the search for serum markers specific to certain pathologies and which would be useful in the prevention, diagnosis and monitoring of cancers and which could be used as a therapeutic tool. METHODS The functional and quantitative analysis of the proteasome is carried out at the serum and subcellular level during a pathological phenomenon in a population of 145 Moroccan patients (sex ratio: 1.10 / average age: 47.9 ± 15, 3 years) using an indirect ELISA test and a follow-up of the fluorescence emitted after enzymatic digestion of specific peptides by proteolytic activity (chymotrypsin-like). RESULTS The evolutionary trend proteasome subcellular is significantly linked to the rate of chymotrypsin-like activity. The entire population of 60 patients called back for a second blood test. After three months of treatment reported a significant drop in the rate and the activity of the proteasome in serum and intracellular level. CONCLUSIONS Although the serum proteasome level is a potential new tool for the monitoring of. Patientswithliquid cancer. TRIAL REGISTRATION retrospectively registered.
Collapse
Affiliation(s)
- Hassan Filali
- Laboratory of Biochemistry, Environment and Agrifood (URAC 36), Faculty of Science and Technology, Mohammedia-University Hassan II of Casablanca, Mohammedia, Morocco.
| | - Ouadie Mohamed El Yaagoubi
- Laboratory of Biochemistry, Environment and Agrifood (URAC 36), Faculty of Science and Technology, Mohammedia-University Hassan II of Casablanca, Mohammedia, Morocco.
| | - Ayoub Lahmadi
- Laboratory of Biochemistry, Environment and Agrifood (URAC 36), Faculty of Science and Technology, Mohammedia-University Hassan II of Casablanca, Mohammedia, Morocco
| | - Asmaa Quessar
- Hematology and Pediatric Oncology Service-Hospital August 20, UniversityHospital Center IBN ROCHD Casablanca, Casablanca, Morocco
| | - Said El Antri
- Laboratory of Biochemistry, Environment and Agrifood (URAC 36), Faculty of Science and Technology, Mohammedia-University Hassan II of Casablanca, Mohammedia, Morocco
| | - Hamid Samaki
- National Institute of Social Actions (INAS), Tanger, Morocco
| | - Souad Aboudkhil
- Laboratory of Biochemistry, Environment and Agrifood (URAC 36), Faculty of Science and Technology, Mohammedia-University Hassan II of Casablanca, Mohammedia, Morocco
| |
Collapse
|
39
|
Sheu ML, Pan LY, Hu HY, Su HL, Sheehan J, Tsou HK, Pan HC. Potential Therapeutic Effects of Thiazolidinedione on Malignant Glioma. Int J Mol Sci 2022; 23:ijms232113510. [PMID: 36362294 PMCID: PMC9657575 DOI: 10.3390/ijms232113510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/25/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary malignant tumor of the central nervous system. GBM has a very low 5-year survival rate and reaching merely a median of ~15 months even with aggressive treatments. PPARγ (Peroxisome proliferator- activated receptor gamma) agonists (ciglitazone), while being widely used on patients of type 2 diabetes mellitus, also have approved anticancer effects. Their action mechanisms on malignant glioma are not fully understood. The aim of this study is to investigate the potential therapeutic effect of PPARγ agonists on maligant glioma. Glioma cell line and in-vivo/ex-vivo animal model intervened by ciglitazone were used to assess the associated mechanism and therapeutic effect. Our results from in vivo and ex vivo experiments showed that ciglitazone not only inhibited tumor growth and its associated angiogenesis, but it also reduced colony formation and migration of tumors. Ciglitazone inhibited the phosphorylation of STAT3 (signal transducer and activator of transcription 3) (at the point of tyrosine 705 by increasing both the amount and activity of SHP-2 (Src homology region 2-containing protein tyrosine phosphatase 2) proteins, based on evidence obtained from immunoprecipitation and immunohistochemistry. Furthermore, ciglitazone activated proteasomes and lysosomes to degrade cell-cycle-related proteins like Cyclin D1, Cyclin E, CDK2 (Cyclin-dependent kinase 2), and CDK4 (Cyclin-dependent kinase 4). Ciglitazone triggered expressions of LC3 (Microtubule-associated protein 1A/1B-light chain 3) and formation of acidic vesicular organelles (AVOs), both of which were implicated in the autophagy pathway. In conclusion, ciglitazone showed the multiple actions to regulate the growth of glioma, which appeared to be a potential candidate for treating malignant glioma.
Collapse
Affiliation(s)
- Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung 402, Taiwan
| | - Liang-Yi Pan
- Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Huai-Yun Hu
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung 402, Taiwan
| | - Hong-Lin Su
- Department of Life Sciences, Agriculture Biotechnology Center, National Chun-Hsing University, Taichung 402, Taiwan
| | - Jason Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Hsi-Kai Tsou
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung 40210, Taiwan
| | - Hung-Chuan Pan
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung 402, Taiwan
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Correspondence: ; Tel.: +886-4-23592525 (ext. 4018)
| |
Collapse
|
40
|
Xu H, Xu G, Xu Q, Xu C, Zhou X, Bai Y, Yin L, Ding Y, Wang W. MLN2238 exerts its anti-tumor effects via regulating ROS/JNK/mitochondrial signaling pathways in intrahepatic cholangiocarcinoma. Front Pharmacol 2022; 13:1040847. [PMID: 36386204 PMCID: PMC9659592 DOI: 10.3389/fphar.2022.1040847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/19/2022] [Indexed: 07/30/2023] Open
Abstract
Background: Intrahepatic Cholangiocarcinoma (iCCA) is a highly malignant tumor with limited treatment options that contributes largely to cancer-related deaths worldwide. Compared with traditional transcriptomic analysis, single-cell RNA sequencing (scRNA-seq) is emerging as a more advanced and popular tool for the in-depth exploration of cellular diversity and molecular complexity. As a next-generation proteasome inhibitor, MLN2238 presents better pharmacodynamics, pharmacokinetics, and therapeutic responses in various cancers. However, its effects and mechanisms of action in iCCA remain unknown. Methods: iCCA tumor heterogeneity was determined based on 4,239 qualified scRNA-seq data from 10 iCCA samples. The potential biological roles of proteasome-related genes in iCCA were investigated using a pseudo-trajectory reconstruction. The effect of MLN2238 on iCCA cell proliferation was estimated using the CCK-8, EdU, and clone formation assays. Flow cytometry was used to examine the effect of added MLN2238 on cell cycle and apoptosis levels. Autophagic flux was detected using AdPlus-mCherry-GFP-LC3B cells. ROS levels and mitochondrial membrane potential were determined using DCFH-DA probing and JC-1 staining. JNK activation and mitochondrial apoptosis were observed using western blotting and immunofluorescence microscopy, respectively. Finally, we used a tumor-bearing mouse model to validate its efficacy in vivo for iCCA treatment. Results: Proteasome-related genes were dysregulated in iCCA progression and expressed at higher levels in tumor tissues. MLN2238 suppressed cell proliferation, blocked the cell cycle in the G2/M phase, promoted apoptosis, and induced cytoprotective autophagy in iCCA cells. Furthermore, MLN2238 increased ROS levels and activated the JNK signaling pathway. Inhibition of ROS and JNK activation by NAC and SP600125 significantly reversed MLN2238-induced apoptosis. MLN2238 also suppressed the growth of iCCA tumors in vivo. Conclusion: Proteasome-related genes play pivotal roles in iCCA development. MLN2238, as a proteasome inhibitor, induces apoptosis in iCCA cells through ROS/JNK/mitochondrial signaling pathways, and hence, making MLN2238 a potential therapeutic choice for iCCA.
Collapse
Affiliation(s)
- Hao Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Guangyu Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Qianhui Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Chang Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Xiaohu Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yang Bai
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Lu Yin
- Department of Pathology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, China
- Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, China
- Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease of Zhejiang University, Hangzhou, China
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
41
|
Viera CR, Stevens BT, Viera T, Zielinski C, Uranga LA, Rogelj S, Patidar PL, Tello-Aburto R. Cystargolide-based amide and ester Pz analogues as proteasome inhibitors and anti-cancer agents. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220358. [PMID: 36177203 PMCID: PMC9515629 DOI: 10.1098/rsos.220358] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/29/2022] [Indexed: 06/16/2023]
Abstract
A series of cystargolide-based β-lactone analogues containing nitrogen atoms at the Pz portion of the scaffold were prepared and evaluated as proteasome inhibitors, and for their cytotoxicity profile toward several cancer cell lines. Inclusion of one, two or even three nitrogen atoms at the Pz portion of the cystargolide scaffold is well tolerated, producing analogues with low nanomolar proteasome inhibition activity, in many cases superior to carfilzomib. Additionally, analogue 8g, containing an ester and pyrazine group at Pz, was shown to possess significant activity toward RPMI 8226 cells (IC50 = 21 nM) and to be less cytotoxic toward the normal tissue model MCF10A cells than carfilzomib.
Collapse
Affiliation(s)
- Carlos R. Viera
- Department of Chemistry, New Mexico Institute of Mining and Technology, Socorro, NM 87801, USA
| | - Bradley T. Stevens
- Department of Biology, New Mexico Institute of Mining and Technology, Socorro, NM 87801, USA
| | - Talysa Viera
- Department of Chemistry, New Mexico Institute of Mining and Technology, Socorro, NM 87801, USA
| | - Cameron Zielinski
- Department of Chemical Engineering, New Mexico Institute of Mining and Technology, Socorro, NM 87801, USA
| | - Lee A. Uranga
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM 88003, USA
| | - Snezna Rogelj
- Department of Biology, New Mexico Institute of Mining and Technology, Socorro, NM 87801, USA
| | - Praveen L. Patidar
- Department of Chemistry, New Mexico Institute of Mining and Technology, Socorro, NM 87801, USA
| | - Rodolfo Tello-Aburto
- Department of Chemistry and Biochemistry, New Mexico State University, Las Cruces, NM 88003, USA
| |
Collapse
|
42
|
Barreto IV, Machado CB, Almeida DB, Pessoa FMCDP, Gadelha RB, Pantoja LDC, Oliveira DDS, Ribeiro RM, Lopes GS, de Moraes Filho MO, de Moraes MEA, Khayat AS, de Oliveira EHC, Moreira-Nunes CA. Kinase Inhibition in Multiple Myeloma: Current Scenario and Clinical Perspectives. Pharmaceutics 2022; 14:pharmaceutics14091784. [PMID: 36145532 PMCID: PMC9506264 DOI: 10.3390/pharmaceutics14091784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a blood cell neoplasm characterized by excessive production of malignant monoclonal plasma cells (activated B lymphocytes) by the bone marrow, which end up synthesizing antibodies or antibody fragments, called M proteins, in excess. The accumulation of this production, both cells themselves and of the immunoglobulins, causes a series of problems for the patient, of a systemic and local nature, such as blood hyperviscosity, renal failure, anemia, bone lesions, and infections due to compromised immunity. MM is the third most common hematological neoplasm, constituting 1% of all cancer cases, and is a disease that is difficult to treat, still being considered an incurable disease. The treatments currently available cannot cure the patient, but only extend their lifespan, and the main and most effective alternative is autologous hematopoietic stem cell transplantation, but not every patient is eligible, often due to age and pre-existing comorbidities. In this context, the search for new therapies that can bring better results to patients is of utmost importance. Protein tyrosine kinases (PTKs) are involved in several biological processes, such as cell growth regulation and proliferation, thus, mutations that affect their functionality can have a great impact on crucial molecular pathways in the cells, leading to tumorigenesis. In the past couple of decades, the use of small-molecule inhibitors, which include tyrosine kinase inhibitors (TKIs), has been a hallmark in the treatment of hematological malignancies, and MM patients may also benefit from TKI-based treatment strategies. In this review, we seek to understand the applicability of TKIs used in MM clinical trials in the last 10 years.
Collapse
Affiliation(s)
- Igor Valentim Barreto
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Caio Bezerra Machado
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | | | - Flávia Melo Cunha de Pinho Pessoa
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Renan Brito Gadelha
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Laudreísa da Costa Pantoja
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil
| | | | | | - Germison Silva Lopes
- Department of Hematology, César Cals General Hospital, Fortaleza 60015-152, CE, Brazil
| | - Manoel Odorico de Moraes Filho
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - Maria Elisabete Amaral de Moraes
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
| | - André Salim Khayat
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil
| | - Edivaldo Herculano Correa de Oliveira
- Faculty of Natural Sciences, Institute of Exact and Natural Sciences, Federal University of Pará (UFPA), Rua Augusto Correa, 01, Belém 66075-990, PA, Brazil
- Laboratory of Cytogenomics and Environmental Mutagenesis, Environment Section (SAMAM), Evandro Chagas Institute (IEC), BR 316, KM 7, s/n, Levilândia, Ananindeua 67030-000, PA, Brazil
| | - Caroline Aquino Moreira-Nunes
- Pharmacogenetics Laboratory, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza 60430-275, CE, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém 66073-005, PA, Brazil
- Northeast Biotechnology Network (RENORBIO), Itaperi Campus, Ceará State University, Fortaleza 60740-903, CE, Brazil
- Correspondence:
| |
Collapse
|
43
|
Masuda T, Haji S, Nakashima Y, Tsuda M, Kimura D, Takamatsu A, Iwahashi N, Umakoshi H, Shiratsuchi M, Kikutake C, Suyama M, Ohkawa Y, Ogawa Y. Identification of a drug-response gene in multiple myeloma through longitudinal single-cell transcriptome sequencing. iScience 2022; 25:104781. [PMID: 35992084 PMCID: PMC9386061 DOI: 10.1016/j.isci.2022.104781] [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: 01/22/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/06/2022] Open
Abstract
Despite recent therapeutic advances for multiple myeloma (MM), relapse is very common. Here, we conducted longitudinal single-cell transcriptome sequencing (scRNA-seq) of MM cells from a patient with relapsed MM, treated with multiple anti-myeloma drugs. We observed five subclusters of MM cells, which appeared and/or disappeared in response to the therapeutic pressure, and identified cluster 3 which emerged during lenalidomide treatment and disappeared after proteasome inhibitor (PI) treatment. Among the differentially expressed genes in cluster 3, we found a candidate drug-response gene; pellino E3 ubiquitin-protein ligase family member 2 (PELI2), which is responsible for PI-induced cell death in in vitro assay. Kaplan-Meier survival analysis of database revealed that higher expression of PELI2 is associated with a better prognosis. Our integrated strategy combining longitudinal scRNA-seq analysis, in vitro functional assay, and database analysis would facilitate the understanding of clonal dynamics of MM in response to anti-myeloma drugs and identification of drug-response genes.
Collapse
Affiliation(s)
- Toru Masuda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shojiro Haji
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yasuhiro Nakashima
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Mariko Tsuda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Daisaku Kimura
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akiko Takamatsu
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Norifusa Iwahashi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hironobu Umakoshi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Motoaki Shiratsuchi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Hematology, Iizuka Hospital, Iizuka 820-8505, Japan
| | - Chie Kikutake
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Mikita Suyama
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
44
|
Zhang H, Ginn J, Zhan W, Liu YJ, Leung A, Toita A, Okamoto R, Wong TT, Imaeda T, Hara R, Yukawa T, Michino M, Vendome J, Beuming T, Sato K, Aso K, Meinke PT, Nathan CF, Kirkman LA, Lin G. Design, Synthesis, and Optimization of Macrocyclic Peptides as Species-Selective Antimalaria Proteasome Inhibitors. J Med Chem 2022; 65:9350-9375. [PMID: 35727231 PMCID: PMC10152543 DOI: 10.1021/acs.jmedchem.2c00611] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
With over 200 million cases and close to half a million deaths each year, malaria is a threat to global health, particularly in developing countries. Plasmodium falciparum, the parasite that causes the most severe form of the disease, has developed resistance to all antimalarial drugs. Resistance to the first-line antimalarial artemisinin and to artemisinin combination therapies is widespread in Southeast Asia and is emerging in sub-Saharan Africa. The P. falciparum proteasome is an attractive antimalarial target because its inhibition kills the parasite at multiple stages of its life cycle and restores artemisinin sensitivity in parasites that have become resistant through mutation in Kelch K13. Here, we detail our efforts to develop noncovalent, macrocyclic peptide malaria proteasome inhibitors, guided by structural analysis and pharmacokinetic properties, leading to a potent, species-selective, metabolically stable inhibitor.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| | - John Ginn
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Wenhu Zhan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| | - Yi J Liu
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| | - Annie Leung
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| | - Akinori Toita
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Rei Okamoto
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Tzu-Tshin Wong
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Toshihiro Imaeda
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Ryoma Hara
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Takafumi Yukawa
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Mayako Michino
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | | | - Thijs Beuming
- Schrödinger, Inc., New York, New York 10036, United States
| | - Kenjiro Sato
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Kazuyoshi Aso
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Peter T Meinke
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Carl F Nathan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| | - Laura A Kirkman
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States.,Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| | - Gang Lin
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| |
Collapse
|
45
|
Cléroux P, Voisin L, Meloche S. Development of a high-throughput assay to identify inhibitors of the ubiquitin-conjugating enzyme UBCH10. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:266-271. [PMID: 35342035 DOI: 10.1016/j.slasd.2022.03.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 06/14/2023]
Abstract
UBCH10 is an ubiquitin-conjugating enzyme (E2) of the anaphase-promoting complex E3 ligase, a key regulator of the cell cycle. The UBCH10 gene and protein are frequently upregulated in multiple solid tumors, associated with an unfavorable outcome. Accumulating evidence from studies of human cancer cell lines, mouse transgenic models, and analyses of clinical samples suggest that UBCH10 is a potential cancer drug target. No small molecule inhibitor of UBCH10 has been reported in the literature. Here, we described the development and optimization of a novel time-resolved fluorescence resonance energy transfer (TR-FRET) UBCH10 assay based on the self-polyubiquitination of the enzyme in the absence of E3. The homogenous assay is robust, sensitive, and scalable to different multi-well formats for high-throughput screening (HTS). We demonstrate the suitability of the TR-FRET assay to identify chemical inhibitors of UBCH10 in a pilot HTS campaign.
Collapse
Affiliation(s)
- Patrick Cléroux
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
| | - Laure Voisin
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
| | - Sylvain Meloche
- Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada; Molecular Biology Program, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada; Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
46
|
Schlesser C, Meul T, Stathopoulos G, Meiners S. Metformin Induces Resistance of Cancer Cells to the Proteasome Inhibitor Bortezomib. Biomolecules 2022; 12:biom12060756. [PMID: 35740881 PMCID: PMC9221333 DOI: 10.3390/biom12060756] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/23/2022] [Accepted: 05/27/2022] [Indexed: 02/06/2023] Open
Abstract
The anti-diabetic drug metformin is currently tested for the treatment of hematological and solid cancers. Proteasome inhibitors, e.g., Bortezomib, are approved for the treatment of multiple myeloma and mantle cell lymphoma but are also studied for lung cancer therapy. We here analyzed the interaction of the two drugs in two cell lines, namely the mantle cell lymphoma Jeko-1 and the non-small-cell lung cancer (NSCLC) H1299 cells, using proliferation and survival assays, native-gel analysis for proteasome activity and assembly, and expression analysis of proteasome assembly factors. Our results demonstrate that metformin treatment induces resistance of cancer cells to the proteasome inhibitor Bortezomib by impairing the activity and assembly of the 26S proteasome complexes. These effects of metformin on proteasome inhibitor sensitivity in cancer cells are of potential relevance for patients that receive proteasome inhibitor therapy.
Collapse
Affiliation(s)
- Camille Schlesser
- Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Ludwig-Maximilians University, Max-Lebsche Platz 31, 81377 Munich, Germany; (C.S.); (T.M.); (G.S.)
| | - Thomas Meul
- Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Ludwig-Maximilians University, Max-Lebsche Platz 31, 81377 Munich, Germany; (C.S.); (T.M.); (G.S.)
| | - Georgios Stathopoulos
- Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Ludwig-Maximilians University, Max-Lebsche Platz 31, 81377 Munich, Germany; (C.S.); (T.M.); (G.S.)
- Member of the German Center for Lung Research (DZL), 35392 Gießen, Germany
- Laboratory for Molecular Respiratory Carcinogenesis, Department of Physiology, Faculty of Medicine, University of Patras, 26504 Rio, Greece
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Ludwig-Maximilians University, Max-Lebsche Platz 31, 81377 Munich, Germany; (C.S.); (T.M.); (G.S.)
- Research Center Borstel/Leibniz Lung Center, Parkallee 1-40, 23845 Borstel, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Sülfeld, Germany
- Institute of Experimental Medicine, Christian-Albrechts University Kiel, 24118 Kiel, Germany
- Correspondence: ; Tel.: +0049-4537-188-5846
| |
Collapse
|
47
|
Luu AK, Cadieux M, Wong M, Macdonald R, Jones R, Choi D, Oblak M, Brisson B, Sauer S, Chafitz J, Warshawsky D, Wood GA, Viloria-Petit AM. Proteomic Assessment of Extracellular Vesicles from Canine Tissue Explants as a Pipeline to Identify Molecular Targets in Osteosarcoma: PSMD14/Rpn11 as a Proof of Principle. Int J Mol Sci 2022; 23:ijms23063256. [PMID: 35328679 PMCID: PMC8953151 DOI: 10.3390/ijms23063256] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma (OS) is a highly malignant bone tumour that has seen little improvement in treatment modalities in the past 30 years. Understanding what molecules contribute to OS biology could aid in the discovery of novel therapies. Extracellular vesicles (EVs) serve as a mode of cell-to-cell communication and have the potential to uncover novel protein signatures. In our research, we developed a novel pipeline to isolate, characterize, and profile EVs from normal bone and osteosarcoma tissue explants from canine OS patients. Proteomic analysis of vesicle preparations revealed a protein signature related to protein metabolism. One molecule of interest, PSMD14/Rpn11, was explored further given its prognostic potential in human and canine OS, and its targetability with the drug capzimin. In vitro experiments demonstrated that capzimin induces apoptosis and reduces clonogenic survival, proliferation, and migration in two metastatic canine OS cell lines. Capzimin also reduces the viability of metastatic human OS cells cultured under 3D conditions that mimic the growth of OS cells at secondary sites. This unique pipeline can improve our understanding of OS biology and identify new prognostic markers and molecular targets for both canine and human OS patients.
Collapse
Affiliation(s)
- Anita K. Luu
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.K.L.); (M.C.); (M.W.); (R.M.)
| | - Mia Cadieux
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.K.L.); (M.C.); (M.W.); (R.M.)
| | - Mackenzie Wong
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.K.L.); (M.C.); (M.W.); (R.M.)
| | - Rachel Macdonald
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.K.L.); (M.C.); (M.W.); (R.M.)
| | - Robert Jones
- Department of Animal Biosciences, Ontario Agricultural College, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Dongsic Choi
- Department of Biochemistry, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea;
| | - Michelle Oblak
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (M.O.); (B.B.)
| | - Brigitte Brisson
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (M.O.); (B.B.)
| | - Scott Sauer
- Vuja De Sciences, Inc., Natick, MA 01760, USA; (S.S.); (D.W.)
| | | | | | - Geoffrey A. Wood
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada;
| | - Alicia M. Viloria-Petit
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (A.K.L.); (M.C.); (M.W.); (R.M.)
- Correspondence:
| |
Collapse
|
48
|
Ni B, Hou J. Promising therapeutic approaches for relapsed/refractory multiple myeloma. Hematology 2022; 27:343-352. [PMID: 35287555 DOI: 10.1080/16078454.2022.2045724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE Treatment strategies for relapsed/refractory MM are particularly complex. In particular, patients who are refractory to the three classes of therapies have limited therapeutic options and poor survival. Fortunately, promising treatments are emerging, but their incorporation into existing treatments still needs to be defined. We will describe the latest trends and emerging developments in the field of therapies for RRMM by analyzing the most recent clinical data and new technologies in drug development. METHODS Pubmed, Embase and Cochrane Library were searched to select eligible studies, the clinical data of new promising treatments were reviewed. The key results of the most recent clinical trial were summarized in Table. RESULTS A total of 13 studies were included in the final analysis involving anti-BCMA CAR T-cell therapy, Combined CAR T-cell therapy, antibody-drug conjugates, bispecific Ab therapy and CELMoDs. The key efficacy and side effects of treatments were summarized. CONCLUSIONS There is great promise for a set of next-generation of rescue therapies, including CAR T-cell therapy, bispecific antibodies, antibody-drug conjugates, and novel PROteolysis Targeting Chimeras. Emerging new treatments for MM provide more choices for relapsed/refractory multiple myeloma (RRMM). The optimal therapy for each patient should be based on disease-related factors, such as previous therapies, duration of response to prior drugs, clinical and biochemical features of relapse, and the relationship of patient comorbidities with known AE profiles of the different therapies.
Collapse
Affiliation(s)
- Beiwen Ni
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jian Hou
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
49
|
Gasparetto C, Schiller GJ, Tuchman SA, Callander NS, Baljevic M, Lentzsch S, Rossi AC, Kotb R, White D, Bahlis NJ, Chen CI, Sutherland HJ, Madan S, LeBlanc R, Sebag M, Venner CP, Bensinger WI, Biran N, Ammu S, Ben-Shahar O, DeCastro A, Van Domelen D, Zhou T, Zhang C, Bentur OS, Shah J, Shacham S, Kauffman M, Lipe B. Once weekly selinexor, carfilzomib and dexamethasone in carfilzomib non-refractory multiple myeloma patients. Br J Cancer 2022; 126:718-725. [PMID: 34802051 PMCID: PMC8605887 DOI: 10.1038/s41416-021-01608-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/20/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Proteasome inhibitors (PIs), including carfilzomib, potentiate the activity of selinexor, a novel, first-in-class, oral selective inhibitor of nuclear export (SINE) compound, in preclinical models of multiple myeloma (MM). METHODS The safety, efficacy, maximum-tolerated dose (MTD) and recommended phase 2 dose (RP2D) of selinexor (80 or 100 mg) + carfilzomib (56 or 70 mg/m2) + dexamethasone (40 mg) (XKd) once weekly (QW) was evaluated in patients with relapsed refractory MM (RRMM) not refractory to carfilzomib. RESULTS Thirty-two patients, median prior therapies 4 (range, 1-8), were enrolled. MM was triple-class refractory in 38% of patients and 53% of patients had high-risk cytogenetics del(17p), t(4;14), t(14;16) and/or gain 1q. Common treatment-related adverse events (all/Grade 3) were thrombocytopenia 72%/47% (G3 and G4), nausea 72%/6%, anaemia 53%/19% and fatigue 53%/9%, all expected and manageable with supportive care and dose modifications. MTD and RP2D were identified as selinexor 80 mg, carfilzomib 56 mg/m2, and dexamethasone 40 mg, all QW. The overall response rate was 78% including 14 (44%) ≥ very good partial responses. Median progression-free survival was 15 months. CONCLUSIONS Weekly XKd is highly effective and well-tolerated. These data support further investigation of XKd in patients with MM.
Collapse
Affiliation(s)
| | - Gary J Schiller
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | | | | | | | | | - Rami Kotb
- Cancer Care Manitoba, Winnipeg, MB, Canada
| | - Darrell White
- Dalhousie University and Queen Elizabeth II Health Sciences Centre, Halifax, NS, Canada
| | - Nizar J Bahlis
- Charbonneau Cancer Research Institute, Calgary, AB, Canada
| | - Christine I Chen
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | | | - Sumit Madan
- Banner MD Anderson Cancer Center, Gilbert, AZ, USA
| | - Richard LeBlanc
- Maisonneuve-Rosemont Hospital, University of Montreal, Montreal, QC, Canada
| | | | | | | | - Noa Biran
- Hackensack Meridian Health, Hackensack University Medical Center, Teaneck, USA
| | - Sonia Ammu
- Karyopharm Therapeutics Inc., Newton, MA, USA
| | | | | | | | | | - Chris Zhang
- Karyopharm Therapeutics Inc., Newton, MA, USA
| | | | - Jatin Shah
- Karyopharm Therapeutics Inc., Newton, MA, USA
| | | | | | - Brea Lipe
- University of Rochester Medical College, Rochester, NY, USA
| |
Collapse
|
50
|
He J, Fan Z, Tian Y, Yang W, Zhou Y, Zhu Q, Zhang W, Qin W, Yi W. Spatiotemporal Activation of Protein O-GlcNAcylation in Living Cells. J Am Chem Soc 2022; 144:4289-4293. [PMID: 35138101 DOI: 10.1021/jacs.1c11041] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
O-linked N-acetylglucosamine (O-GlcNAc) is a prevalent protein modification that plays fundamental roles in both cell physiology and pathology. O-GlcNAc is catalyzed solely by O-GlcNAc transferase (OGT). The study of protein O-GlcNAc function is limited by the lack of tools to control OGT activity with spatiotemporal resolution in cells. Here, we report light control of OGT activity in cells by replacing a catalytically essential lysine residue with a genetically encoded photocaged lysine. This enables the expression of a transiently inactivated form of OGT, which can be rapidly reactivated by photo-decaging. We demonstrate the activation of OGT activity by monitoring the time-dependent increase of cellular O-GlcNAc and profile glycoproteins using mass-spectrometry-based quantitative proteomics. We further apply this activation strategy to control the morphological contraction of fibroblasts. Furthermore, we achieved spatial activation of OGT activity predominantly in the cytosol. Thus, our approach provides a valuable chemical tool to control cellular O-GlcNAc with much needed spatiotemporal precision, which aids in a better understanding of O-GlcNAc function.
Collapse
Affiliation(s)
- Jiahui He
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhiya Fan
- National Center for Protein Sciences Beijing, State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yinping Tian
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China.,Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Weiwei Yang
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yichao Zhou
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiang Zhu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wanjun Zhang
- National Center for Protein Sciences Beijing, State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Weijie Qin
- National Center for Protein Sciences Beijing, State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Wen Yi
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|