151
|
RNA-binding protein 39: a promising therapeutic target for cancer. Cell Death Discov 2021; 7:214. [PMID: 34389703 PMCID: PMC8363639 DOI: 10.1038/s41420-021-00598-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/04/2021] [Accepted: 05/29/2021] [Indexed: 12/14/2022] Open
Abstract
RNA-binding motif protein 39 (RBM39), as a key factor in tumor-targeted mRNA and protein expression, not only plays a vital role in tumorigenesis, but also has broad development prospects in clinical treatment and drug research. Moreover, since RBM39 was identified as a target of sulfonamides, it has played a key role in the emerging field of molecule drug development. Hence, it is of great significance to study the interaction between RBM39 and tumors and the clinical application of drug-targeted therapy. In this paper, we describe the possible multi-level regulation of RBM39, including gene transcription, protein translation, and alternative splicing. Importantly, the molecular function of RBM39 as an important splicing factor in most common tumors is systematically outlined. Furthermore, we briefly introduce RBM39’s tumor-targeted drug research and its clinical application, hoping to give reference significance for the molecular mechanism of RBM39 in tumors, and provide reliable ideas for in-depth research for future therapeutic strategies.
Collapse
|
152
|
Nguyen J, Ghazali R, Batterham P, Perry T. Inhibiting the proteasome reduces molecular and biological impacts of the natural product insecticide, spinosad. PEST MANAGEMENT SCIENCE 2021; 77:3777-3786. [PMID: 33481333 DOI: 10.1002/ps.6290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 01/01/2021] [Accepted: 01/22/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Insecticide targets are often identified by mutations that confer resistance, but the intricacies of insecticide binding and downstream processes leading to insect death often remain obscure. Mutations in α6-like nicotinic acetylcholine receptor subunit genes have been associated with high levels of resistance to spinosad in many insect species, including Drosophila melanogaster. Here, we aimed to expand our understanding of the effects of the natural product insecticide spinosad on its protein target, the α6 subunit, using genetic tools available in D. melanogaster. RESULTS Functional, fluorescently tagged Dα6 subunits (Dα6YFP ) were developed to allow observation of the protein in vivo. Larvae expressing Dα6YFP were exposed to a sub-lethal concentration of spinosyn A (0.025 ppm) for 6 days, leading to a 64% reduction in fluorescence relative to unexposed larvae. Direct application of high doses of spinosyn A to dissected larval brains resulted in a visible 38.25% decrease in Dα6YFP within 20 min, indicating that degradation of the Dα6 protein occurred in response to spinosyn A exposure. Chemical inhibition of the proteasome system using the multiple myeloma treatment drug, PS-341 reduced loss of Dα6YFP in response to spinosyn A at the 20-min time point to 6.35%. In addition, in vivo administration of PS-341 prior to spinosad exposure reduced the effect of spinosad on larval activity. CONCLUSION Based on these data, we propose that exposure to spinosad leads to degradation of the α6-like target protein, a potentially novel element in the mode of action of spinosyns that may contribute to their toxicity towards insects. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Joseph Nguyen
- School of BioSciences, Bio21 Institute, The University of Melbourne, Parkville, Australia
| | - Razi Ghazali
- School of BioSciences, Bio21 Institute, The University of Melbourne, Parkville, Australia
| | - Philip Batterham
- School of BioSciences, Bio21 Institute, The University of Melbourne, Parkville, Australia
| | - Trent Perry
- School of BioSciences, Bio21 Institute, The University of Melbourne, Parkville, Australia
| |
Collapse
|
153
|
Penke LRK, Speth J, Wettlaufer S, Draijer C, Peters-Golden M. Bortezomib Inhibits Lung Fibrosis and Fibroblast Activation Without Proteasome Inhibition. Am J Respir Cell Mol Biol 2021; 66:23-37. [PMID: 34236953 DOI: 10.1165/rcmb.2021-0112oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The FDA-approved proteasomal inhibitor bortezomib (BTZ) has attracted interest for its potential anti-fibrotic actions. However, neither its in vivo efficacy in lung fibrosis nor its dependence on proteasome inhibition has been conclusively defined. In this study, we assessed the therapeutic efficacy of BTZ in a mouse model of pulmonary fibrosis, developed an in vitro protocol to define its actions on diverse fibroblast activation parameters, determined its reliance on proteasome inhibition for these actions in vivo and in vitro and explored alternative mechanisms of action. The therapeutic administration of BTZ diminished the severity of pulmonary fibrosis without reducing proteasome activity in the lung. In experiments designed to mimic this lack of proteasome inhibition in vitro, BTZ reduced fibroblast proliferation, differentiation into myofibroblasts, and collagen synthesis. It promoted de-differentiation of myofibroblasts and overcame their characteristic resistance to apoptosis. Mechanistically, BTZ inhibited kinases important for fibroblast activation while inducing expression of dual-specificity phosphatase 1 or DUSP1, and knockdown of DUSP1 abolished its anti-fibrotic actions in fibroblasts. Collectively, these findings suggest that BTZ exhibits a multidimensional profile of robust inhibitory actions on lung fibroblasts as well as anti-fibrotic actions in vivo. Unexpectedly, these actions appear to be independent of proteasome inhibition, and instead attributable to induction of DUSP1.
Collapse
Affiliation(s)
| | - Jennifer Speth
- University of Michigan, 1259, Ann Arbor, Michigan, United States
| | - Scott Wettlaufer
- University of Michigan, 1259, Division of Pulmonary and Critical Care Medicine, Ann Arbor, Michigan, United States
| | | | - Marc Peters-Golden
- University of Michigan Health System, 21707, Ann Arbor, Michigan, United States;
| |
Collapse
|
154
|
Sorrentino VG, Thota S, Gonzalez EA, Rameshwar P, Chang VT, Etchegaray JP. Hypomethylating Chemotherapeutic Agents as Therapy for Myelodysplastic Syndromes and Prevention of Acute Myeloid Leukemia. Pharmaceuticals (Basel) 2021; 14:641. [PMID: 34358067 PMCID: PMC8308509 DOI: 10.3390/ph14070641] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
Myelodysplastic Syndromes (MDSs) affect the elderly and can progress to Acute Myeloid Leukemia (AML). Epigenetic alterations including DNA methylation and chromatin modification may contribute to the initiation and progression of these malignancies. DNA hypomethylating agents such as decitabine and azacitidine are used as therapeutic treatments and have shown to promote expression of genes involved in tumor suppression, apoptosis, and immune response. Another anti-cancer drug, the proteasome inhibitor bortezomib, is used as a chemotherapeutic treatment for multiple myeloma (MM). Phase III clinical trials of decitabine and azacitidine used alone and in combination with other chemotherapeutics demonstrated their capacity to treat hematological malignancies and prolong the survival of MDS and AML patients. Although phase III clinical trials examining bortezomib's role in MDS and AML patients are limited, its underlying mechanisms in MM highlight its potential as a chemotherapeutic for such malignancies. Further research is needed to better understand how the epigenetic mechanisms mediated by these chemotherapeutic agents and their targeted gene networks are associated with the development and progression of MDS into AML. This review discusses the mechanisms by which decitabine, azacitidine, and bortezomib alter epigenetic programs and their results from phase III clinical trials.
Collapse
Affiliation(s)
- Vincent G. Sorrentino
- Department of Biological Sciences, Rutgers University—Newark, Newark, NJ 07102, USA; (V.G.S.); (S.T.); (E.A.G.)
| | - Srijan Thota
- Department of Biological Sciences, Rutgers University—Newark, Newark, NJ 07102, USA; (V.G.S.); (S.T.); (E.A.G.)
| | - Edward A. Gonzalez
- Department of Biological Sciences, Rutgers University—Newark, Newark, NJ 07102, USA; (V.G.S.); (S.T.); (E.A.G.)
| | - Pranela Rameshwar
- Department of Medicine, Division of Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA;
| | - Victor T. Chang
- Department of Medicine, Division of Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA;
- Veteran Affairs New Jersey Health Care System, East Orange, NJ 07018, USA;
| | - Jean-Pierre Etchegaray
- Department of Biological Sciences, Rutgers University—Newark, Newark, NJ 07102, USA; (V.G.S.); (S.T.); (E.A.G.)
| |
Collapse
|
155
|
Sannino S, Yates ME, Schurdak ME, Oesterreich S, Lee AV, Wipf P, Brodsky JL. Unique integrated stress response sensors regulate cancer cell susceptibility when Hsp70 activity is compromised. eLife 2021; 10:64977. [PMID: 34180400 PMCID: PMC8275131 DOI: 10.7554/elife.64977] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 06/27/2021] [Indexed: 12/11/2022] Open
Abstract
Molecular chaperones, such as Hsp70, prevent proteotoxicity and maintain homeostasis. This is perhaps most evident in cancer cells, which overexpress Hsp70 and thrive even when harboring high levels of misfolded proteins. To define the response to proteotoxic challenges, we examined adaptive responses in breast cancer cells in the presence of an Hsp70 inhibitor. We discovered that the cells bin into distinct classes based on inhibitor sensitivity. Strikingly, the most resistant cells have higher autophagy levels, and autophagy was maximally activated only in resistant cells upon Hsp70 inhibition. In turn, resistance to compromised Hsp70 function required the integrated stress response transducer, GCN2, which is commonly associated with amino acid starvation. In contrast, sensitive cells succumbed to Hsp70 inhibition by activating PERK. These data reveal an unexpected route through which breast cancer cells adapt to proteotoxic insults and position GCN2 and autophagy as complementary mechanisms to ensure survival when proteostasis is compromised.
Collapse
Affiliation(s)
- Sara Sannino
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, United States
| | - Megan E Yates
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women Research Institute, Pittsburgh, United States.,Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, United States.,Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Mark E Schurdak
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, United States.,University of Pittsburgh Drug Discovery Institute, Pittsburgh, United States
| | - Steffi Oesterreich
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women Research Institute, Pittsburgh, United States.,Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, United States.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Adrian V Lee
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Magee-Women Research Institute, Pittsburgh, United States.,Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, United States.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, United States
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, United States
| |
Collapse
|
156
|
Immunoproteasome Function in Normal and Malignant Hematopoiesis. Cells 2021; 10:cells10071577. [PMID: 34206607 PMCID: PMC8305381 DOI: 10.3390/cells10071577] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) is a central part of protein homeostasis, degrading not only misfolded or oxidized proteins but also proteins with essential functions. The fact that a healthy hematopoietic system relies on the regulation of protein homeostasis and that alterations in the UPS can lead to malignant transformation makes the UPS an attractive therapeutic target for the treatment of hematologic malignancies. Herein, inhibitors of the proteasome, the last and most important component of the UPS enzymatic cascade, have been approved for the treatment of these malignancies. However, their use has been associated with side effects, drug resistance, and relapse. Inhibitors of the immunoproteasome, a proteasomal variant constitutively expressed in the cells of hematopoietic origin, could potentially overcome the encountered problems of non-selective proteasome inhibition. Immunoproteasome inhibitors have demonstrated their efficacy and safety against inflammatory and autoimmune diseases, even though their development for the treatment of hematologic malignancies is still in the early phases. Various immunoproteasome inhibitors have shown promising preliminary results in pre-clinical studies, and one inhibitor is currently being investigated in clinical trials for the treatment of multiple myeloma. Here, we will review data on immunoproteasome function and inhibition in hematopoietic cells and hematologic cancers.
Collapse
|
157
|
Baveloni FG, Riccio BVF, Di Filippo LD, Fernandes MA, Meneguin AB, Chorilli M. Nanotechnology-based Drug Delivery Systems as Potential for Skin Application: A Review. Curr Med Chem 2021; 28:3216-3248. [PMID: 32867631 DOI: 10.2174/0929867327666200831125656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/21/2020] [Accepted: 07/25/2020] [Indexed: 11/22/2022]
Abstract
Administration of substances through the skin represents a promising alternative, in relation to other drug administration routes, due to its large body surface area, in order to offer ideal and multiple sites for drug administration. In addition, the administration of drugs through the skin avoids the first-pass metabolism, allowing an increase in the bioavailability of drugs, as well as reducing their side effects. However, the stratum corneum (SC) comprises the main barrier of protection against external agents, mainly due to its structure, composition and physicochemical properties, becoming the main limitation for the administration of substances through the skin. In view of the above, pharmaceutical technology has allowed the development of multiple drug delivery systems (DDS), which include liquid crystals (LC), cubosomes, liposomes, polymeric nanoparticles (PNP), nanoemulsions (NE), as well as cyclodextrins (CD) and dendrimers (DND). It appears that the DDS circumvents the problems of drug absorption through the SC layer of the skin, ensuring the release of the drug, as well as optimizing the therapeutic effect locally. This review aims to highlight the DDS that include LC, cubosomes, lipid systems, PNP, as well as CD and DND, to optimize topical skin therapies.
Collapse
Affiliation(s)
- Franciele Garcia Baveloni
- School of Pharmaceutical Sciences, Department of Drugs and Medicines, Sao Paulo State University, Rodovia Araraquara-Jau, km 01, Araraquara, SP, CEP 14800-903, Brazil
| | - Bruno Vincenzo Fiod Riccio
- School of Pharmaceutical Sciences, Department of Drugs and Medicines, Sao Paulo State University, Rodovia Araraquara-Jau, km 01, Araraquara, SP, CEP 14800-903, Brazil
| | - Leonardo Delello Di Filippo
- School of Pharmaceutical Sciences, Department of Drugs and Medicines, Sao Paulo State University, Rodovia Araraquara-Jau, km 01, Araraquara, SP, CEP 14800-903, Brazil
| | - Mariza Aires Fernandes
- School of Pharmaceutical Sciences, Department of Drugs and Medicines, Sao Paulo State University, Rodovia Araraquara-Jau, km 01, Araraquara, SP, CEP 14800-903, Brazil
| | - Andréia Bagliotti Meneguin
- School of Pharmaceutical Sciences, Department of Drugs and Medicines, Sao Paulo State University, Rodovia Araraquara-Jau, km 01, Araraquara, SP, CEP 14800-903, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Sciences, Department of Drugs and Medicines, Sao Paulo State University, Rodovia Araraquara-Jau, km 01, Araraquara, SP, CEP 14800-903, Brazil
| |
Collapse
|
158
|
Small-Molecule Inhibitors Targeting Proteasome-Associated Deubiquitinases. Int J Mol Sci 2021; 22:ijms22126213. [PMID: 34207520 PMCID: PMC8226605 DOI: 10.3390/ijms22126213] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 02/06/2023] Open
Abstract
The 26S proteasome is the principal protease for regulated intracellular proteolysis. This multi-subunit complex is also pivotal for clearance of harmful proteins that are produced throughout the lifetime of eukaryotes. Recent structural and kinetic studies have revealed a multitude of conformational states of the proteasome in substrate-free and substrate-engaged forms. These conformational transitions demonstrate that proteasome is a highly dynamic machinery during substrate processing that can be also controlled by a number of proteasome-associated factors. Essentially, three distinct family of deubiquitinases–USP14, RPN11, and UCH37–are associated with the 19S regulatory particle of human proteasome. USP14 and UCH37 are capable of editing ubiquitin conjugates during the process of their dynamic engagement into the proteasome prior to the catalytic commitment. In contrast, RPN11-mediated deubiquitination is directly coupled to substrate degradation by sensing the proteasome’s conformational switch into the commitment steps. Therefore, proteasome-bound deubiquitinases are likely to tailor the degradation events in accordance with substrate processing steps and for dynamic proteolysis outcomes. Recent chemical screening efforts have yielded highly selective small-molecule inhibitors for targeting proteasomal deubiquitinases, such as USP14 and RPN11. USP14 inhibitors, IU1 and its progeny, were found to promote the degradation of a subset of substrates probably by overriding USP14-imposed checkpoint on the proteasome. On the other hand, capzimin, a RPN11 inhibitor, stabilized the proteasome substrates and showed the anti-proliferative effects on cancer cells. It is highly conceivable that these specific inhibitors will aid to dissect the role of each deubiquitinase on the proteasome. Moreover, customized targeting of proteasome-associated deubiquitinases may also provide versatile therapeutic strategies for induced or repressed protein degradation depending on proteolytic demand and cellular context.
Collapse
|
159
|
Łuczkowska K, Rogińska D, Ulańczyk Z, Safranow K, Paczkowska E, Baumert B, Milczarek S, Osękowska B, Górska M, Borowiecka E, Sommerfeld K, Zawodny P, Szudy-Szczyrek A, Hus M, Machaliński B. microRNAs as the biomarkers of chemotherapy-induced peripheral neuropathy in patients with multiple myeloma. Leuk Lymphoma 2021; 62:2768-2776. [PMID: 34092168 DOI: 10.1080/10428194.2021.1933478] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multiple myeloma (MM) is a malignant, incurable neoplastic disease. The currently used treatment significantly improves the prognosis and extends the survival time of patients. Unfortunately, a common side effect of the therapy is peripheral neuropathy, which may lead to dose reduction or complete treatment discontinuation/modification. In this study, we examined the changes in plasma levels of circulating miRNAs in myeloma patients to define potential factors characteristic for drug-induced peripheral neuropathy (DiPN). Global miRNA expression profile in the plasma of patients with MM during treatment was determined using miRNA microarray technology. Receiver operating characteristic (ROC) analysis allowed the identification of three miRNAs (miR-22-3p; miR-23a-3p; miR-24-3p) that could be a potential biomarker of PN. The most promising results were obtained for miR-22-3p, which was characterized by ROC area under curve (AUC) = 0.807. Our results suggest a relationship between the DiPN in patients with MM and the level of selected miRNAs in the plasma.
Collapse
Affiliation(s)
- Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Dorota Rogińska
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Zofia Ulańczyk
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | - Edyta Paczkowska
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland.,Department of Bone Marrow Transplantation, Pomeranian Medical University, Szczecin, Poland
| | - Bartłomiej Baumert
- Department of Bone Marrow Transplantation, Pomeranian Medical University, Szczecin, Poland
| | - Sławomir Milczarek
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland.,Department of Bone Marrow Transplantation, Pomeranian Medical University, Szczecin, Poland
| | - Bogumiła Osękowska
- Department of Bone Marrow Transplantation, Pomeranian Medical University, Szczecin, Poland
| | - Martyna Górska
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Ewa Borowiecka
- Department of Bone Marrow Transplantation, Pomeranian Medical University, Szczecin, Poland
| | - Krzysztof Sommerfeld
- Department of Bone Marrow Transplantation, Pomeranian Medical University, Szczecin, Poland
| | - Piotr Zawodny
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Aneta Szudy-Szczyrek
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| | - Marek Hus
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, Lublin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland.,Department of Bone Marrow Transplantation, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
160
|
Hernández ÁP, Juanes-Velasco P, Landeira-Viñuela A, Bareke H, Montalvillo E, Góngora R, Fuentes M. Restoring the Immunity in the Tumor Microenvironment: Insights into Immunogenic Cell Death in Onco-Therapies. Cancers (Basel) 2021; 13:2821. [PMID: 34198850 PMCID: PMC8201010 DOI: 10.3390/cancers13112821] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/31/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
Immunogenic cell death (ICD) elicited by cancer therapy reshapes the tumor immune microenvironment. A long-term adaptative immune response can be initiated by modulating cell death by therapeutic approaches. Here, the major hallmarks of ICD, endoplasmic reticulum (ER) stress, and damage-associated molecular patterns (DAMPs) are correlated with ICD inducers used in clinical practice to enhance antitumoral activity by suppressing tumor immune evasion. Approaches to monitoring the ICD triggered by antitumoral therapeutics in the tumor microenvironment (TME) and novel perspective in this immune system strategy are also reviewed to give an overview of the relevance of ICD in cancer treatment.
Collapse
Affiliation(s)
- Ángela-Patricia Hernández
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (Á.-P.H.); (P.J.-V.); (A.L.-V.); (H.B.); (E.M.); (R.G.)
| | - Pablo Juanes-Velasco
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (Á.-P.H.); (P.J.-V.); (A.L.-V.); (H.B.); (E.M.); (R.G.)
| | - Alicia Landeira-Viñuela
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (Á.-P.H.); (P.J.-V.); (A.L.-V.); (H.B.); (E.M.); (R.G.)
| | - Halin Bareke
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (Á.-P.H.); (P.J.-V.); (A.L.-V.); (H.B.); (E.M.); (R.G.)
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Institute of Health Sciences, Marmara University, 34722 Istanbul, Turkey
| | - Enrique Montalvillo
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (Á.-P.H.); (P.J.-V.); (A.L.-V.); (H.B.); (E.M.); (R.G.)
| | - Rafael Góngora
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (Á.-P.H.); (P.J.-V.); (A.L.-V.); (H.B.); (E.M.); (R.G.)
| | - Manuel Fuentes
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (Á.-P.H.); (P.J.-V.); (A.L.-V.); (H.B.); (E.M.); (R.G.)
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
161
|
Coghi PS, Zhu Y, Xie H, Hosmane NS, Zhang Y. Organoboron Compounds: Effective Antibacterial and Antiparasitic Agents. Molecules 2021; 26:3309. [PMID: 34072937 PMCID: PMC8199504 DOI: 10.3390/molecules26113309] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 11/16/2022] Open
Abstract
The unique electron deficiency and coordination property of boron led to a wide range of applications in chemistry, energy research, materials science and the life sciences. The use of boron-containing compounds as pharmaceutical agents has a long history, and recent developments have produced encouraging strides. Boron agents have been used for both radiotherapy and chemotherapy. In radiotherapy, boron neutron capture therapy (BNCT) has been investigated to treat various types of tumors, such as glioblastoma multiforme (GBM) of brain, head and neck tumors, etc. Boron agents playing essential roles in such treatments and other well-established areas have been discussed elsewhere. Organoboron compounds used to treat various diseases besides tumor treatments through BNCT technology have also marked an important milestone. Following the clinical introduction of bortezomib as an anti-cancer agent, benzoxaborole drugs, tavaborole and crisaborole, have been approved for clinical use in the treatments of onychomycosis and atopic dermatitis. Some heterocyclic organoboron compounds represent potentially promising candidates for anti-infective drugs. This review highlights the clinical applications and perspectives of organoboron compounds with the natural boron atoms in disease treatments without neutron irradiation. The main topic focuses on the therapeutic applications of organoboron compounds in the diseases of tuberculosis and antifungal activity, malaria, neglected tropical diseases and cryptosporidiosis and toxoplasmosis.
Collapse
Affiliation(s)
- Paolo Saul Coghi
- School of Pharmacy Macau, University of Science and Technology, Taipa Macau 999078, China;
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa Macau 999078, China
| | - Yinghuai Zhu
- The State Key Laboratory of Anti-Infective Drug Development (NO. 2015DQ780357), Sunshine Lake Pharma Co., Ltd., Dongguan 523871, China;
| | - Hongming Xie
- The State Key Laboratory of Anti-Infective Drug Development (NO. 2015DQ780357), Sunshine Lake Pharma Co., Ltd., Dongguan 523871, China;
| | - Narayan S. Hosmane
- Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, IL 60115, USA
| | - Yingjun Zhang
- The State Key Laboratory of Anti-Infective Drug Development (NO. 2015DQ780357), Sunshine Lake Pharma Co., Ltd., Dongguan 523871, China;
| |
Collapse
|
162
|
Devis-Jauregui L, Eritja N, Davis ML, Matias-Guiu X, Llobet-Navàs D. Autophagy in the physiological endometrium and cancer. Autophagy 2021; 17:1077-1095. [PMID: 32401642 PMCID: PMC8143243 DOI: 10.1080/15548627.2020.1752548] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 03/18/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a highly conserved catabolic process and a major cellular pathway for the degradation of long-lived proteins and cytoplasmic organelles. An increasing body of evidence has unveiled autophagy as an indispensable biological function that helps to maintain normal tissue homeostasis and metabolic fitness that can also lead to severe consequences for the normal cellular functioning when altered. Recent accumulating data point to autophagy as a key player in a wide variety of physiological and pathophysiological conditions in the human endometrium, one of the most proficient self-regenerating tissues in the human body and an instrumental player in placental species reproductive function. The current review highlights the most recent findings regarding the process of autophagy in the normal and cancerous endometrial tissue. Current research efforts aiming to therapeutically exploit autophagy and the methodological approaches used are discussed.Abbreviations: 3-MA: 3-methyladenine; ACACA (acetyl-CoA carboxylase alpha); AICAR: 5-aminoimidazole-4-carboximide riboside; AKT: AKT serine/threonine kinase; AMPK: AMP-activated protein kinase; ATG: autophagy related; ATG12: autophagy related 12; ATG16L1: autophagy related 16 like 1; ATG3: autophagy related 3; ATG4C: autophagy related 4C cysteine peptidase; ATG5: autophagy related 5; ATG7: autophagy related 7; ATG9: autophagy related 9; Baf A1: bafilomycin A1; BAX: BCL2 associated X, apoptosis regulator; BCL2: BCL2 apoptosis regulator; BECN1: beclin 1; CACNA1D: calcium voltage-gated channel subunit alpha1 D; CASP3: caspase 3; CASP7: caspase 7; CASP8: caspase 8; CASP9: caspase 9; CD44: CD44 molecule (Indian blood group); CDH1: cadherin 1; CDKN1A: cyclin dependent kinase inhibitor 1A; CDKN2A: cyclin dependent kinase inhibitor 2A; CMA: chaperone-mediated autophagy; CQ: chloroquine; CTNNB1: catenin beta 1; DDIT3: DNA damage inducible transcript 3; EC: endometrial cancer; EGFR: epidermal growth factor receptor; EH: endometrial hyperplasia; EIF4E: eukaryotic translation initiation factor 4E; EPHB2/ERK: EPH receptor B2; ER: endoplasmic reticulum; ERBB2: er-b2 receptor tyrosine kinase 2; ERVW-1: endogenous retrovirus group W member 1, envelope; ESR1: estrogen receptor 1; FSH: follicle-stimulating hormone; GCG/GLP1: glucagon; GFP: green fluorescent protein; GIP: gastric inhibitory polypeptide; GLP1R: glucagon-like peptide-1 receptor; GLS: glutaminase; H2AX: H2A.X variant histone; HIF1A: hypoxia inducible factor 1 alpha; HMGB1: high mobility group box 1; HOTAIR: HOX transcript antisense RNA; HSPA5: heat shock protein family A (HSP70) member 5; HSPA8: heat shock protein family A (HSP70) member 8; IGF1: insulin like growth factor 1; IL27: interleukin 27; INS: insulin; ISL: isoliquiritigenin; KRAS: KRAS proto-oncogene, GTPase; LAMP2: lysosomal-associated membrane protein 2; lncRNA: long-non-coding RNA; MAP1LC3A/LC3A: microtubule associated protein 1 light chain 3 alpha; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MAPK8: mitogen-activated protein kinase 8; MAPK9: mitogen-activated protein kinase 9; MPA: medroxyprogesterone acetate; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1; MTORC2: mechanistic target of rapamycin kinase complex 2; MYCBP: MYC-binding protein; NFE2L2: nuclear factor, erythroid 2 like 2; NFKB: nuclear factor kappa B; NFKBIA: NFKB inhibitor alpha; NK: natural killer; NR5A1: nuclear receptor subfamily 5 group A member 1; PARP1: poly(ADP-ribose) polymerase 1; PAX2: paired box 2; PDK1: pyruvate dehydrogenase kinase 1; PDX: patient-derived xenograft; PIK3C3/Vps34: phosphatidylinositol 3-kinase catalytic subunit type 3; PIK3CA: phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha; PIK3R1: phosphoinositide-3-kinase regulatory subunit 1; PIKFYVE: phosphoinositide kinase, FYVE-type zinc finger containing; PPD: protopanaxadiol; PRKCD: protein kinase C delta; PROM1/CD133: prominin 1; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; PTEN: phosphatase and tensin homolog; RB1CC1/FIP200: RB1 inducible coiled-coil 1; RFP: red fluorescent protein; RPS6KB1/S6K1: ribosomal protein S6 kinase B1; RSV: resveratrol; SGK1: serum/glucocorticoid regulated kinase 1; SGK3: serum/glucocorticoid regulated kinase family member 3; SIRT: sirtuin; SLS: stone-like structures; SMAD2: SMAD family member 2; SMAD3: SMAD family member 3; SQSTM1: sequestosome 1; TALEN: transcription activator-like effector nuclease; TGFBR2: transforming growth factor beta receptor 2; TP53: tumor protein p53; TRIB3: tribbles pseudokinase 3; ULK1: unc-51 like autophagy activating kinase 1; ULK4: unc-51 like kinase 4; VEGFA: vascular endothelial growth factor A; WIPI2: WD repeat domain, phosphoinositide interacting 2; XBP1: X-box binding protein 1; ZFYVE1: zinc finger FYVE domain containing 1.
Collapse
Affiliation(s)
- Laura Devis-Jauregui
- Laboratory of Precision Medicine, Oncobell Program. Bellvitge Biomedical Research Institute (IDIBELL), Gran via De l’Hospitalet, Barcelona, Spain
| | - Núria Eritja
- Department of Pathology-Hospital Universitari Arnau De Vilanova, Universitat De Lleida, IRBLLEIDA, CIBERONC, Lleida, Spain
| | - Meredith Leigh Davis
- Institute of Genetic Medicine-International Centre for Life, Newcastle University. Central Parkway, Newcastle upon Tyne, UK
| | - Xavier Matias-Guiu
- Laboratory of Precision Medicine, Oncobell Program. Bellvitge Biomedical Research Institute (IDIBELL), Gran via De l’Hospitalet, Barcelona, Spain
- Department of Pathology-Hospital Universitari Arnau De Vilanova, Universitat De Lleida, IRBLLEIDA, CIBERONC, Lleida, Spain
- Department of Pathology-Hospital, Universitari De Bellvitge, Barcelona, Spain
| | - David Llobet-Navàs
- Laboratory of Precision Medicine, Oncobell Program. Bellvitge Biomedical Research Institute (IDIBELL), Gran via De l’Hospitalet, Barcelona, Spain
| |
Collapse
|
163
|
Omi K, Matsuo Y, Ueda G, Aoyama Y, Kato T, Hayashi Y, Imafuji H, Saito K, Tsuboi K, Morimoto M, Ogawa R, Takahashi H, Takiguchi S. Escin inhibits angiogenesis by suppressing interleukin‑8 and vascular endothelial growth factor production by blocking nuclear factor‑κB activation in pancreatic cancer cell lines. Oncol Rep 2021; 45:55. [PMID: 33760162 PMCID: PMC7962110 DOI: 10.3892/or.2021.8006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/21/2020] [Indexed: 12/20/2022] Open
Abstract
Pancreatic cancer (PaCa) is one of the most aggressive types of cancer. Thus, the development of new and more effective therapies is urgently required. Escin, a pentacyclic triterpenoid from the horse chestnut, has been reported to exhibit antitumor potential by reducing cell proliferation and blocking the nuclear factor‑κB (NF‑κB) signaling pathway in several types of cancer. Our previous study reported that NF‑κB enhanced the secretion of interleukin (IL)‑8 and vascular endothelial growth factor (VEGF), thereby inducing angiogenesis in PaCa cell lines. In the present study, it was examined whether escin inhibited angiogenesis by blocking NF‑κB activation in PaCa. It was initially confirmed that escin, at concentrations >10 µM, significantly inhibited the proliferation of several PaCa cell lines. Next, using immunocytochemical staining, it was found that escin inhibited the nuclear translocation of NF‑κB. Furthermore, ELISA confirmed that NF‑κB activity in the escin‑treated PaCa cells was significantly inhibited and reverse transcription‑quantitative PCR showed that the mRNA expression levels of tumor necrosis factor‑α‑induced IL‑8 and VEGF were significantly suppressed following escin treatment in the PaCa cell lines. ELISA also showed that escin decreased the secretion of IL‑8 and VEGF from the PaCa cells. Furthermore, tube formation in immortalized human endothelial cells was inhibited following incubation with the supernatants from escin‑treated PaCa cells. These results indicated that escin inhibited angiogenesis by reducing the secretion of IL‑8 and VEGF by blocking NF‑κB activity in PaCa. In conclusion, escin could be used as a novel molecular therapy for PaCa.
Collapse
Affiliation(s)
- Kan Omi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Goro Ueda
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Yoshinaga Aoyama
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Tomokatsu Kato
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Yuichi Hayashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Hiroyuki Imafuji
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Kenta Saito
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Ken Tsuboi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Mamoru Morimoto
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Ryo Ogawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| |
Collapse
|
164
|
Liu L, Luan S, Zhang C, Wang R, Zhang Y, Zhang M, Sheng Q, Han G, Wang T, Song S. Encapsulation and pH-responsive release of bortezomib by dopamine grafted hyaluronate nanogels. Int J Biol Macromol 2021; 183:369-378. [PMID: 33932413 DOI: 10.1016/j.ijbiomac.2021.04.161] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/12/2021] [Accepted: 04/24/2021] [Indexed: 11/28/2022]
Abstract
Hydrophobic drugs loaded nanogels were always associated with low encapsulation efficiency and immature burst release. In this work, dopamine grafted hyaluronate nanogels were designed for bortezomib (BTZ), a hydrophobic anticancer drug and a proteasome inhibitor. It was found that there was a more efficient loading and pH-controlled release of BTZ due to the presence of dopamine groups on the skeleton of the nanogels. The drug loading content (DLC) were up to 8.58% as the nanogels modified with 29% dopamine, compared to the DLC of less than 1% for nanogels without dopamine modification. It was the pH-sensitive nature of the borated bonds between BTZ and catechol groups that endowed the pH-responsive release behavior of BTZ in vitro. In vitro study proved good biocompatibility and efficient cell uptake of the nanogels. In vivo anti-tumor experiments demonstrated that bortezomib loading into the nanogel significantly enhanced the therapeutic effect of the drug. After 14-day treatment, the average tumor volume of BTZ loaded nanogel group was reduced by 200% more than that of free BTZ group. Combined with CD44 receptor targeting ability of hyaluronate and the merits of nanogel, the catechol modified hyaluronate nanogel exhibited as an efficient chemotherapeutic formulation of BTZ for cancer treatment.
Collapse
Affiliation(s)
- Lei Liu
- Institute of Pharmacy, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Shujuan Luan
- Department of Health Service and Management, Henan Technician College of Medicine & Health, Kaifeng 475000, China
| | - Chunli Zhang
- Institute of Pharmacy, School of Pharmacy, Henan University, Kaifeng 475004, China.
| | - Rui Wang
- Institute of Pharmacy, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Yanan Zhang
- Institute of Pharmacy, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Mengying Zhang
- Institute of Pharmacy, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Qianli Sheng
- Institute of Pharmacy, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Guang Han
- Institute of Pharmacy, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Tianshun Wang
- Institute of Pharmacy, School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Shiyong Song
- Institute of Pharmacy, School of Pharmacy, Henan University, Kaifeng 475004, China.
| |
Collapse
|
165
|
Lenin S, Ponthier E, Scheer KG, Yeo ECF, Tea MN, Ebert LM, Oksdath Mansilla M, Poonnoose S, Baumgartner U, Day BW, Ormsby RJ, Pitson SM, Gomez GA. A Drug Screening Pipeline Using 2D and 3D Patient-Derived In Vitro Models for Pre-Clinical Analysis of Therapy Response in Glioblastoma. Int J Mol Sci 2021; 22:4322. [PMID: 33919246 PMCID: PMC8122466 DOI: 10.3390/ijms22094322] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma is one of the most common and lethal types of primary brain tumor. Despite aggressive treatment with chemotherapy and radiotherapy, tumor recurrence within 6-9 months is common. To overcome this, more effective therapies targeting cancer cell stemness, invasion, metabolism, cell death resistance and the interactions of tumor cells with their surrounding microenvironment are required. In this study, we performed a systematic review of the molecular mechanisms that drive glioblastoma progression, which led to the identification of 65 drugs/inhibitors that we screened for their efficacy to kill patient-derived glioma stem cells in two dimensional (2D) cultures and patient-derived three dimensional (3D) glioblastoma explant organoids (GBOs). From the screening, we found a group of drugs that presented different selectivity on different patient-derived in vitro models. Moreover, we found that Costunolide, a TERT inhibitor, was effective in reducing the cell viability in vitro of both primary tumor models as well as tumor models pre-treated with chemotherapy and radiotherapy. These results present a novel workflow for screening a relatively large groups of drugs, whose results could lead to the identification of more personalized and effective treatment for recurrent glioblastoma.
Collapse
Affiliation(s)
- Sakthi Lenin
- Centre for Cancer Biology, SA Pathology and the University of South of Australia, Adelaide, SA 5000, Australia; (S.L.); (E.P.); (K.G.S.); (E.C.F.Y.); (M.N.T.); (L.M.E.); (M.O.M.); (S.M.P.)
| | - Elise Ponthier
- Centre for Cancer Biology, SA Pathology and the University of South of Australia, Adelaide, SA 5000, Australia; (S.L.); (E.P.); (K.G.S.); (E.C.F.Y.); (M.N.T.); (L.M.E.); (M.O.M.); (S.M.P.)
| | - Kaitlin G. Scheer
- Centre for Cancer Biology, SA Pathology and the University of South of Australia, Adelaide, SA 5000, Australia; (S.L.); (E.P.); (K.G.S.); (E.C.F.Y.); (M.N.T.); (L.M.E.); (M.O.M.); (S.M.P.)
| | - Erica C. F. Yeo
- Centre for Cancer Biology, SA Pathology and the University of South of Australia, Adelaide, SA 5000, Australia; (S.L.); (E.P.); (K.G.S.); (E.C.F.Y.); (M.N.T.); (L.M.E.); (M.O.M.); (S.M.P.)
| | - Melinda N. Tea
- Centre for Cancer Biology, SA Pathology and the University of South of Australia, Adelaide, SA 5000, Australia; (S.L.); (E.P.); (K.G.S.); (E.C.F.Y.); (M.N.T.); (L.M.E.); (M.O.M.); (S.M.P.)
| | - Lisa M. Ebert
- Centre for Cancer Biology, SA Pathology and the University of South of Australia, Adelaide, SA 5000, Australia; (S.L.); (E.P.); (K.G.S.); (E.C.F.Y.); (M.N.T.); (L.M.E.); (M.O.M.); (S.M.P.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Mariana Oksdath Mansilla
- Centre for Cancer Biology, SA Pathology and the University of South of Australia, Adelaide, SA 5000, Australia; (S.L.); (E.P.); (K.G.S.); (E.C.F.Y.); (M.N.T.); (L.M.E.); (M.O.M.); (S.M.P.)
| | - Santosh Poonnoose
- Flinders Health and Medical Research Institute, College of Medicine & Public Health, Flinders University, Adelaide, SA 5042, Australia; (S.P.); (R.J.O.)
- Department of Neurosurgery, Flinders Medical Centre, Adelaide, SA 5042, Australia
| | - Ulrich Baumgartner
- Cell and Molecular Biology Department, Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (U.B.); (B.W.D.)
- Faculty of Health, Queensland University of Technology, Brisbane, QLD 4006, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
| | - Bryan W. Day
- Cell and Molecular Biology Department, Sid Faithfull Brain Cancer Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; (U.B.); (B.W.D.)
- Faculty of Health, Queensland University of Technology, Brisbane, QLD 4006, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
| | - Rebecca J. Ormsby
- Flinders Health and Medical Research Institute, College of Medicine & Public Health, Flinders University, Adelaide, SA 5042, Australia; (S.P.); (R.J.O.)
| | - Stuart M. Pitson
- Centre for Cancer Biology, SA Pathology and the University of South of Australia, Adelaide, SA 5000, Australia; (S.L.); (E.P.); (K.G.S.); (E.C.F.Y.); (M.N.T.); (L.M.E.); (M.O.M.); (S.M.P.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
| | - Guillermo A. Gomez
- Centre for Cancer Biology, SA Pathology and the University of South of Australia, Adelaide, SA 5000, Australia; (S.L.); (E.P.); (K.G.S.); (E.C.F.Y.); (M.N.T.); (L.M.E.); (M.O.M.); (S.M.P.)
| |
Collapse
|
166
|
Xiong C, Zhou L, Tan J, Song S, Bao X, Zhang N, Ding H, Zhao J, He JX, Miao ZH, Zhang A. Development of Potent NEDD8-Activating Enzyme Inhibitors Bearing a Pyrimidotriazole Scaffold. J Med Chem 2021; 64:6161-6178. [PMID: 33857374 DOI: 10.1021/acs.jmedchem.1c00242] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The ubiquitin-like protein NEDD8 is a critical signaling molecule implicated in the functional maintenance and homeostasis of cells. Dysregulation of this process is involved in a variety of human diseases, including cancer. Therefore, NEDD8-activating enzyme E1 (NAE), the only activation enzyme of the neddylation pathway, has been an emergent anticancer target. In view of the single-agent modest response of the clinical NAE inhibitor, pevonedistat (compound 1, MLN4924), efforts on development of new inhibitors with both high potency and better safety profiles are urgently needed. Here, we report a structural hopping strategy by optimizing the central deazapurine framework and the solvent interaction region of compound 1, leading to compound 26 bearing a pyrimidotriazole scaffold. Compound 26 not only has compatible potency in the biochemical and cell assays but also possesses improved pharmacokinetic (PK) properties than compound 1. In vivo, compound 26 showed significant antitumor efficacy and good safety in xenograft models.
Collapse
Affiliation(s)
- Chaodong Xiong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai 201203, China.,Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lina Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai 201203, China.,Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shanshan Song
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xubin Bao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ning Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huaqian Ding
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiannan Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai 201203, China.,Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jin-Xue He
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ze-Hong Miao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ao Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai 201203, China.,Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Ministry of Education of China, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
167
|
Long MJC, Kulkarni A, Aye Y. Can Precision Electrophile Signaling Make a Meaningful and Lasting Impression in Drug Design? Chembiochem 2021; 23:e202100051. [PMID: 33826211 DOI: 10.1002/cbic.202100051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/04/2021] [Indexed: 12/14/2022]
Abstract
For several years, drugs with reactive electrophilic appendages have been developed. These units typically confer prolonged residence time of the drugs on their protein targets, and may assist targeting shallow binding sites and/or improving the drug-protein target spectrum. Studies on natural electrophilic molecules have indicated that, in many instances, natural electrophiles use similar mechanisms to alter signaling pathways. However, natural reactive species are also endowed with other important mechanisms to hone signaling properties that are uncommon in drug design. These include ability to be active at low occupancy and elevated inhibitor kinetics. Herein, we discuss how we have begun to harness these properties in inhibitor design.
Collapse
Affiliation(s)
- Marcus J C Long
- University of Lausanne, Department of Biochemistry, Chemin des boveresses 155, Epalinges, 1066, Lausanne, Switzerland
| | - Amogh Kulkarni
- Swiss Federal Institute of Technology in Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Yimon Aye
- Swiss Federal Institute of Technology in Lausanne (EPFL), 1015, Lausanne, Switzerland
| |
Collapse
|
168
|
Tian W, Maresh ME, Trader DJ. Approaches to Evaluate the Impact of a Small-Molecule Binder to a Noncatalytic Site of the Proteasome. Chembiochem 2021; 22:1961-1965. [PMID: 33617657 DOI: 10.1002/cbic.202100023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/11/2021] [Indexed: 11/11/2022]
Abstract
Proteasome activity is crucial for cell survival and proliferation. In recent years, small molecules have been discovered that can affect the catalytic activity of the proteasome. Rather than targeting the active sites of the proteasome, it might be possible to affect ubiquitin-dependent degradation of proteins by limiting the association of the 19S regulatory particle (19S RP) with the 20S core particle (20S CP) of the proteasome. We recently described the discovery of TXS-8, a peptoid that binds to Rpn-6. Rpn-6 is a proteasome-associated protein that makes critical contacts with the 19S RP and the 20S CP. Herein, we present a general workflow to evaluate the impact of a small-molecule binder on proteasome activity by using TXS-8 as an example. This workflow contains three steps in which specific probes or overexpressed proteins in cells are used to determine whether the hydrolysis activity of the proteasome is affected. Although, in our case, TXS-8 did not affect proteasome activity, our workflow is highly amenable to studying a variety of small-molecule-proteasome subunit interactions.
Collapse
Affiliation(s)
- Wenzhi Tian
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, 47907, USA
| | - Marianne E Maresh
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, 47907, USA
| | - Darci J Trader
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, 47907, USA
| |
Collapse
|
169
|
Deesrisak K, Yingchutrakul Y, Krobthong S, Roytrakul S, Chatupheeraphat C, Subkorn P, Anurathapan U, Tanyong D. Bioactive peptide isolated from sesame seeds inhibits cell proliferation and induces apoptosis and autophagy in leukemic cells. EXCLI JOURNAL 2021; 20:709-721. [PMID: 33907539 PMCID: PMC8073838 DOI: 10.17179/excli2021-3406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/17/2021] [Indexed: 12/24/2022]
Abstract
Leukemia is the most common type of hematological malignancies. Several natural products including bioactive peptides have been explored and studied for their anti-leukemic activities. In the present study, anti-leukemic peptide, IGTLILM (IM-7), was isolated and identified from the protein hydrolysate of sesame seeds by reverse phase-solid phase extraction, off-gel fractionation and nano LC-MS/MS. The cytotoxic effects of IM-7 were studied in MOLT-4 and NB4 acute leukemic cell lines using an MTT assay. The induction of apoptosis and autophagy was investigated by flow cytometry using Annexin V-FITC/PI staining and anti-LC3/FITC antibodies, respectively. The mRNA alterations of apoptotic and autophagic-related genes were determined by reverse transcription-quantitative PCR. The present study found that IM-7 inhibited the proliferation of MOLT-4 and NB4 cells in dose-dependent manner, but it showed a minimal effect on healthy mononuclear cells. IM-7 activated apoptosis and autophagy through the upregulation of CASP3, ULK1 and BECN1 and the downregulation of BCL2. In addition, IM-7 enhanced the cytotoxic effect of the anti-leukemic drug, daunorubicin. The findings suggested that IM-7 was potent to suppress the proliferation of MOLT-4 and NB4 leukemic cells and induce apoptosis and autophagy through the regulation of caspase 3-Bcl-2 and ULK1-Beclin1, respectively.
Collapse
Affiliation(s)
- Kamolchanok Deesrisak
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Yodying Yingchutrakul
- Proteomics Research Team, National Omics Center, National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Sucheewin Krobthong
- Proteomics Research Team, National Omics Center, National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Sittiruk Roytrakul
- Functional Proteomics Technology Laboratory, Functional Ingredients and FoodInnovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology for Development Agency, Pathum Thani 12120,Thailand
| | - Chawalit Chatupheeraphat
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Paweena Subkorn
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Usanarat Anurathapan
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Dalina Tanyong
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand
| |
Collapse
|
170
|
Vozandychova V, Stojkova P, Hercik K, Rehulka P, Stulik J. The Ubiquitination System within Bacterial Host-Pathogen Interactions. Microorganisms 2021; 9:638. [PMID: 33808578 PMCID: PMC8003559 DOI: 10.3390/microorganisms9030638] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/17/2022] Open
Abstract
Ubiquitination of proteins, like phosphorylation and acetylation, is an important regulatory aspect influencing numerous and various cell processes, such as immune response signaling and autophagy. The study of ubiquitination has become essential to learning about host-pathogen interactions, and a better understanding of the detailed mechanisms through which pathogens affect ubiquitination processes in host cell will contribute to vaccine development and effective treatment of diseases. Pathogenic bacteria (e.g., Salmonella enterica, Legionella pneumophila and Shigella flexneri) encode many effector proteins, such as deubiquitinating enzymes (DUBs), targeting the host ubiquitin machinery and thus disrupting pertinent ubiquitin-dependent anti-bacterial response. We focus here upon the host ubiquitination system as an integral unit, its interconnection with the regulation of inflammation and autophagy, and primarily while examining pathogens manipulating the host ubiquitination system. Many bacterial effector proteins have already been described as being translocated into the host cell, where they directly regulate host defense processes. Due to their importance in pathogenic bacteria progression within the host, they are regarded as virulence factors essential for bacterial evasion. However, in some cases (e.g., Francisella tularensis) the host ubiquitination system is influenced by bacterial infection, although the responsible bacterial effectors are still unknown.
Collapse
Affiliation(s)
- Vera Vozandychova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 50001 Hradec Kralove, Czech Republic; (V.V.); (P.S.); (K.H.); (P.R.)
| | - Pavla Stojkova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 50001 Hradec Kralove, Czech Republic; (V.V.); (P.S.); (K.H.); (P.R.)
| | - Kamil Hercik
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 50001 Hradec Kralove, Czech Republic; (V.V.); (P.S.); (K.H.); (P.R.)
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, 16000 Prague, Czech Republic
| | - Pavel Rehulka
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 50001 Hradec Kralove, Czech Republic; (V.V.); (P.S.); (K.H.); (P.R.)
| | - Jiri Stulik
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Trebesska 1575, 50001 Hradec Kralove, Czech Republic; (V.V.); (P.S.); (K.H.); (P.R.)
| |
Collapse
|
171
|
Lopes R, Ferreira BV, Caetano J, Barahona F, Carneiro EA, João C. Boosting Immunity against Multiple Myeloma. Cancers (Basel) 2021; 13:1221. [PMID: 33799565 PMCID: PMC8001641 DOI: 10.3390/cancers13061221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/12/2021] [Accepted: 03/01/2021] [Indexed: 01/10/2023] Open
Abstract
Despite the improvement of patient's outcome obtained by the current use of immunomodulatory drugs, proteasome inhibitors or anti-CD38 monoclonal antibodies, multiple myeloma (MM) remains an incurable disease. More recently, the testing in clinical trials of novel drugs such as anti-BCMA CAR-T cells, antibody-drug conjugates or bispecific antibodies broadened the possibility of improving patients' survival. However, thus far, these treatment strategies have not been able to steadily eliminate all malignant cells, and the aim has been to induce a long-term complete response with minimal residual disease (MRD)-negative status. In this sense, approaches that target not only myeloma cells but also the surrounding microenvironment are promising strategies to achieve a sustained MRD negativity with prolonged survival. This review provides an overview of current and future strategies used for immunomodulation of MM focusing on the impact on bone marrow (BM) immunome.
Collapse
Affiliation(s)
- Raquel Lopes
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medicine, University of Lisbon, 1649-028 Lisbon, Portugal
| | - Bruna Velosa Ferreira
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
| | - Joana Caetano
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
- Hemato-Oncology Department, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Filipa Barahona
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
| | - Emilie Arnault Carneiro
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
| | - Cristina João
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
- Hemato-Oncology Department, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| |
Collapse
|
172
|
Vizovisek M, Ristanovic D, Menghini S, Christiansen MG, Schuerle S. The Tumor Proteolytic Landscape: A Challenging Frontier in Cancer Diagnosis and Therapy. Int J Mol Sci 2021; 22:ijms22052514. [PMID: 33802262 PMCID: PMC7958950 DOI: 10.3390/ijms22052514] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
In recent decades, dysregulation of proteases and atypical proteolysis have become increasingly recognized as important hallmarks of cancer, driving community-wide efforts to explore the proteolytic landscape of oncologic disease. With more than 100 proteases currently associated with different aspects of cancer development and progression, there is a clear impetus to harness their potential in the context of oncology. Advances in the protease field have yielded technologies enabling sensitive protease detection in various settings, paving the way towards diagnostic profiling of disease-related protease activity patterns. Methods including activity-based probes and substrates, antibodies, and various nanosystems that generate reporter signals, i.e., for PET or MRI, after interaction with the target protease have shown potential for clinical translation. Nevertheless, these technologies are costly, not easily multiplexed, and require advanced imaging technologies. While the current clinical applications of protease-responsive technologies in oncologic settings are still limited, emerging technologies and protease sensors are poised to enable comprehensive exploration of the tumor proteolytic landscape as a diagnostic and therapeutic frontier. This review aims to give an overview of the most relevant classes of proteases as indicators for tumor diagnosis, current approaches to detect and monitor their activity in vivo, and associated therapeutic applications.
Collapse
|
173
|
The paradoxical pharmacological mechanisms of lenalidomide and bortezomib in the treatment of multiple myeloma. Anticancer Drugs 2021; 32:227-232. [PMID: 33534410 DOI: 10.1097/cad.0000000000001041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The combination of bortezomib (Velcade, PS-341) and lenalidomide (Revlimid) for the treatment of multiple myeloma was proved by USA Food and Drug Administration in 2006. Lenalidomide prevents the proliferation of multiple myeloma cells through binding to cereblon and promoting the ubiquitinational degradation of IKZF1 (Ikaros)/IKZF3 (Aiolos). However, the proteasome inhibitor bortezomib would inhibit the ubiquitinational degradation of IKZF1/IKZF3. How bortezomib could not block the antiproliferative effect of lenalidomide on multiple myeloma cells, which is the paradoxical pharmacological mechanisms in multiple myeloma. In this review, we summarized recent advances in molecular mechanisms underlying the combination of bortezomib and lenalidomide for the treatment multiple myeloma, discussed the paradoxical pharmacological mechanisms of lenalidomide and bortezomib in the treatment of multiple myeloma.
Collapse
|
174
|
Joutsen J, Da Silva AJ, Luoto JC, Budzynski MA, Nylund AS, de Thonel A, Concordet JP, Mezger V, Sabéran-Djoneidi D, Henriksson E, Sistonen L. Heat Shock Factor 2 Protects against Proteotoxicity by Maintaining Cell-Cell Adhesion. Cell Rep 2021; 30:583-597.e6. [PMID: 31940498 DOI: 10.1016/j.celrep.2019.12.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/15/2019] [Accepted: 12/12/2019] [Indexed: 12/13/2022] Open
Abstract
Maintenance of protein homeostasis, through inducible expression of molecular chaperones, is essential for cell survival under protein-damaging conditions. The expression and DNA-binding activity of heat shock factor 2 (HSF2), a member of the heat shock transcription factor family, increase upon exposure to prolonged proteotoxicity. Nevertheless, the specific roles of HSF2 and the global HSF2-dependent gene expression profile during sustained stress have remained unknown. Here, we found that HSF2 is critical for cell survival during prolonged proteotoxicity. Strikingly, our RNA sequencing (RNA-seq) analyses revealed that impaired viability of HSF2-deficient cells is not caused by inadequate induction of molecular chaperones but is due to marked downregulation of cadherin superfamily genes. We demonstrate that HSF2-dependent maintenance of cadherin-mediated cell-cell adhesion is required for protection against stress induced by proteasome inhibition. This study identifies HSF2 as a key regulator of cadherin superfamily genes and defines cell-cell adhesion as a determinant of proteotoxic stress resistance.
Collapse
Affiliation(s)
- Jenny Joutsen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland
| | - Alejandro Jose Da Silva
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland
| | - Jens Christian Luoto
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland
| | - Marek Andrzej Budzynski
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland
| | - Anna Serafia Nylund
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland
| | - Aurelie de Thonel
- CNRS, UMR 7216 "Epigenetic and Cell Fate," 75250 Paris Cedex 13, France; University of Paris Diderot, Sorbonne Paris Cité, 75250 Paris Cedex 13, France; Département Hospitalo-Universitaire DHU PROTECT, Paris, France
| | - Jean-Paul Concordet
- INSERM U1154, CNRS UMR 7196, Muséum National d'Histoire Naturelle, Paris, France
| | - Valérie Mezger
- CNRS, UMR 7216 "Epigenetic and Cell Fate," 75250 Paris Cedex 13, France; University of Paris Diderot, Sorbonne Paris Cité, 75250 Paris Cedex 13, France; Département Hospitalo-Universitaire DHU PROTECT, Paris, France
| | - Délara Sabéran-Djoneidi
- CNRS, UMR 7216 "Epigenetic and Cell Fate," 75250 Paris Cedex 13, France; University of Paris Diderot, Sorbonne Paris Cité, 75250 Paris Cedex 13, France; Département Hospitalo-Universitaire DHU PROTECT, Paris, France
| | - Eva Henriksson
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland
| | - Lea Sistonen
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, 20520 Turku, Finland.
| |
Collapse
|
175
|
Zhang X, Meng T, Cui S, Feng L, Liu D, Pang Q, Wang P. Ubiquitination of Nonhistone Proteins in Cancer Development and Treatment. Front Oncol 2021; 10:621294. [PMID: 33643919 PMCID: PMC7905169 DOI: 10.3389/fonc.2020.621294] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 12/24/2020] [Indexed: 12/14/2022] Open
Abstract
Ubiquitination, a crucial post-translation modification, regulates the localization and stability of the substrate proteins including nonhistone proteins. The ubiquitin-proteasome system (UPS) on nonhistone proteins plays a critical role in many cellular processes such as DNA repair, transcription, signal transduction, and apoptosis. Its dysregulation induces various diseases including cancer, and the identification of this process may provide potential therapeutic targets for cancer treatment. In this review, we summarize the regulatory roles of key UPS members on major nonhistone substrates in cancer-related processes, such as cell cycle, cell proliferation, apoptosis, DNA damage repair, inflammation, and T cell dysfunction in cancer. In addition, we also highlight novel therapeutic interventions targeting the UPS members (E1s, E2s, E3s, proteasomes, and deubiquitinating enzymes). Furthermore, we discuss the application of proteolysis-targeting chimeras (PROTACs) technology as a novel anticancer therapeutic strategy in modulating protein target levels with the aid of UPS.
Collapse
Affiliation(s)
- Xiuzhen Zhang
- School of Life Sciences, Shandong University of Technology, Zibo, China
| | - Tong Meng
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Shuaishuai Cui
- School of Life Sciences, Shandong University of Technology, Zibo, China
| | - Ling Feng
- School of Life Sciences, Shandong University of Technology, Zibo, China
| | - Dongwu Liu
- School of Life Sciences, Shandong University of Technology, Zibo, China
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, China
| | - Qiuxiang Pang
- School of Life Sciences, Shandong University of Technology, Zibo, China
| | - Ping Wang
- School of Life Sciences, Shandong University of Technology, Zibo, China
| |
Collapse
|
176
|
Kim BJ, Fang Y, He H, Xu B. Trypsin-Instructed Self-Assembly on Endoplasmic Reticulum for Selectively Inhibiting Cancer Cells: Dedicated to Professor George M. Whitesides on the occasion of his 80th birthday. Adv Healthc Mater 2021; 10:e2000416. [PMID: 32342647 PMCID: PMC7725443 DOI: 10.1002/adhm.202000416] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/07/2020] [Indexed: 12/15/2022]
Abstract
Selectively targeting the endoplasmic reticulum (ER) of cancer cells, though promising a new strategy for cancer therapy, remains underdeveloped. Enzyme-instructed self-assembly (EISA) is emerging as a promising approach for selectively targeting ER of cancer cells. This work reports an easily accessible branched peptide that consists of a D-tetrapeptide backbone and a branch with the sequence of KYDKKKKDG (K: lysine; Y: tyrosine; D: aspratic acid; G: glycine), being an EISA substrate of typsin-1 (PRSS1), selectively inhibits cancer cells. Depending on the type of cells, the level of PRSS1 expression dictates the cytotoxicity of the branched peptide. Moreover, immunostaining and fluorescent imaging reveal that PRSS1 overexpresses on the ER of a high-grade serous ovarian cancer cell line (OVSAHO). The overexpression of PRSS1 renders the branched peptide to exhibit high selectivity against OVSAHO by the in situ formation of the peptide assemblies on the ER of OVSAHO cells, which causes ER stress and eventual cell death. This work, illustrating trypsin-guided EISA for inhibiting cancer cells by enzymatic reaction on ER for the first time, offers a new way to target the subcellular organelles of cancer cells for potential cancer therapy.
Collapse
Affiliation(s)
- Beom Jin Kim
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Yu Fang
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Hongjian He
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| | - Bing Xu
- Department of Chemistry, Brandeis University, 415 South Street, Waltham, MA, 02453, USA
| |
Collapse
|
177
|
Santopolo S, Riccio A, Rossi A, Santoro MG. The proteostasis guardian HSF1 directs the transcription of its paralog and interactor HSF2 during proteasome dysfunction. Cell Mol Life Sci 2021; 78:1113-1129. [PMID: 32607595 PMCID: PMC11071745 DOI: 10.1007/s00018-020-03568-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 05/03/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023]
Abstract
Protein homeostasis is essential for life in eukaryotes. Organisms respond to proteotoxic stress by activating heat shock transcription factors (HSFs), which play important roles in cytoprotection, longevity and development. Of six human HSFs, HSF1 acts as a proteostasis guardian regulating stress-induced transcriptional responses, whereas HSF2 has a critical role in development, in particular of brain and reproductive organs. Unlike HSF1, that is a stable protein constitutively expressed, HSF2 is a labile protein and its expression varies in different tissues; however, the mechanisms regulating HSF2 expression remain poorly understood. Herein we demonstrate that the proteasome inhibitor anticancer drug bortezomib (Velcade), at clinically relevant concentrations, triggers de novo HSF2 mRNA transcription in different types of cancers via HSF1 activation. Similar results were obtained with next-generation proteasome inhibitors ixazomib and carfilzomib, indicating that induction of HSF2 expression is a general response to proteasome dysfunction. HSF2-promoter analysis, electrophoretic mobility shift assays, and chromatin immunoprecipitation studies unexpectedly revealed that HSF1 is recruited to a heat shock element located at 1.397 bp upstream from the transcription start site in the HSF2-promoter. More importantly, we found that HSF1 is critical for HSF2 gene transcription during proteasome dysfunction, representing an interesting example of transcription factor involved in controlling the expression of members of the same family. Moreover, bortezomib-induced HSF2 was found to localize in the nucleus, interact with HSF1, and participate in bortezomib-mediated control of cancer cell migration. The results shed light on HSF2-expression regulation, revealing a novel level of HSF1/HSF2 interplay that may lead to advances in pharmacological modulation of these fundamental transcription factors.
Collapse
Affiliation(s)
- Silvia Santopolo
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Anna Riccio
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy
| | - Antonio Rossi
- Institute of Translational Pharmacology, CNR, Rome, Italy
| | - M Gabriella Santoro
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133, Rome, Italy.
- Institute of Translational Pharmacology, CNR, Rome, Italy.
| |
Collapse
|
178
|
Saitoh T, Oda T. DNA Damage Response in Multiple Myeloma: The Role of the Tumor Microenvironment. Cancers (Basel) 2021; 13:504. [PMID: 33525741 PMCID: PMC7865954 DOI: 10.3390/cancers13030504] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy characterized by genomic instability. MM cells present various forms of genetic instability, including chromosomal instability, microsatellite instability, and base-pair alterations, as well as changes in chromosome number. The tumor microenvironment and an abnormal DNA repair function affect genetic instability in this disease. In addition, states of the tumor microenvironment itself, such as inflammation and hypoxia, influence the DNA damage response, which includes DNA repair mechanisms, cell cycle checkpoints, and apoptotic pathways. Unrepaired DNA damage in tumor cells has been shown to exacerbate genomic instability and aberrant features that enable MM progression and drug resistance. This review provides an overview of the DNA repair pathways, with a special focus on their function in MM, and discusses the role of the tumor microenvironment in governing DNA repair mechanisms.
Collapse
Affiliation(s)
- Takayuki Saitoh
- Department of Laboratory Sciences, Graduate School of Health Sciences, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Tsukasa Oda
- Laboratory of Molecular Genetics, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma 371-8512, Japan;
| |
Collapse
|
179
|
Lin KM, Nightingale K, Soday L, Antrobus R, Weekes MP. Rapid Degradation Pathways of Host Proteins During HCMV Infection Revealed by Quantitative Proteomics. Front Cell Infect Microbiol 2021; 10:578259. [PMID: 33585265 PMCID: PMC7873559 DOI: 10.3389/fcimb.2020.578259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/08/2020] [Indexed: 12/30/2022] Open
Abstract
Human cytomegalovirus (HCMV) is an important pathogen in immunocompromised individuals and neonates, and a paradigm for viral immune evasion. We previously developed a quantitative proteomic approach that identified 133 proteins degraded during the early phase of HCMV infection, including known and novel antiviral factors. The majority were rescued from degradation by MG132, which is known to inhibit lysosomal cathepsins in addition to the proteasome. Global definition of the precise mechanisms of host protein degradation is important both to improve our understanding of viral biology, and to inform novel antiviral therapeutic strategies. We therefore developed and optimized a multiplexed comparative proteomic analysis using the selective proteasome inhibitor bortezomib in addition to MG132, to provide a global mechanistic view of protein degradation. Of proteins rescued from degradation by MG132, 34-47 proteins were also rescued by bortezomib, suggesting both that the predominant mechanism of protein degradation employed by HCMV is via the proteasome, and that alternative pathways for degradation are nevertheless important. Our approach and data will enable improved mechanistic understanding of HCMV and other viruses, and provide a shortlist of candidate restriction factors for further analysis.
Collapse
Affiliation(s)
| | | | | | | | - Michael P. Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
180
|
Lai D, Ghosh S, Hajra A. Light-induced borylation: developments and mechanistic insights. Org Biomol Chem 2021; 19:4397-4428. [PMID: 33913460 DOI: 10.1039/d1ob00323b] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Organoboron compounds are very important derivatives because of their profound impacts on medicinal, biological as well as industrial applications. The development of several novel borylation methodologies has achieved momentous interest among synthetic chemists. In this scenario, eco-friendly light-induced borylation is progressively becoming one of the best synthetic tools in recent days to prepare organoboronic ester and acid derivatives based on green chemistry rules. In this article, we have discussed all the UV- and visible-light-induced borylation strategies developed in the last decade. Furthermore, special attention is given to the mechanisms of these borylation methodologies for better understanding of reaction insights.
Collapse
Affiliation(s)
- Dipti Lai
- Department of Chemistry, Visva-Bharati (A Central University), Santiniketan 731235, India.
| | - Sumit Ghosh
- Department of Chemistry, Visva-Bharati (A Central University), Santiniketan 731235, India.
| | - Alakananda Hajra
- Department of Chemistry, Visva-Bharati (A Central University), Santiniketan 731235, India.
| |
Collapse
|
181
|
Maresh ME, Salazar-Chaparro AF, Trader DJ. Methods for the discovery of small molecules to monitor and perturb the activity of the human proteasome. Future Med Chem 2021; 13:99-116. [PMID: 33275045 PMCID: PMC7857359 DOI: 10.4155/fmc-2020-0288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Regulating protein production and degradation is critical to maintaining cellular homeostasis. The proteasome is a key player in keeping proteins at the proper levels. However, proteasome activity can be altered in certain disease states, such as blood cancers and neurodegenerative diseases. Cancers often exhibit enhanced proteasomal activity, as protein synthesis is increased in these cells compared with normal cells. Conversely, neurodegenerative diseases are characterized by protein accumulation, leading to reduced proteasome activity. As a result, the proteasome has emerged as a target for therapeutic intervention. The potential of the proteasome as a therapeutic target has come from studies involving chemical stimulators and inhibitors, and the development of a suite of assays and probes that can be used to monitor proteasome activity with purified enzyme and in live cells.
Collapse
Affiliation(s)
- Marianne E Maresh
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, IN 47907, USA
| | - Andres F Salazar-Chaparro
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, IN 47907, USA
| | - Darci J Trader
- Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, 575 West Stadium Avenue, West Lafayette, IN 47907, USA
| |
Collapse
|
182
|
Machado V, Carvalho R, Mendes JJ, Botelho J. The Ubiquitin Proteasome System in Periodontal Disease: A Comprehensive Review. FRONTIERS IN DENTAL MEDICINE 2020. [DOI: 10.3389/fdmed.2020.613080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The turnover of intracellular proteins is a highly selective and regulated process. This process is responsible for avoiding injury and irreparable breakdown of cellular constituents. Its impairment disrupts cellular stability, integrity, and homeostasis. The ubiquitin-proteasome system (UPS) is responsible for this programmed degradation of most intracellular proteins. This process involves a cascade of enzymes that involves the ubiquitin conjugation to a target substrate protein, its recognition and degradation by the proteasome. The turn-over of intracellular proteins is a non-stop ubiquitous process that regulates a series of mechanisms, for instance transcription, translation, endocytosis. In addition, proteasome act by releasing peptides that may serve to other purposes, such as antigen presentation in immune actions and enzymatic flagging toward biosynthesis and gluconeogenesis. The role of the UPS impairment in periodontal diseases is gaining growing. This acquaintance might contribute to the development of novel therapeutic applications. Thus, this review focuses on the latest progresses on the role of the UPS and its signaling pathways in Periodontal Medicine. Furthermore, we discuss the potential of UPS-based drugs development to be used in periodontal disease therapy.
Collapse
|
183
|
Misiewicz-Krzeminska I, de Ramón C, Corchete LA, Krzeminski P, Rojas EA, Isidro I, García-Sanz R, Martínez-López J, Oriol A, Bladé J, Lahuerta JJ, San Miguel J, Rosiñol L, Mateos MV, Gutiérrez NC. Quantitative expression of Ikaros, IRF4, and PSMD10 proteins predicts survival in VRD-treated patients with multiple myeloma. Blood Adv 2020; 4:6023-6033. [PMID: 33284947 PMCID: PMC7724915 DOI: 10.1182/bloodadvances.2020002711] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
The search for biomarkers based on the mechanism of drug action has not been thoroughly addressed in the therapeutic approaches to multiple myeloma (MM), mainly because of the difficulty in analyzing proteins obtained from purified plasma cells. Here, we investigated the prognostic impact of the expression of 12 proteins involved in the mechanism of action of bortezomib, lenalidomide, and dexamethasone (VRD), quantified by capillary nanoimmunoassay, in CD138-purified samples from 174 patients with newly diagnosed MM treated according to the PETHEMA/GEM2012 study. A high level of expression of 3 out of 5 proteasome components tested (PSMD1, PSMD4, and PSMD10) negatively influenced survival. The 5 analyzed proteins involved in lenalidomide's mode of action were associated with time to progression (TTP); low levels of cereblon and IRF4 protein and high levels of Ikaros, AGO2, and Aiolos were significantly associated with shorter TTP. Although the glucocorticoid receptor (GCR) level by itself had no significant impact on MM prognosis, a high XPO1 (exportin 1)/GCR ratio was associated with shorter TTP and progression-free survival (PFS). The multivariate Cox model identified high levels of PSMD10 (hazard ratio [HR] TTP, 3.49; P = .036; HR PFS, 5.33; P = .004) and Ikaros (HR TTP, 3.01, P = .014; HR PFS, 2.57; P = .028), and low levels of IRF4 protein expression (HR TTP, 0.33; P = .004; HR PFS, 0.35; P = .004) along with high-risk cytogenetics (HR TTP, 3.13; P < .001; HR PFS, 2.69; P = .002), as independently associated with shorter TTP and PFS. These results highlight the value of assessing proteins related to the mechanism of action of drugs used in MM for predicting treatment outcome.
Collapse
Affiliation(s)
- Irena Misiewicz-Krzeminska
- Hematology Department, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca, Salamanca, Spain
- Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, University of Salamanca-National Research Council (USAL-CSIC), Salamanca, Spain
- Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Cristina de Ramón
- Hematology Department, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca, Salamanca, Spain
| | - Luis A Corchete
- Hematology Department, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca, Salamanca, Spain
- Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, University of Salamanca-National Research Council (USAL-CSIC), Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Patryk Krzeminski
- Hematology Department, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca, Salamanca, Spain
- Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, University of Salamanca-National Research Council (USAL-CSIC), Salamanca, Spain
- Department of Nanobiotechnology and Experimental Ecology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Elizabeta A Rojas
- Hematology Department, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca, Salamanca, Spain
- Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, University of Salamanca-National Research Council (USAL-CSIC), Salamanca, Spain
| | - Isabel Isidro
- Hematology Department, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca, Salamanca, Spain
- Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, University of Salamanca-National Research Council (USAL-CSIC), Salamanca, Spain
| | - Ramón García-Sanz
- Hematology Department, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca, Salamanca, Spain
- Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, University of Salamanca-National Research Council (USAL-CSIC), Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| | - Joaquín Martínez-López
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Medicine Department, Complutense University, Madrid, Spain
- Hematology Department, Hospital 12 de Octubre, Madrid, Spain
- Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Albert Oriol
- Hospital Germans Trias i Pujol, Barcelona, Spain
| | | | | | - Jesús San Miguel
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Clínica Universidad de Navarra, Centro de Investigaciones Médicas Aplicadas, Pamplona, Spain
| | | | - María-Victoria Mateos
- Hematology Department, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca, Salamanca, Spain
- Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, University of Salamanca-National Research Council (USAL-CSIC), Salamanca, Spain
| | - Norma C Gutiérrez
- Hematology Department, University Hospital of Salamanca, Institute of Biomedical Research of Salamanca, Salamanca, Spain
- Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, University of Salamanca-National Research Council (USAL-CSIC), Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
| |
Collapse
|
184
|
Meep, a Novel Regulator of Insulin Signaling, Supports Development and Insulin Sensitivity via Maintenance of Protein Homeostasis in Drosophila melanogaster. G3-GENES GENOMES GENETICS 2020; 10:4399-4410. [PMID: 32998936 PMCID: PMC7718763 DOI: 10.1534/g3.120.401688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Insulin signaling is critical for developmental growth and adult homeostasis, yet the downstream regulators of this signaling pathway are not completely understood. Using the model organism Drosophila melanogaster, we took a genomic approach to identify novel mediators of insulin signaling. These studies led to the identification of Meep, encoded by the gene CG32335. Expression of this gene is both insulin receptor- and diet-dependent. We found that Meep was specifically required in the developing fat body to tolerate a high-sugar diet (HSD). Meep is not essential on a control diet, but when reared on an HSD, knockdown of meep causes hyperglycemia, reduced growth, developmental delay, pupal lethality, and reduced longevity. These phenotypes stem in part from Meep’s role in promoting insulin sensitivity and protein stability. This work suggests a critical role for protein homeostasis in development during overnutrition. Because Meep is conserved and obesity-associated in mammals, future studies on Meep may help to understand the role of proteostasis in insulin-resistant type 2 diabetes.
Collapse
|
185
|
Shen J, Wang R, Wang Q, Zhang M, Liu C, Tao Z, Su G. The improved anticancer effects of Bortezomib-loaded hollow mesoporous silica nanospheres on lymphoma development. Aging (Albany NY) 2020; 13:411-423. [PMID: 33290262 PMCID: PMC7835069 DOI: 10.18632/aging.202146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/01/2020] [Indexed: 11/25/2022]
Abstract
As the first clinical proteasome inhibitor, Bortezomib (BTZ) has been reported to improve the outcome of lymphoma. However, due to the unstable property, low bioavailability, and hydrophobic properties of BTZ, it is needed to develop effective drug delivery systems to deliver BTZ into targeted cells or organs. Here we developed a bortezomib (BTZ)-loaded HMSNs (BTZ@HMSNs) system, which can sustain the release of BTZ in targeted tissues. In vitro assays showed that BTZ@HMSNs limited cell proliferation and augmented apoptosis of lymphoma SNK-1 cells. Moreover, BTZ@HMSNs significantly diminished migration and invasion of SNK-1 cells as compared with BTZ. In contrast to the upregulation of SHP-1, BTZ@HMSNs decreased the mRNA levels of c-Kit, NF-κB, and JAK1, which elicit oncogenic role in lymphoma development. Importantly, lymphoma mice model showed that BTZ@HMSNs significantly activated p53 signaling and reduced tumor volume and weight compared with free BTZ. Our data thus demonstrate that BTZ@HMSNs manifests improved tumor-suppressing effect in vitro and in vivo compared to free BTZ. We believe that HMSNs is a promising strategy for delivering therapeutic agents for cancer treatment.
Collapse
Affiliation(s)
- Jie Shen
- Department of Hematology, Centre Hospital of Cangzhou, Cangzhou, Hebei Province, China
| | - Ruihuan Wang
- Department of Hematology, Centre Hospital of Cangzhou, Cangzhou, Hebei Province, China
| | - Qing Wang
- Department of Hematology, Centre Hospital of Cangzhou, Cangzhou, Hebei Province, China
| | - Minjuan Zhang
- Department of Hematology, Centre Hospital of Cangzhou, Cangzhou, Hebei Province, China
| | - Chunyan Liu
- Department of Hematology, Centre Hospital of Cangzhou, Cangzhou, Hebei Province, China
| | - Zhenxia Tao
- Department of Central Laboratory, Centre Hospital of Cangzhou, Cangzhou, Hebei Province, China
| | - Guohong Su
- Department of Hematology, Centre Hospital of Cangzhou, Cangzhou, Hebei Province, China
| |
Collapse
|
186
|
Shao F, Lyu X, Miao K, Xie L, Wang H, Xiao H, Li J, Chen Q, Ding R, Chen P, Xing F, Zhang X, Luo G, Zhu W, Cheng G, Lon NW, Martin SE, Wang G, Chen G, Dai Y, Deng C. Enhanced Protein Damage Clearance Induces Broad Drug Resistance in Multitype of Cancers Revealed by an Evolution Drug-Resistant Model and Genome-Wide siRNA Screening. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001914. [PMID: 33304752 PMCID: PMC7709997 DOI: 10.1002/advs.202001914] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/09/2020] [Indexed: 05/08/2023]
Abstract
Resistance to therapeutic drugs occurs in virtually all types of cancers, and the tolerance to one drug frequently becomes broad therapy resistance; however, the underlying mechanism remains elusive. Combining a whole whole-genome-wide RNA interference screening and an evolutionary drug pressure model with MDA-MB-231 cells, it is found that enhanced protein damage clearance and reduced mitochondrial respiratory activity are responsible for cisplatin resistance. Screening drug-resistant cancer cells and human patient-derived organoids for breast and colon cancers with many anticancer drugs indicates that activation of mitochondrion protein import surveillance system enhances proteasome activity and minimizes caspase activation, leading to broad drug resistance that can be overcome by co-treatment with a proteasome inhibitor, bortezomib. It is further demonstrated that cisplatin and bortezomib encapsulated into nanoparticle further enhance their therapeutic efficacy and alleviate side effects induced by drug combination treatment. These data demonstrate a feasibility for eliminating broad drug resistance by targeting its common mechanism to achieve effective therapy for multiple cancers.
Collapse
Affiliation(s)
- Fangyuan Shao
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Xueying Lyu
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Kai Miao
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Lisi Xie
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Haitao Wang
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Hao Xiao
- Guangdong Key Laboratory of Animal Breeding and NutritionInstitute of Animal ScienceGuangdong Academy of Agricultural SciencesGuangzhou510640China
| | - Jie Li
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Qiang Chen
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Renbo Ding
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Ping Chen
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Fuqiang Xing
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Department of BiologySouthern University of Science and TechnologyShenzhen518055China
| | - Xu Zhang
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | | | | | - Gregory Cheng
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Ng Wai Lon
- Centro Hospitalar Conde de S. JanuárioMacau820004China
| | - Scott E. Martin
- Division of Pre‐Clinical InnovationNational Center for Advancing Translational Sciences (NCATS)National Institutes of HealthBethesdaMD20892USA
| | - Guanyu Wang
- Department of BiologySouthern University of Science and TechnologyShenzhen518055China
| | - Guokai Chen
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Yunlu Dai
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Chu‐Xia Deng
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| |
Collapse
|
187
|
Zhou H, Lei M, Wang W, Guo M, Wang J, Zhang H, Qiao L, Feng H, Liu Z, Chen L, Hou J, Wang X, Gu C, Zhao B, Izumchenko E, Yang Y, Zhu Y. In vitro and in vivo efficacy of the novel oral proteasome inhibitor NNU546 in multiple myeloma. Aging (Albany NY) 2020; 12:22949-22974. [PMID: 33203800 PMCID: PMC7746380 DOI: 10.18632/aging.104023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/15/2020] [Indexed: 01/14/2023]
Abstract
Proteasome inhibition demonstrates highly effective impact on multiple myeloma (MM) treatment. Here, we aimed to examine anti-tumor efficiency and underlying mechanisms of a novel well tolerated orally applicable proteasome inhibitor NNU546 and its hydrolyzed pharmacologically active form NNU219. NNU219 showed more selective inhibition to proteasome catalytic subunits and less off-target effect than bortezomib ex vivo. Moreover, intravenous and oral administration of either NNU219 or NNU546 led to more sustained pharmacodynamic inhibitions of proteasome activities compared with bortezomib. Importantly, NNU219 exhibited potential anti-MM activity in both MM cell lines and primary samples in vitro. The anti-MM activity of NNU219 was associated with induction of G2/M-phase arrest and apoptosis via activation of the caspase cascade and endoplasmic reticulum stress response. Significant growth-inhibitory effects of NNU219 and NNU546 were observed in 3 different human MM xenograft mouse models. Furthermore, such observation was even found in the presence of a bone marrow microenvironment. Taken together, these findings provided the basis for clinical trial of NNU546 to determine its potential as a candidate for MM treatment.
Collapse
Affiliation(s)
- Hui Zhou
- College of Life Science, Nanjing Normal University, Nanjing 210046, PR China
| | - Meng Lei
- College of Science, Nanjing Forestry University, Nanjing 210037, PR China
| | - Wang Wang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Mengjie Guo
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Jia Wang
- Jiangsu Chia Tai Fenghai Pharmaceutical Co. Ltd., Nanjing 210046, PR China
| | - Haoyang Zhang
- College of Life Science, Nanjing Normal University, Nanjing 210046, PR China
| | - Li Qiao
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Huayun Feng
- College of Science, Nanjing Forestry University, Nanjing 210037, PR China
| | - Zhaogang Liu
- Jiangsu Chia Tai Fenghai Pharmaceutical Co. Ltd., Nanjing 210046, PR China
| | - Lijuan Chen
- The 1st Affiliated Hospital of Nanjing Medical University, Nanjing 210029, PR China
| | - Jianhao Hou
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Xueyuan Wang
- College of Life Science, Nanjing Normal University, Nanjing 210046, PR China
| | - Chenxi Gu
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, PR China
| | - Bo Zhao
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, PR China
| | - Evgeny Izumchenko
- Department of Medicine, Section of Hematology and Oncology, University of Chicago, Chicago, IL 60637, USA
| | - Ye Yang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.,The 3rd Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Yongqiang Zhu
- College of Life Science, Nanjing Normal University, Nanjing 210046, PR China.,Jiangsu Chia Tai Fenghai Pharmaceutical Co. Ltd., Nanjing 210046, PR China
| |
Collapse
|
188
|
Kim YM, Kim HJ. Proteasome Inhibitor MG132 is Toxic and Inhibits the Proliferation of Rat Neural Stem Cells but Increases BDNF Expression to Protect Neurons. Biomolecules 2020; 10:biom10111507. [PMID: 33147870 PMCID: PMC7692322 DOI: 10.3390/biom10111507] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/21/2020] [Accepted: 10/27/2020] [Indexed: 12/31/2022] Open
Abstract
Regulation of protein expression is essential for maintaining normal cell function. Proteasomes play important roles in protein degradation and dysregulation of proteasomes is implicated in neurodegenerative disorders. In this study, using a proteasome inhibitor MG132, we showed that proteasome inhibition reduces neural stem cell (NSC) proliferation and is toxic to NSCs. Interestingly, MG132 treatment increased the percentage of neurons in both proliferation and differentiation culture conditions of NSCs. Proteasome inhibition reduced B-cell lymphoma 2 (Bcl-2)/Bcl-2 associated X protein ratio. In addition, MG132 treatment induced cAMP response element-binding protein phosphorylation and increased the expression of brain-derived neurotrophic factor transcripts and proteins. These data suggest that proteasome function is important for NSC survival and differentiation. Moreover, although MG132 is toxic to NSCs, it may increase neurogenesis. Therefore, by modifying MG132 chemical structure and developing none toxic proteasome inhibitors, neurogenic chemicals can be developed to control NSC cell fate.
Collapse
Affiliation(s)
| | - Hyun-Jung Kim
- Correspondence: ; Tel.: +82-2-820-5619; Fax: +82-2-816-7338
| |
Collapse
|
189
|
Kumar V, Khan I, Gupta U. Lipid-dendrimer nanohybrid system or dendrosomes: evidences of enhanced encapsulation, solubilization, cellular uptake and cytotoxicity of bortezomib. APPLIED NANOSCIENCE 2020. [DOI: 10.1007/s13204-020-01515-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
190
|
Dopamine-modified poly(ε-caprolactone) micelles for pH controlled delivery of bortezomib. Int J Pharm 2020; 590:119885. [DOI: 10.1016/j.ijpharm.2020.119885] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/20/2020] [Accepted: 09/12/2020] [Indexed: 12/12/2022]
|
191
|
Ghannam K, Martinez Gamboa L, Kedor C, Spengler L, Kuckelkorn U, Häupl T, Burmester G, Feist E. Response to abatacept is associated with the inhibition of proteasome β1i expression in T cells of patients with rheumatoid arthritis. RMD Open 2020; 6:rmdopen-2020-001248. [PMID: 32998980 PMCID: PMC7547540 DOI: 10.1136/rmdopen-2020-001248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/27/2020] [Accepted: 08/21/2020] [Indexed: 11/25/2022] Open
Abstract
Objective Abatacept is a biological disease-modifying antirheumatic drug (DMARD) used for the treatment of rheumatoid arthritis (RA) and modulates the costimulatory signal by cluster of differentiation (CD)28:CD80/CD86 interaction required for T cell activation. Since CD28-mediated signalling regulates many T cell functions including cytokine production of, for example, interferons (IFNs), it is of interest to clarify, whether response to abatacept has an effect on the IFN inducible immunoproteasome, as a central regulator of the immune response. Methods Effects of abatacept on the proteasome were investigated in 39 patients with RA over a period of 24 weeks. Using real-time PCR, transcript levels of constitutive and corresponding immunoproteasome catalytic subunits were investigated at baseline (T0), week 16 (T16) and week 24 (T24) in sorted blood cells. Proteasomal activity and induction of apoptosis after proteasome inhibition were also evaluated. Results Abatacept achieved remission or low disease activity in 55% of patients at T16 and in 70% of patients at T24. By two-way analysis of variance (ANOVA), a significant reduction of proteasome immunosubunit β1i was shown only in CD4+ and CD8+ T cells of sustained responders at both T16 and T24. One-way ANOVA analysis for each response group confirmed the results and showed a significant reduction at T24 in CD4+ and CD8+ T cells of the same group. Abatacept did not influence chymotrypsin-like activity of proteasome and had no effect on induction of apoptosis under exposure to a proteasome inhibitor in vitro. Conclusion The reduction of proteasome immunosubunit β1i in T cells of patients with RA with sustained response to abatacept suggests association of the immunoproteasome of T cells with RA disease activity.
Collapse
Affiliation(s)
- Khetam Ghannam
- Department of Rheumatology and Clinical Immunology, Charite University Hospital Berlin, Berlin, Germany
| | - Lorena Martinez Gamboa
- Department of Rheumatology and Clinical Immunology, Charite University Hospital Berlin, Berlin, Germany
| | - Claudia Kedor
- Department of Rheumatology and Clinical Immunology, Charite University Hospital Berlin, Berlin, Germany
| | - Lydia Spengler
- Department of Rheumatology and Clinical Immunology, Charite University Hospital Berlin, Berlin, Germany
| | - Ulrike Kuckelkorn
- Institute of Biochemistry, Charite University Hospital Berlin, Berlin, Germany
| | - Thomas Häupl
- Department of Rheumatology and Clinical Immunology, Charite University Hospital Berlin, Berlin, Germany
| | - Gerd Burmester
- Department of Rheumatology and Clinical Immunology, Charite University Hospital Berlin, Berlin, Germany
| | - Eugen Feist
- Helios Fachklinik Vogelsang-Gommern GmbH, Vogelsang-Gommern, Germany.,Department of Rheumatology and Clinical Immunology, Charite University Hospital Berlin, Berlin, Germany
| |
Collapse
|
192
|
Ribeiro GH, Guedes APM, de Oliveira TD, de Correia CRSTB, Colina-Vegas L, Lima MA, Nóbrega JA, Cominetti MR, Rocha FV, Ferreira AG, Castellano EE, Teixeira FR, Batista AA. Ruthenium(II) Phosphine/Mercapto Complexes: Their in Vitro Cytotoxicity Evaluation and Actions as Inhibitors of Topoisomerase and Proteasome Acting as Possible Triggers of Cell Death Induction. Inorg Chem 2020; 59:15004-15018. [PMID: 32997499 DOI: 10.1021/acs.inorgchem.0c01835] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this paper, a series of new ruthenium complexes of the general formula [Ru(NS)(dpphpy)(dppb)]PF6 (Ru1-Ru3), where dpphpy = diphenyl-2-pyridylphosphine, NS ligands = 2-thiazoline-2-thiol (tzdt, Ru1), 2-mercaptopyrimidine (pySm, Ru2), and 4,6-diamino-2-mercaptopyrimidine (damp, Ru3), and dppb = 1,4-bis(diphenylphosphino)butane, were synthesized and characterized by elemental analysis, spectroscopic techniques (IR, UV/visible, and 1D and 2D NMR), and X-ray diffraction. In the characterization, the correlation between the phosphorus atoms and their respective aromatic hydrogen atoms of the compounds in the assignment stands outs, by 1H-31P HMBC experiments. The compounds show anticancer activities against A549 (lung) and MDA-MB-231 (breast) cancer cell lines, higher than the clinical drug cisplatin. All of the complexes are more cytotoxic against the cancer cell lines than against the MRC-5 (lung) and MCF-10A (breast) nontumorigenic human cell lines. For A549 tumor cells, cell cycle analysis upon treatment with Ru2 showed that it inhibits the mitotic phase because arrest was observed in the Sub-G1 phase. Additionally, the compound induces cell death by an apoptotic pathway in a dose-dependent manner, according to annexin V-PE assay. The multitargeted character of the compounds was investigated, and the biomolecules were DNA, topoisomerase IB, and proteasome, as well as the fundamental biomolecule in the pharmacokinetics of drugs, human serum albumin. The experimental results indicate that the complexes do not target DNA in the cells. At low concentrations, the compounds showed the ability to partially inhibit the catalytic activity of topoisomerase IB in the process of relaxation of the DNA plasmid. Among the complexes assayed in cultured cells, complex Ru3 was able to diminish the proteasomal chymotrypsin-like activity to a greater extent.
Collapse
Affiliation(s)
- Gabriel H Ribeiro
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Adriana P M Guedes
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Tamires D de Oliveira
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Camila R S T B de Correia
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Legna Colina-Vegas
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil.,Instituto de Química, Universidade Federal do Rio Grande do Sul, CP 15003, 91501-970 Porto Alegre, Rio Grande do Sul, Brazil
| | - Mauro A Lima
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Joaquim A Nóbrega
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Márcia R Cominetti
- Departamento de Gerontologia, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo Brazil
| | - Fillipe V Rocha
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Antônio G Ferreira
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Eduardo E Castellano
- Instituto de Física de São Carlos, Universidade de São Paulo, CEP 13560-970 São Carlos, São Paulo, Brazil
| | - Felipe R Teixeira
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos, CEP 13565-905 São Carlos, São Paulo, Brazil
| |
Collapse
|
193
|
Montefusco V, Mussetti A, Salas MQ, Martinelli G, Cerchione C. Old and new generation proteasome inhibitors in multiple myeloma. Panminerva Med 2020; 62:193-206. [PMID: 32957744 DOI: 10.23736/s0031-0808.20.04148-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Proteasome inhibitors (PIs) represent a recently developed drug class that inhibit the ubiquitin-proteasome system, thus interfering with the intracellular machinery who has the duty of misfolded proteins disposal. Myeloma plasma cells are structurally aimed at the production of large quantities of immunoglobulins. This explains their vulnerability to any perturbation of intracellular protein homeostasis. Bortezomib is the first-in-class PI and nowadays, in combination with other compounds, is the cornerstone of multiple myeloma (MM) treatment in several settings. Bortezomib has several attractive features for its inclusion in the induction phase of therapy: high efficacy, rapid cytoreduction, absence of nephrotoxicity, fast reduction of plasmacytomas, and fast pain control. However, the safety profile of bortezomib is characterized by a not negligible peripheral neuropathy. Newer PIs, such as carfilzomib and ixazomib, have been developed and each offers specific advantages. Carfilzomib is extremely efficient in proteasome inhibition. This results in high efficacy but suffers from a significant cardiotoxicity. Ixazomib is the first oral PI with a proteasome inhibition profile similar to bortezomib, with lower neurotoxicity. PIs mechanism of action is complementary with other drug classes, and this explains the synergism between PIs and other drugs, in particular steroids and immunomodulators. PIs are frequently used in doublets and triplets. Also, they can be associated with anti-CD38 monoclonal antibodies. This review summarizes the principal biological and clinical features of PIs in the MM treatment.
Collapse
Affiliation(s)
| | - Alberto Mussetti
- Department Clinical Hematology, Institut Català d'Oncologia-Hospitalet, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Maria Q Salas
- Department Clinical Hematology, Institut Català d'Oncologia-Hospitalet, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Giovanni Martinelli
- Unit of Hematology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| | - Claudio Cerchione
- Unit of Hematology, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Forlì-Cesena, Italy
| |
Collapse
|
194
|
Kiuchi T, Tomaru U, Ishizu A, Imagawa M, Iwasaki S, Suzuki A, Otsuka N, Ohhara Y, Kinoshita I, Matsuno Y, Dosaka-Akita H, Kasahara M. Expression of the immunoproteasome subunit β5i in non-small cell lung carcinomas. J Clin Pathol 2020; 74:300-306. [PMID: 32943490 DOI: 10.1136/jclinpath-2020-206618] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/24/2020] [Accepted: 06/27/2020] [Indexed: 12/25/2022]
Abstract
AIM The immunoproteasome is a specific proteasome isoform whose proteolytic activity enhances the generation of antigenic peptides to be presented by major histocompatibility complex class I molecules to CD8+ T cells. Physiologically, it is expressed abundantly in immune cells and is induced in somatic cells by cytokines, especially interferon-γ. Recently, variable expression of immunoproteasomes has been demonstrated in different types of cancers. However, the clinical significance of immunoproteasome expression in malignant tumours is poorly understood. In this study, we performed clinicopathological evaluation of immunoproteasome subunit β5i in non-small cell lung carcinomas (NSCLCs). METHODS Tumour tissues were collected from 155 patients with NSCLCs, and immunohistochemical analysis for β5i was performed in relation to the prognosis of patients. RESULTS High expression of β5i was found in about 20% of all NSCLCs and was found significantly more frequently (40%) in the adenocarcinoma subset. High expression of β5i was associated with a better 5-year relative survival rate in patients with pStage I to II adenocarcinoma and was also a significant and independent favourable prognostic factor in adenocarcinoma patients. In addition, when we performed in vitro analysis using NSCLC cell lines, combined treatment with the immunoproteasome-specific inhibitor ONX0914 and the proteasome inhibitor MG132 enhanced cell death in β5i-expressing NSCLC cell lines. CONCLUSION The expression of immunoproteasome can be explored as both a prognostic factor and a potential therapeutic target in NSCLCs. Since immunoproteasomes have crucial role in the antigen presentation, further studies may help to provide essential knowledge for therapeutic strategies in anticancer immunotherapy.
Collapse
Affiliation(s)
- Takayuki Kiuchi
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Utano Tomaru
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiro Ishizu
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Makoto Imagawa
- Department of Diagnostic Pathology, KKR Sapporo Medical Center, Sapporo, Hokkaido, Japan
| | - Sari Iwasaki
- Department of Diagnostic Pathology, KKR Sapporo Medical Center, Sapporo, Hokkaido, Japan
| | - Akira Suzuki
- Department of Diagnostic Pathology, KKR Sapporo Medical Center, Sapporo, Hokkaido, Japan
| | - Noriyuki Otsuka
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshihito Ohhara
- Department of Medical Oncology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ichiro Kinoshita
- Department of Medical Oncology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Hirotoshi Dosaka-Akita
- Department of Medical Oncology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masanori Kasahara
- Department of Pathology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
195
|
Zhang Z, Gu J, Yin M, Wang D, Ma C, Du J, Lin Z, Hu S, Wang X, Li Y, Tan G, Luo H, Wei G. Establishment and Investigation of a Multiple Gene Expression Signature to Predict Long-Term Survival in Pancreatic Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1570862. [PMID: 33015155 PMCID: PMC7516738 DOI: 10.1155/2020/1570862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 07/01/2020] [Indexed: 11/23/2022]
Abstract
Pancreatic cancer remains a lethal type of cancer with poor prognosis. Molecular classification enables in-depth, precise prognostic assessment. This study is aimed at identifying a robust and simple mRNA signature to predict the overall survival (OS) of pancreatic cancer (PC) patients. Differentially expressed genes (DEGs) between 45 paired pancreatic tumor samples and adjacent healthy tissues were selected. For risk determination, a LASSO Cox regression model with DEGs was used to generate the OS-associated risk score formula for the training cohort containing 177 PC patients. Another five independent datasets were used as the testing cohort to determine the predictive efficiency for further validation. In total, 441 DEGs were selected after considering the enrichment of classical pathways, such as EMT, cell cycle, cell adhesion, and PI3K-AKT. A five-gene signature for risk discrimination was established with high efficacy using LASSO Cox regression in the training group. External validation showed that patients identified by the gene expression signature to be in the high-risk group had poorer prognosis compared with the low-risk patients. Further investigation identified the differential epigenetic modification patterns of the five genes, which indicated their roles in tumor progression and their effect on therapy. In conclusion, we constructed a robust five-gene expression signature that could predict the OS of PC patients, offering a new insight for risk discrimination in daily clinical practice.
Collapse
Affiliation(s)
- Zhiqiang Zhang
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jiangning Gu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Menghong Yin
- Department of Sports Medicine, Dalian Municipal Central Hospital, Dalian Medical University, Dalian, Liaoning Province, China
| | - Di Wang
- Department of Scientific Research, Eyes & ENT Hospital of Fudan University, Shanghai, China
| | - Chi Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Jian Du
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Zhikun Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Siling Hu
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xuelong Wang
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ying Li
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guang Tan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Haifeng Luo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Gang Wei
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
196
|
Complete Tumor Regression by Liposomal Bortezomib in a Humanized Mouse Model of Multiple Myeloma. Hemasphere 2020; 4:e463. [PMID: 32923984 PMCID: PMC7455224 DOI: 10.1097/hs9.0000000000000463] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/29/2020] [Indexed: 11/25/2022] Open
Abstract
Supplemental Digital Content is available in the text
Collapse
|
197
|
Maiti P, Manna J, Burch ZN, Flaherty DB, Larkin JD, Dunbar GL. Ameliorative Properties of Boronic Compounds in In Vitro and In Vivo Models of Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21186664. [PMID: 32933008 PMCID: PMC7555802 DOI: 10.3390/ijms21186664] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by amyloid (Aβ) aggregation, hyperphosphorylated tau, neuroinflammation, and severe memory deficits. Reports that certain boronic compounds can reduce amyloid accumulation and neuroinflammation prompted us to compare trans-2-phenyl-vinyl-boronic-acid-MIDA-ester (TPVA) and trans-beta-styryl-boronic-acid (TBSA) as treatments of deficits in in vitro and in vivo models of AD. We hypothesized that these compounds would reduce neuropathological deficits in cell-culture and animal models of AD. Using a dot-blot assay and cultured N2a cells, we observed that TBSA inhibited Aβ42 aggregation and increased cell survival more effectively than did TPVA. These TBSA-induced benefits were extended to C. elegans expressing Aβ42 and to the 5xFAD mouse model of AD. Oral administration of 0.5 mg/kg dose of TBSA or an equivalent amount of methylcellulose vehicle to groups of six- and 12-month-old 5xFAD or wild-type mice over a two-month period prevented recognition- and spatial-memory deficits in the novel-object recognition and Morris-water-maze memory tasks, respectively, and reduced the number of pyknotic and degenerated cells, Aβ plaques, and GFAP and Iba-1 immunoreactivity in the hippocampus and cortex of these mice. These findings indicate that TBSA exerts neuroprotective properties by decreasing amyloid plaque burden and neuroinflammation, thereby preventing neuronal death and preserving memory function in the 5xFAD mice.
Collapse
Affiliation(s)
- Panchanan Maiti
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48859, USA
- Department of Psychology, Central Michigan University, Mt. Pleasant, MI 48859, USA
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI 48859, USA
- Field Neurosciences Institute, Ascension St. Mary, Saginaw, MI 48604, USA;
- College of Health and Human Services, Saginaw Valley State University, Saginaw, MI 48604, USA
- Correspondence: (P.M.); (G.L.D.); Tel.: +1-989-497-3026 (P.M.); +1-989-497-3105 (G.L.D.)
| | - Jayeeta Manna
- Field Neurosciences Institute, Ascension St. Mary, Saginaw, MI 48604, USA;
| | - Zoe N. Burch
- Department of Biology, Eckerd College, St. Petersburg, FL 33711, USA; (Z.N.B.); (D.B.F.)
| | - Denise B. Flaherty
- Department of Biology, Eckerd College, St. Petersburg, FL 33711, USA; (Z.N.B.); (D.B.F.)
| | - Joseph D. Larkin
- Department of Chemistry, Eckerd College, St. Petersburg, FL 33711, USA;
| | - Gary L. Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48859, USA
- Department of Psychology, Central Michigan University, Mt. Pleasant, MI 48859, USA
- Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI 48859, USA
- Field Neurosciences Institute, Ascension St. Mary, Saginaw, MI 48604, USA;
- Correspondence: (P.M.); (G.L.D.); Tel.: +1-989-497-3026 (P.M.); +1-989-497-3105 (G.L.D.)
| |
Collapse
|
198
|
Tundo GR, Sbardella D, Santoro AM, Coletta A, Oddone F, Grasso G, Milardi D, Lacal PM, Marini S, Purrello R, Graziani G, Coletta M. The proteasome as a druggable target with multiple therapeutic potentialities: Cutting and non-cutting edges. Pharmacol Ther 2020; 213:107579. [PMID: 32442437 PMCID: PMC7236745 DOI: 10.1016/j.pharmthera.2020.107579] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/10/2023]
Abstract
Ubiquitin Proteasome System (UPS) is an adaptable and finely tuned system that sustains proteostasis network under a large variety of physiopathological conditions. Its dysregulation is often associated with the onset and progression of human diseases; hence, UPS modulation has emerged as a promising new avenue for the development of treatments of several relevant pathologies, such as cancer and neurodegeneration. The clinical interest in proteasome inhibition has considerably increased after the FDA approval in 2003 of bortezomib for relapsed/refractory multiple myeloma, which is now used in the front-line setting. Thereafter, two other proteasome inhibitors (carfilzomib and ixazomib), designed to overcome resistance to bortezomib, have been approved for treatment-experienced patients, and a variety of novel inhibitors are currently under preclinical and clinical investigation not only for haematological malignancies but also for solid tumours. However, since UPS collapse leads to toxic misfolded proteins accumulation, proteasome is attracting even more interest as a target for the care of neurodegenerative diseases, which are sustained by UPS impairment. Thus, conceptually, proteasome activation represents an innovative and largely unexplored target for drug development. According to a multidisciplinary approach, spanning from chemistry, biochemistry, molecular biology to pharmacology, this review will summarize the most recent available literature regarding different aspects of proteasome biology, focusing on structure, function and regulation of proteasome in physiological and pathological processes, mostly cancer and neurodegenerative diseases, connecting biochemical features and clinical studies of proteasome targeting drugs.
Collapse
Affiliation(s)
- G R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| | | | - A M Santoro
- CNR, Institute of Crystallography, Catania, Italy
| | - A Coletta
- Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - F Oddone
- IRCCS-Fondazione Bietti, Rome, Italy
| | - G Grasso
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - D Milardi
- CNR, Institute of Crystallography, Catania, Italy
| | - P M Lacal
- Laboratory of Molecular Oncology, IDI-IRCCS, Rome, Italy
| | - S Marini
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - R Purrello
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - G Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - M Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
199
|
Neurotoxicity of antineoplastic drugs: Mechanisms, susceptibility, and neuroprotective strategies. Adv Med Sci 2020; 65:265-285. [PMID: 32361484 DOI: 10.1016/j.advms.2020.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 12/22/2019] [Accepted: 04/13/2020] [Indexed: 02/06/2023]
Abstract
This review summarizes the adverse effects on the central and/or peripheral nervous systems that may occur in response to antineoplastic drugs. In particular, we describe the neurotoxic side effects of the most commonly used drugs, such as platinum compounds, doxorubicin, ifosfamide, 5-fluorouracil, vinca alkaloids, taxanes, methotrexate, bortezomib and thalidomide. Neurotoxicity may result from direct action of compounds on the nervous system or from metabolic alterations produced indirectly by these drugs, and either the central nervous system or the peripheral nervous system, or both, may be affected. The incidence and severity of neurotoxicity are principally related to the dose, to the duration of treatment, and to the dose intensity, though other factors, such as age, concurrent pathologies, and genetic predisposition may enhance the occurrence of side effects. To avoid or reduce the onset and severity of these neurotoxic effects, the use of neuroprotective compounds and/or strategies may be helpful, thereby enhancing the therapeutic effectiveness of antineoplastic drug.
Collapse
|
200
|
Alpha-lipoic acid alters the antitumor effect of bortezomib in melanoma cells in vitro. Sci Rep 2020; 10:14287. [PMID: 32868799 PMCID: PMC7459291 DOI: 10.1038/s41598-020-71138-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 08/04/2020] [Indexed: 12/27/2022] Open
Abstract
Bortezomib (BOZ) is a proteasome inhibitor chemotherapeutic agent utilized to treat multiple myeloma and recently offered to cure melanoma. Bortezomib-induced neuropathy is one of the dose-limiting side-effects, which can be treated with antioxidants (e.g. alpha-lipoic acid-ALA and Vitamin B1-vit B1). We hypothesized that these antioxidants may counteract the antitumor activity by disrupting the BOZ-induced pathways (e.g. proteasome inhibition or reactive oxygen species generation). The objectives were: (i) to verify the anti-proliferative effect of BOZ; (ii) to compare the influence of the antioxidants on the antitumor effect of BOZ in melanoma (A2058) and myeloma (U266) cells. At first, the reduction in the anti-proliferative effect of BOZ by ALA was proved in melanoma cells. Analysis of p53 phosphorylation and the cell cycle progression revealed that ALA failed to counteract these effects of BOZ. Nevertheless, a good correlation was found between the inhibition of the anti-proliferative effect, the anti-proteasome activity and the oxidative stress level after the co-treatment with 20 ng/mL BOZ + 100 μg/mL ALA. Downregulation of apoptotic proteins such as HO-1 and Claspin along with the inhibition of the cleavage of Caspase-3 indicated the proteomic background of the altered responsiveness of the melanoma cells exposed to BOZ + ALA. This phenomenon draws attention to the proper application of cancer supportive care to avoid possible interactions.
Collapse
|