Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nature ; 626(8000): 874-880, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38297121

ABSTRACT

Stress response pathways detect and alleviate adverse conditions to safeguard cell and tissue homeostasis, yet their prolonged activation induces apoptosis and disrupts organismal health1-3. How stress responses are turned off at the right time and place remains poorly understood. Here we report a ubiquitin-dependent mechanism that silences the cellular response to mitochondrial protein import stress. Crucial to this process is the silencing factor of the integrated stress response (SIFI), a large E3 ligase complex mutated in ataxia and in early-onset dementia that degrades both unimported mitochondrial precursors and stress response components. By recognizing bifunctional substrate motifs that equally encode protein localization and stability, the SIFI complex turns off a general stress response after a specific stress event has been resolved. Pharmacological stress response silencing sustains cell survival even if stress resolution failed, which underscores the importance of signal termination and provides a roadmap for treating neurodegenerative diseases caused by mitochondrial import defects.


Subject(s)
Mitochondria , Mitochondrial Proteins , Mutation , Neurodegenerative Diseases , Stress, Physiological , Ubiquitin-Protein Ligases , Apoptosis/drug effects , Ataxia/genetics , Cell Survival/drug effects , Dementia/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Stability/drug effects , Protein Transport/drug effects , Proteolysis/drug effects , Stress, Physiological/drug effects , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
2.
Artif Life ; 29(4): 468-486, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37610755

ABSTRACT

In sociotechnical settings, human operators are increasingly assisted by decision support systems. By employing such systems, important properties of sociotechnical systems, such as self-adaptation and self-optimization, are expected to improve further. To be accepted by and engage efficiently with operators, decision support systems need to be able to provide explanations regarding the reasoning behind specific decisions. In this article, we propose the use of learning classifier systems (LCSs), a family of rule-based machine learning methods, to facilitate and highlight techniques to improve transparent decision-making. Furthermore, we present a novel approach to assessing application-specific explainability needs for the design of LCS models. For this, we propose an application-independent template of seven questions. We demonstrate the approach's use in an interview-based case study for a manufacturing scenario. We find that the answers received do yield useful insights for a well-designed LCS model and requirements for stakeholders to engage actively with an intelligent agent.


Subject(s)
Intelligence , Machine Learning , Humans
3.
Blood Adv ; 7(4): 469-481, 2023 02 28.
Article in English | MEDLINE | ID: mdl-35917568

ABSTRACT

Proteasome inhibition is a highly effective treatment for multiple myeloma (MM). However, virtually all patients develop proteasome inhibitor resistance, which is associated with a poor prognosis. Hyperactive small ubiquitin-like modifier (SUMO) signaling is involved in both cancer pathogenesis and cancer progression. A state of increased SUMOylation has been associated with aggressive cancer biology. We found that relapsed/refractory MM is characterized by a SUMO-high state, and high expression of the SUMO E1-activating enzyme (SAE1/UBA2) is associated with poor overall survival. Consistently, continuous treatment of MM cell lines with carfilzomib (CFZ) enhanced SUMO pathway activity. Treatment of MM cell lines with the SUMO E1-activating enzyme inhibitor subasumstat (TAK-981) showed synergy with CFZ in both CFZ-sensitive and CFZ-resistant MM cell lines, irrespective of the TP53 state. Combination therapy was effective in primary MM cells and in 2 murine MM xenograft models. Mechanistically, combination treatment with subasumstat and CFZ enhanced genotoxic and proteotoxic stress, and induced apoptosis was associated with activity of the prolyl isomerase PIN1. In summary, our findings reveal activated SUMOylation as a therapeutic target in MM and point to combined SUMO/proteasome inhibition as a novel and potent strategy for the treatment of proteasome inhibitor-resistant MM.


Subject(s)
Multiple Myeloma , Proteasome Inhibitors , Humans , Animals , Mice , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Sumoylation , Proteasome Endopeptidase Complex/metabolism , Apoptosis , Ubiquitin-Activating Enzymes/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/pharmacology
4.
EMBO J ; 41(20): e110871, 2022 10 17.
Article in English | MEDLINE | ID: mdl-36059274

ABSTRACT

Deubiquitylases (DUBs) are therapeutically amenable components of the ubiquitin machinery that stabilize substrate proteins. Their inhibition can destabilize oncoproteins that may otherwise be undruggable. Here, we screened for DUB vulnerabilities in multiple myeloma, an incurable malignancy with dependency on the ubiquitin proteasome system and identified OTUD6B as an oncogene that drives the G1/S-transition. LIN28B, a suppressor of microRNA biogenesis, is specified as a bona fide cell cycle-specific substrate of OTUD6B. Stabilization of LIN28B drives MYC expression at G1/S, which in turn allows for rapid S-phase entry. Silencing OTUD6B or LIN28B inhibits multiple myeloma outgrowth in vivo and high OTUD6B expression evolves in patients that progress to symptomatic multiple myeloma and results in an adverse outcome of the disease. Thus, we link proteolytic ubiquitylation with post-transcriptional regulation and nominate OTUD6B as a potential mediator of the MGUS-multiple myeloma transition, a central regulator of MYC, and an actionable vulnerability in multiple myeloma and other tumors with an activated OTUD6B-LIN28B axis.


Subject(s)
Endopeptidases , MicroRNAs , Multiple Myeloma , Proto-Oncogene Proteins c-myc , RNA-Binding Proteins , Cell Cycle , Cell Line, Tumor , Endopeptidases/genetics , Humans , MicroRNAs/genetics , Multiple Myeloma/genetics , Proteasome Endopeptidase Complex/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA-Binding Proteins/genetics , Ubiquitins/metabolism
5.
EJNMMI Res ; 12(1): 4, 2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35099620

ABSTRACT

RATIONALE: Multiple myeloma (MM) cells synthesize large amounts of paraproteins, making radiolabeled amino acids promising candidates for PET imaging of MM patients. METHODS: We compare tumor uptake of the two amino acid analogs [18F]-fluoroethyltyrosine and [18F]-FACBC in a MM xenograft model and show the feasibility of PET imaging with [18F]-FACBC in a MM patient. RESULTS: Preclinically [18F]-FACBC showed superior performance, mainly due to the uptake via the ASC-system. In a subsequent proof-of-concept investigation [18F]-FACBC PET was performed in a MM patient. It allowed identification of both lesions with and without CT correlate (SUVmean 8.0 or 7.9) based on higher uptake compared to normal bone marrow (SUVmean 5.7). Bone signal was elevated compared to non-MM patients, and, thus [18F]-FACBC potentially allows the assessment of bone marrow infiltration. CONCLUSION: The FDA/EMA approved PET agent [18F]-FACBC is promising for imaging MM and should be further evaluated in prospective clinical studies.

6.
Blood Adv ; 6(2): 515-520, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34768284

ABSTRACT

Biomarkers that predict response to lenalidomide maintenance therapy in patients with multiple myeloma (MM) have remained elusive. We have shown that immunomodulatory drugs (IMiDs) exert anti-MM activity via destabilization of MCT1 and CD147. In this study, cell samples of 654 patients with MM who received lenalidomide (n = 455), thalidomide (n = 98), or bortezomib (n = 101) maintenance were assessed by gene expression profiling and RNA sequencing, followed by correlation of MCT1 and CD147 expression with data for progression-free survival (PFS) and overall survival (OS). Patients with high expression levels of MCT1 showed significantly reduced PFS (31.9 months vs 48.2 months in MCT1high vs MCT1low; P = .03) and OS (75.9 months vs not reached [NR] in MCT1high vs MCT1low; P = .001) in cases with lenalidomide maintenance, whereas MCT1 expression had no significant impact on PFS or OS in cases with bortezomib maintenance. We validated the predictive role of MCT1 for IMiD-based maintenance in an independent cohort of patients who received thalidomide (OS, 83.6 months vs NR in MCT1high vs MCT1low; P = .03). Functional validation showed that MCT1 overexpression in human MM cell lines significantly reduced the efficacy of lenalidomide, whereas no change was observed with bortezomib treatment, either in vitro or in a MM xenograft model. Our findings have established MCT1 expression as a predictive marker for response to lenalidomide-based maintenance in patients with MM.


Subject(s)
Multiple Myeloma , Biomarkers , Bortezomib/pharmacology , Bortezomib/therapeutic use , Humans , Lenalidomide/therapeutic use , Multiple Myeloma/therapy , Thalidomide/pharmacology , Thalidomide/therapeutic use
7.
Oncol Res Treat ; 44(12): 672-681, 2021.
Article in English | MEDLINE | ID: mdl-34749378

ABSTRACT

BACKGROUND: Multiple myeloma is the second most common hematologic malignancy, which to date remains incurable despite advances in treatment strategies including the use of novel substances such as proteasome inhibitors, immunomodulatory drugs, and monoclonal antibodies. SUMMARY: The bone marrow-based disease is preceded by the 2 sequential premalignant conditions: monoclonal gammo-pathy of undetermined significance and smoldering myeloma. Plasma cell leukemia and extramedullary disease occur, when malignant clones lose their dependency on the bone marrow. Key genetic features of these plasma cell dyscrasias include chromosomal aberrations such as translocations and hyperdiploidy, which occur during error-prone physiologic processes in B-cell development. Next-generation sequencing studies have identified mutations in major oncogenic pathways and tumor suppressors, which contribute to the pathogenesis of multiple myeloma and have revealed insights into the clonal evolution of the disease, particularly along different lines of therapy. More recently, the importance of epigenetic alterations and the role of the bone marrow microenvironment, including immune and osteogenic cells, have become evident. Key Messages: We herein review the current knowledge of the pathogenesis of multiple myeloma, which is crucial for the development of novel targeted therapeutic strategies. These can contribute to the endeavor to make multiple myeloma a curable disease.


Subject(s)
Multiple Myeloma , Antibodies, Monoclonal , Bone Marrow , Clonal Evolution/genetics , Humans , Immunomodulating Agents , Multiple Myeloma/genetics , Tumor Microenvironment
8.
Transfus Apher Sci ; 60(5): 103197, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34175195

ABSTRACT

High-dose chemotherapy followed by autologous stem cell transplantation is a major component in the treatment of patients with multiple myeloma. As a prerequisite, the successful collection of a sufficient number of viable peripheral blood hematopoietic CD34+ cells is critical. A common standard protocol for mobilization is currently not defined and critically discussed especially in German-speaking Europe. In times of the Covid-19 pandemic, safe and effective strategies have to be chosen to minimize hospitalization times and severe courses. In this single-center retrospective analysis, safety and efficacy of cyclophosphamide plus etoposide (CE) and growth-factor support (n = 33) was compared to cyclophosphamide mono treatment and growth-factor support (n = 49) in 82 patients with multiple myeloma at first diagnosis. CE was superior to cyclophosphamide mono with a significantly higher number of collected CD34+ cells (15.46 × 106 CD34+ cells/kg vs. 9.92 × 106 CD34+ cells/kg), significantly faster engraftment of granulocytes after stem cell transplantation (day 10.5 vs. day 11.6), shorter duration of the inpatient stay (17.47 days vs. 19.16 days) and significantly less transfusions (8.82 % vs. 30.61 % patients receiving transfusions). The safety profile was comparable in both groups and in line with published data. We conclude that CE is a safe and highly effective mobilization protocol in patients with multiple myeloma at first diagnosis and appears to be superior to the commonly used cyclophosphamide mono regimen.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cyclophosphamide/pharmacology , Etoposide/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/drug effects , Multiple Myeloma/therapy , Peripheral Blood Stem Cell Transplantation/methods , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , COVID-19 , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Etoposide/administration & dosage , Etoposide/adverse effects , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Male , Melphalan/administration & dosage , Middle Aged , Multiple Myeloma/blood , Myeloma Proteins/analysis , Pandemics , Retrospective Studies , SARS-CoV-2 , Transplantation, Autologous
9.
Mol Cell ; 81(6): 1170-1186.e10, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33571422

ABSTRACT

The complex architecture of transmembrane proteins requires quality control (QC) of folding, membrane positioning, and trafficking as prerequisites for cellular homeostasis and intercellular communication. However, it has remained unclear whether transmembrane protein-specific QC hubs exist. Here we identify cereblon (CRBN), the target of immunomodulatory drugs (IMiDs), as a co-chaperone that specifically determines chaperone activity of HSP90 toward transmembrane proteins by means of counteracting AHA1. This function is abrogated by IMiDs, which disrupt the interaction of CRBN with HSP90. Among the multiple transmembrane protein clients of CRBN-AHA1-HSP90 revealed by cell surface proteomics, we identify the amino acid transporter LAT1/CD98hc as a determinant of IMiD activity in multiple myeloma (MM) and present an Anticalin-based CD98hc radiopharmaceutical for MM radio-theranostics. These data establish the CRBN-AHA1-HSP90 axis in the biogenesis of transmembrane proteins, link IMiD activity to tumor metabolism, and nominate CD98hc and LAT1 as attractive diagnostic and therapeutic targets in MM.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Fusion Regulatory Protein 1, Heavy Chain/metabolism , HSP90 Heat-Shock Proteins/metabolism , Immunologic Factors/pharmacology , Large Neutral Amino Acid-Transporter 1/metabolism , Molecular Chaperones/metabolism , Multiple Myeloma/metabolism , Neoplasm Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Tumor Cells, Cultured
10.
Br J Haematol ; 190(3): 361-370, 2020 08.
Article in English | MEDLINE | ID: mdl-32350858

ABSTRACT

Hypomethylating agents (HMAs) are widely used in patients with higher-risk myelodysplastic syndromes (MDS) not eligible for stem cell transplantation; however, the response rate is <50%. Reliable predictors of response are still missing, and it is a major challenge to develop new treatment strategies. One current approach is the combination of azacytidine (AZA) with checkpoint inhibitors; however, the potential benefit of targeting the immunomodulator indoleamine-2,3-dioxygenase (IDO-1) has not yet been evaluated. We observed moderate to strong IDO-1 expression in 37% of patients with high-risk MDS. IDO-1 positivity was predictive of treatment failure and shorter overall survival. Moreover, IDO-1 positivity correlated inversely with the number of infiltrating CD8+ T cells, and IDO-1+ patients failed to show an increase in CD8+ T cells under AZA treatment. In vitro experiments confirmed tryptophan catabolism and depletion of CD8+ T cells in IDO-1+ MDS, suggesting that IDO-1 expression induces an immunosuppressive microenvironment in MDS, thereby leading to treatment failure under AZA treatment. In conclusion, IDO-1 is expressed in more than one-third of patients with higher-risk MDS, and is predictive of treatment failure and shorter overall survival. Therefore, IDO-1 is emerging as a promising predictor and therapeutic target, especially for combination therapies with HMAs or checkpoint inhibitors.


Subject(s)
Azacitidine/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/blood , Myelodysplastic Syndromes/blood , Aged , Aged, 80 and over , Azacitidine/pharmacology , Biomarkers , Bone Marrow/pathology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Combined Modality Therapy , DNA Methylation/drug effects , Drug Therapy, Combination , Enzyme Induction/drug effects , Female , Hematopoietic Stem Cell Transplantation , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Macrophages/enzymology , Macrophages/ultrastructure , Male , Middle Aged , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/therapy , Prognosis , Risk , Tryptophan/metabolism
11.
Nat Med ; 22(7): 735-43, 2016 07.
Article in English | MEDLINE | ID: mdl-27294876

ABSTRACT

Immunomodulatory drugs (IMiDs), such as thalidomide and its derivatives lenalidomide and pomalidomide, are key treatment modalities for hematologic malignancies, particularly multiple myeloma (MM) and del(5q) myelodysplastic syndrome (MDS). Cereblon (CRBN), a substrate receptor of the CRL4 ubiquitin ligase complex, is the primary target by which IMiDs mediate anticancer and teratogenic effects. Here we identify a ubiquitin-independent physiological chaperone-like function of CRBN that promotes maturation of the basigin (BSG; also known as CD147) and solute carrier family 16 member 1 (SLC16A1; also known as MCT1) proteins. This process allows for the formation and activation of the CD147-MCT1 transmembrane complex, which promotes various biological functions, including angiogenesis, proliferation, invasion and lactate export. We found that IMiDs outcompete CRBN for binding to CD147 and MCT1, leading to destabilization of the CD147-MCT1 complex. Accordingly, IMiD-sensitive MM cells lose CD147 and MCT1 expression after being exposed to IMiDs, whereas IMiD-resistant cells retain their expression. Furthermore, del(5q) MDS cells have elevated CD147 expression, which is attenuated after IMiD treatment. Finally, we show that BSG (CD147) knockdown phenocopies the teratogenic effects of thalidomide exposure in zebrafish. These findings provide a common mechanistic framework to explain both the teratogenic and pleiotropic antitumor effects of IMiDs.


Subject(s)
Basigin/drug effects , Cell Cycle Proteins/drug effects , Immunologic Factors/pharmacology , Immunosuppressive Agents/pharmacology , Oncogene Proteins/drug effects , Peptide Hydrolases/drug effects , RNA, Messenger/drug effects , Teratogenesis/drug effects , Thalidomide/pharmacology , Adaptor Proteins, Signal Transducing , Basigin/genetics , Basigin/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Humans , Lenalidomide , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Teratogenesis/genetics , Thalidomide/analogs & derivatives , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL
...