1
|
Saha G, Ghosh MK. The key vulnerabilities and therapeutic opportunities in the USP7-p53/MDM2 axis in cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119908. [PMID: 39880128 DOI: 10.1016/j.bbamcr.2025.119908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/10/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
The MDM2/MDMX-p53 circuitry is essential for controlling the development, apoptosis, immune response, angiogenesis, senescence, cell cycle progression, and proliferation of cancer cells. Research has demonstrated that USP7 exerts strong control over p53, MDM2, and MDMX stability, with multiple mediator proteins influencing the USP7-p53-MDM2/MDMX axis to modify p53 expression level and function. In cases where p53 is of the wild type (Wt-p53) in tumors, inhibiting USP7 promotes the degradation of MDM2/MDMX, leading to the activation of p53 signaling. This, in turn, results in cell cycle arrest and apoptosis. Hence, targeting USP7 presents a promising avenue for cancer therapy. Targeting USP7 in tumors that harbor mutant p53 (Mut-p53) is unlikely and remains largely unexplored due to the existence of numerous USP7 targets that function independently of p53. Considering that Mut-p53 exhibits resistance to degradation by MDM2 and other E3 ligases and also shares the same signaling pathways as Wt-p53, it is reasonable to suggest that USP7 may play a role in stabilizing Mut-p53. However, there is still much to be done in this area. If the hypothesis is correct, USP7 may be a potent target in cancers containing both Wt-p53 and Mut-p53.
Collapse
Affiliation(s)
- Gouranga Saha
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata- 700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Mrinal K Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata- 700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India.
| |
Collapse
|
2
|
Martano S, Faktor J, Kote S, Cascione M, Di Corato R, Faktorova D, Semeraro P, Rizzello L, Leporatti S, Rinaldi R, De Matteis V. DIA/SWATH-Mass Spectrometry Revealing Melanoma Cell Proteome Transformations with Silver Nanoparticles: An Innovative Comparative Study. Int J Mol Sci 2025; 26:2029. [PMID: 40076651 PMCID: PMC11901134 DOI: 10.3390/ijms26052029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Melanoma is an aggressive cancer with rising incidence and high mortality rates, largely due to chemotherapy resistance and molecular dysregulation. Nanotechnology, particularly silver nanoparticles (AgNPs), has emerged as a promising therapeutic avenue because of the nanoparticles' ability to induce oxidative stress and apoptosis in cancer cells. However, conventional colloidal AgNPs lack selectivity, often causing significant damage to healthy cells. In this study, we introduce a green synthesis of AgNPs using plant extracts, providing an eco-friendly alternative with improved antitumor selectivity compared to traditional colloidal AgNPs. Leveraging label-free Data-Independent Acquisition/Sequential Window Acquisition of All Theoretical Mass Spectrometry (DIA/SWATH MS) quantitative proteomics, we investigated the antitumor effects of green-synthesized versus traditional AgNPs on A375 melanoma cells at 24 and 48 h. Our findings reveal that green AgNPs selectively reduced melanoma cell viability while sparing healthy keratinocytes (HaCaT), a benefit not observed with colloidal AgNPs. Proteomic analysis highlighted that green AgNPs significantly downregulated oncogenes, enhanced carbohydrate metabolism, and disrupted copper homeostasis in melanoma cells. This marks the first study to explore the differential effects of green and traditional AgNPs on melanoma using an integrated proteomic approach, underscoring the molecular potential of green AgNPs as a targeted and sustainable option for cancer therapy.
Collapse
Affiliation(s)
- Simona Martano
- Department of Mathematics and Physics “Ennio De Giorgi”, University of Salento, Via Arnesano, 73100 Lecce, Italy; (S.M.); (M.C.); (R.R.)
| | - Jakub Faktor
- International Centre for Cancer Vaccine Science, University of Gdansk, Kladki 24, 80-822 Gdansk, Poland;
| | - Sachin Kote
- International Centre for Cancer Vaccine Science, University of Gdansk, Kladki 24, 80-822 Gdansk, Poland;
| | - Mariafrancesca Cascione
- Department of Mathematics and Physics “Ennio De Giorgi”, University of Salento, Via Arnesano, 73100 Lecce, Italy; (S.M.); (M.C.); (R.R.)
- Institute for Microelectronics and Microsystems (IMM), CNR, Via Monteroni, 73100 Lecce, Italy;
| | - Riccardo Di Corato
- Institute for Microelectronics and Microsystems (IMM), CNR, Via Monteroni, 73100 Lecce, Italy;
- Center for Biomolecular Nanotechnologies, Istituto Italiano di Tecnologia (IIT), 73010 Arnesano, Italy
| | - Dagmar Faktorova
- Faculty of Special Technology, Alexander Dubček University of Trenčín, 911 06 Trenčín, Slovakia;
| | - Paola Semeraro
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Monteroni, 73100 Lecce, Italy;
| | - Loris Rizzello
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy;
| | - Stefano Leporatti
- CNR Nanotec-Istituto Di Nanotecnologia, C/O Campus Ecotekne, Via Monteroni, 73100 Lecce, Italy;
| | - Rosaria Rinaldi
- Department of Mathematics and Physics “Ennio De Giorgi”, University of Salento, Via Arnesano, 73100 Lecce, Italy; (S.M.); (M.C.); (R.R.)
- Institute for Microelectronics and Microsystems (IMM), CNR, Via Monteroni, 73100 Lecce, Italy;
| | - Valeria De Matteis
- Institute for Microelectronics and Microsystems (IMM), CNR, Via Monteroni, 73100 Lecce, Italy;
- Department of Experimental Medicine, University of Salento, Via Monteroni, 73100 Lecce, Italy
| |
Collapse
|
3
|
Ray P, Jaiswal S, Ferrer-Torres D, Wang Z, Nancarrow D, Curtin M, Martinho MS, Lacy SM, Kasturirangan S, Thomas D, Spence JR, Truttmann MC, Lagisetty KH, Lawrence TS, Wang TD, Beer DG, Ray D. GRAIL1 Stabilizes Misfolded Mutant p53 through a Ubiquitin Ligase-Independent, Chaperone Regulatory Function. Mol Cancer Res 2024; 22:996-1010. [PMID: 39018356 PMCID: PMC11530312 DOI: 10.1158/1541-7786.mcr-24-0361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/25/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024]
Abstract
Frequent (>70%) TP53 mutations often promote its protein stabilization, driving esophageal adenocarcinoma (EAC) development linked to poor survival and therapy resistance. We previously reported that during Barrett's esophagus progression to EAC, an isoform switch occurs in the E3 ubiquitin ligase RNF128 (aka GRAIL-gene related to anergy in lymphocytes), enriching isoform 1 (hereby GRAIL1) and stabilizing the mutant p53 protein. Consequently, GRAIL1 knockdown degrades mutant p53. But, how GRAIL1 stabilizes the mutant p53 protein remains unclear. In search for a mechanism, here, we performed biochemical and cell biology studies to identify that GRAIL has a binding domain (315-PMCKCDILKA-325) for heat shock protein 40/DNAJ. This interaction can influence DNAJ chaperone activity to modulate misfolded mutant p53 stability. As predicted, either the overexpression of a GRAIL fragment (Frag-J) encompassing the DNAJ binding domain or a cell-permeable peptide (Pep-J) encoding the above 10 amino acids can bind and inhibit DNAJ-Hsp70 co-chaperone activity, thus degrading misfolded mutant p53. Consequently, either Frag-J or Pep-J can reduce the survival of mutant p53 containing dysplastic Barrett's esophagus and EAC cells and inhibit the growth of patient-derived organoids of dysplastic Barrett's esophagus in 3D cultures. The misfolded mutant p53 targeting and growth inhibitory effects of Pep-J are comparable with simvastatin, a cholesterol-lowering drug that can degrade misfolded mutant p53 also via inhibiting DNAJA1, although by a distinct mechanism. Implications: We identified a novel ubiquitin ligase-independent, chaperone-regulating domain in GRAIL and further synthesized a first-in-class novel misfolded mutant p53 degrading peptide having future translational potential.
Collapse
Affiliation(s)
- Paramita Ray
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109
| | - Sangeeta Jaiswal
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | | | - Zhuwen Wang
- Department of Thoracic Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Derek Nancarrow
- Department of Thoracic Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Meghan Curtin
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109
| | - May San Martinho
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109
| | - Shannon M. Lacy
- Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109
| | | | - Dafydd Thomas
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Jason R. Spence
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Matthias C. Truttmann
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109
| | - Kiran H. Lagisetty
- Department of Thoracic Surgery, University of Michigan, Ann Arbor, MI 48109
| | | | - Thomas D. Wang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - David G. Beer
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Dipankar Ray
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
4
|
Li Y, Du Y, Li R, Zhong W, Zou X, Li L, Xu L, Wu L, Che X. Spatial transcriptomics in pancreatic cancer: Advances, prospects and challenges. Crit Rev Oncol Hematol 2024; 203:104430. [PMID: 38942220 DOI: 10.1016/j.critrevonc.2024.104430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024] Open
Abstract
Pancreatic cancer remains one of the deadliest malignancies with an overall 5-year survival rate of 13 %. This dismal fact can be partly attributed to currently limited understanding of tumor heterogeneity and immune microenvironment. Traditional bulk-sequencing techniques overlook the diversity of tumor cells, while single-cell sequencing disorganizes the position localizing of cells in tumor microenvironment. The advent of spatial transcriptomics (ST) presents a novel solution by integrating location and whole transcript expression information. This technology allows for detailed observation of spatio-temporal changes across various cell subtypes within the pancreatic tumor microenvironment, providing insights into their potential functions. This review offers an overview of recent studies implementing ST in pancreatic cancer research, highlighting its instrumental role in investigating the heterogeneity and functions of tumor cells, stromal cells, and immune cells. On the basis, we also prospected and summarized the clinical application scenarios, technical limitations and challenges of ST technology in pancreatic cancer.
Collapse
Affiliation(s)
- Yunlong Li
- Department of Pancreatic and Gastric Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yongxing Du
- Department of Pancreatic and Gastric Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Rui Li
- BGI, Shenzhen 518083, China; BGI Research, Shenzhen 518083, China; Institute of Intelligent Medical Research (IIMR), BGI Genomics, Shenzhen 518083, China
| | - Wenhui Zhong
- Department of Pancreatic and Gastric Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xuanxuan Zou
- BGI, Shenzhen 518083, China; BGI Research, Chongqing 401329, China; BGI Research, Shenzhen 518083, China
| | - Liji Li
- College of Life Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Lin Xu
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518172, China
| | - Liang Wu
- BGI, Shenzhen 518083, China; BGI Research, Chongqing 401329, China; BGI Research, Shenzhen 518083, China.
| | - Xu Che
- Department of Pancreatic and Gastric Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518172, China.
| |
Collapse
|
5
|
Pang B, Wang Q, Chen H, Liu Z, Han M, Gong J, Yue L, Ding X, Wang S, Yan Z, Chen Y, Malouf D, Bucci J, Guo T, Zhou C, Jiang J, Li Y. Proteomic Identification of Small Extracellular Vesicle Proteins LAMB1 and Histone H4 for Prostate Cancer Diagnosis and Risk Stratification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402509. [PMID: 38590132 PMCID: PMC11187897 DOI: 10.1002/advs.202402509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Indexed: 04/10/2024]
Abstract
Diagnosis and stratification of prostate cancer (PCa) patients using the prostate-specific antigen (PSA) test is challenging. Extracellular vesicles (EVs), as a new star of liquid biopsy, has attracted interest to complement inaccurate PSA screening and invasiveness of tissue biopsy. In this study, a panel of potential small EV (sEV) protein biomarkers is identified from PCa cell lines using label-free LC-MS/MS proteomics. These biomarkers underwent further validation with plasma and urine samples from different PCa stages through parallel reaction monitoring-based targeted proteomics, western blotting, and ELISA. Additionally, a tissue microarray containing cancerous and noncancerous tissues is screened to provide additional evidence of selected sEV proteins associated with cancer origin. Results indicate that sEV protein LAMB1 is highly expressed in human plasma of metastatic PCa patients compared with localised PCa patients and control subjects, while sEV protein Histone H4 is highly expressed in human urine of high-risk PCa patients compared to low-risk PCa patients and control subjects. These two sEV proteins demonstrate higher specificity and sensitivity than the PSA test and show promise for metastatic PCa diagnosis, progression monitoring, and risk stratification.
Collapse
Affiliation(s)
- Bairen Pang
- Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Ningbo Clinical Research Centre for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Translational Research Laboratory for UrologyThe Key Laboratory of NingboThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Zhejiang Engineering Research Center of Innovative technologies and diagnostic and therapeutic equipment for urinary system diseasesNingboZhejiang315010China
| | - Qi Wang
- Cancer Care CentreSt George HospitalKogarahNSW2217Australia
- St. George and Sutherland Clinical CampusesSchool of Clinical MedicineUNSW SydneyKensingtonNSW2052Australia
| | - Haotian Chen
- Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Ningbo Clinical Research Centre for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Health Science CentreNingbo UniversityNingboZhejiang315211China
| | - Zhihan Liu
- Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Ningbo Clinical Research Centre for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Health Science CentreNingbo UniversityNingboZhejiang315211China
| | - Meng Han
- Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Ningbo Clinical Research Centre for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Translational Research Laboratory for UrologyThe Key Laboratory of NingboThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Zhejiang Engineering Research Center of Innovative technologies and diagnostic and therapeutic equipment for urinary system diseasesNingboZhejiang315010China
| | - Jie Gong
- Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Ningbo Clinical Research Centre for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Translational Research Laboratory for UrologyThe Key Laboratory of NingboThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
| | - Liang Yue
- Westlake Centre for Intelligent ProteomicsWestlake Laboratory of Life Sciences and BiomedicineHangzhouZhejiang310030China
- Key Laboratory of Structural Biology of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhouZhejiang310030China
| | - Xuan Ding
- Westlake Centre for Intelligent ProteomicsWestlake Laboratory of Life Sciences and BiomedicineHangzhouZhejiang310030China
- Key Laboratory of Structural Biology of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhouZhejiang310030China
| | - Suying Wang
- Department of PathologyNingbo Diagnostic Pathology CentreNingboZhejiang315021China
| | - Zejun Yan
- Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
| | - Yingzhi Chen
- Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
| | - David Malouf
- Department of UrologySt George HospitalKogarahNSW2217Australia
| | - Joseph Bucci
- Cancer Care CentreSt George HospitalKogarahNSW2217Australia
- St. George and Sutherland Clinical CampusesSchool of Clinical MedicineUNSW SydneyKensingtonNSW2052Australia
| | - Tiannan Guo
- Westlake Centre for Intelligent ProteomicsWestlake Laboratory of Life Sciences and BiomedicineHangzhouZhejiang310030China
- Key Laboratory of Structural Biology of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhouZhejiang310030China
| | - Cheng Zhou
- Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Ningbo Clinical Research Centre for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Translational Research Laboratory for UrologyThe Key Laboratory of NingboThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Zhejiang Engineering Research Center of Innovative technologies and diagnostic and therapeutic equipment for urinary system diseasesNingboZhejiang315010China
| | - Junhui Jiang
- Department of UrologyThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Ningbo Clinical Research Centre for Urological DiseaseThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Translational Research Laboratory for UrologyThe Key Laboratory of NingboThe First Affiliated Hospital of Ningbo UniversityNingboZhejiang315010China
- Zhejiang Engineering Research Center of Innovative technologies and diagnostic and therapeutic equipment for urinary system diseasesNingboZhejiang315010China
| | - Yong Li
- Cancer Care CentreSt George HospitalKogarahNSW2217Australia
- St. George and Sutherland Clinical CampusesSchool of Clinical MedicineUNSW SydneyKensingtonNSW2052Australia
| |
Collapse
|
6
|
Li D, Kok CYL, Wang C, Ray D, Osterburg S, Dötsch V, Ghosh S, Sabapathy K. Dichotomous transactivation domains contribute to growth inhibitory and promotion functions of TAp73. Proc Natl Acad Sci U S A 2024; 121:e2318591121. [PMID: 38739802 PMCID: PMC11127001 DOI: 10.1073/pnas.2318591121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/22/2024] [Indexed: 05/16/2024] Open
Abstract
The transcription factor p73, a member of the p53 tumor-suppressor family, regulates cell death and also supports tumorigenesis, although the mechanistic basis for the dichotomous functions is poorly understood. We report here the identification of an alternate transactivation domain (TAD) located at the extreme carboxyl (C) terminus of TAp73β, a commonly expressed p73 isoform. Mutational disruption of this TAD significantly reduced TAp73β's transactivation activity, to a level observed when the amino (N)-TAD that is similar to p53's TAD, is mutated. Mutation of both TADs almost completely abolished TAp73β's transactivation activity. Expression profiling highlighted a unique set of targets involved in extracellular matrix-receptor interaction and focal adhesion regulated by the C-TAD, resulting in FAK phosphorylation, distinct from the N-TAD targets that are common to p53 and are involved in growth inhibition. Interestingly, the C-TAD targets are also regulated by the oncogenic, amino-terminal-deficient DNp73β isoform. Consistently, mutation of C-TAD reduces cellular migration and proliferation. Mechanistically, selective binding of TAp73β to DNAJA1 is required for the transactivation of C-TAD target genes, and silencing DNAJA1 expression abrogated all C-TAD-mediated effects. Taken together, our results provide a mechanistic basis for the dichotomous functions of TAp73 in the regulation of cellular growth through its distinct TADs.
Collapse
Affiliation(s)
- Dan Li
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore168583, Singapore
| | - Catherine Yen Li Kok
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore168583, Singapore
| | - Chao Wang
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore168583, Singapore
| | - Debleena Ray
- Programme in Cancer and Stem Cell Biology, Duke-National University of Singapore (NUS) Medical School, Singapore169857, Singapore
| | - Susanne Osterburg
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University, Frankfurt am Main60438, Germany
| | - Volker Dötsch
- Institute of Biophysical Chemistry and Center for Biomolecular Magnetic Resonance and Cluster of Excellence Macromolecular Complexes (CEF), Goethe University, Frankfurt am Main60438, Germany
| | - Sujoy Ghosh
- Centre for Computational Biology & Programme in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore (NUS) Medical School, Singapore169857, Singapore
| | - Kanaga Sabapathy
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore168583, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore637551, Singapore
| |
Collapse
|
7
|
Zoltsman G, Dang TL, Kuchersky M, Faust O, Silva MS, Ilani T, Wentink AS, Bukau B, Rosenzweig R. A unique chaperoning mechanism in class A JDPs recognizes and stabilizes mutant p53. Mol Cell 2024; 84:1512-1526.e9. [PMID: 38508184 DOI: 10.1016/j.molcel.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 12/14/2023] [Accepted: 02/20/2024] [Indexed: 03/22/2024]
Abstract
J-domain proteins (JDPs) constitute a large family of molecular chaperones that bind a broad spectrum of substrates, targeting them to Hsp70, thus determining the specificity of and activating the entire chaperone functional cycle. The malfunction of JDPs is therefore inextricably linked to myriad human disorders. Here, we uncover a unique mechanism by which chaperones recognize misfolded clients, present in human class A JDPs. Through a newly identified β-hairpin site, these chaperones detect changes in protein dynamics at the initial stages of misfolding, prior to exposure of hydrophobic regions or large structural rearrangements. The JDPs then sequester misfolding-prone proteins into large oligomeric assemblies, protecting them from aggregation. Through this mechanism, class A JDPs bind destabilized p53 mutants, preventing clearance of these oncoproteins by Hsp70-mediated degradation, thus promoting cancer progression. Removal of the β-hairpin abrogates this protective activity while minimally affecting other chaperoning functions. This suggests the class A JDP β-hairpin as a highly specific target for cancer therapeutics.
Collapse
Affiliation(s)
- Guy Zoltsman
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 761000, Israel
| | - Thi Lieu Dang
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH-Alliance, Im Neuenheimer Feld 282, Heidelberg 69120, Germany
| | - Miriam Kuchersky
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 761000, Israel
| | - Ofrah Faust
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 761000, Israel
| | - Micael S Silva
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 761000, Israel
| | - Tal Ilani
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 761000, Israel
| | - Anne S Wentink
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH-Alliance, Im Neuenheimer Feld 282, Heidelberg 69120, Germany; Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333CC Leiden, the Netherlands
| | - Bernd Bukau
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH-Alliance, Im Neuenheimer Feld 282, Heidelberg 69120, Germany.
| | - Rina Rosenzweig
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 761000, Israel.
| |
Collapse
|
8
|
Wang J, Lu C, Wang J, Wang Y, Bi H, Zheng J, Ding X. Necroptosis-related genes allow novel insights into predicting graft loss and diagnosing delayed graft function in renal transplantation. Genomics 2024; 116:110778. [PMID: 38163575 DOI: 10.1016/j.ygeno.2023.110778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 12/13/2023] [Accepted: 12/29/2023] [Indexed: 01/03/2024]
Abstract
Ischemia-reperfusion injury (IRI) is an inevitable pathophysiological phenomenon in kidney transplantation. Necroptosis is an undoubtedly important contributing mechanism in renal IRI. We first screened differentially expressed necroptosis-related genes (DENRGs) from public databases. Eight DENRGs were validated by independent datasets and verified by qRT-PCR in a rat IRI model. We used univariate and multivariate Cox regression analyses to establish a prognostic signature, and graft survival analysis was performed. Immune infiltrating landscape analysis and gene set enrichment analysis (GSEA) were performed to understand the underlying mechanisms of graft loss, which suggested that necroptosis may aggravate the immune response, resulting in graft loss. Subsequently, a delayed graft function (DGF) diagnostic signature was constructed using the Least Absolute Shrinkage and Selection Operator (LASSO) and exhibited robust efficacy in validation datasets. After comprehensively analyzing DENRGs during IRI, we successfully constructed a prognostic signature and DGF predictive signature, which may provide clinical insights for kidney transplant.
Collapse
Affiliation(s)
- Jiale Wang
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Cuinan Lu
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingwen Wang
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Wang
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huanjing Bi
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Zheng
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoming Ding
- Department of Renal Transplantation, Hospital of Nephrology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
9
|
Wu J, Yang Q, Zhu Y, Xia T, Yi L, Wang J, Ren X. DNAJA1 promotes proliferation and metastasis of breast cancer by activating mutant P53/NF-κB pathway. Pathol Res Pract 2023; 252:154921. [PMID: 37977037 DOI: 10.1016/j.prp.2023.154921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE Breast cancer is one of the most common tumors with high malignancy and metastatic rate. DNAJA1 is closely related to tumor progress in several tumors. However, the role and mechanisms of DNAJA1 in the metastasis and proliferation of breast cancer are unknown. METHODS Immunohistochemistry and western blot were used to detect the protein expression genes. In vivo and vitro experiments were performed to evaluate the proliferation, invasive and metastatic abilities of breast cancer cells. RESULTS DNAJA1 was high expressed in 234 cases of breast cancer tissues and associated with metastasis, p53 expression and poor survival for patients. Knock down of DNAJA1 decreased the number of plate clone formation and the OD value of CCK8 assays in breast cancer cells. Depletion of DNAJA1 also in decreased the invasive abilities of breast cancer cells. In vivo, knock down DNAJA1 decreased the growth of subcutaneous tumor and lung metastatic nodes. Mechanically, DNAJA1 could bind with P53-R175H and reduced its degradation. Up regulation of DNAJA1 in mutant P53-R175H breast cancer cell promoted the nuclear translocation of p65, activated NF-κB pathway and enhanced the transcription of its downstream genes such as MMP9, CXCL10 et al. Blockade of NF-κB pathway effectively rescued the effects of DNAJA1 on proliferation and metastasis in breast cancer. CONCLUSION Our study reveals that DNAJA1 is up regulated in breast cancer and promotes breast cancer cells proliferation and metastasis via P53-R175H/NF-κB pathway. It might be a potential prognosis marker for the breast cancer patients.
Collapse
Affiliation(s)
- Jiao Wu
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Department of Pathology, Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China
| | - Qiao Yang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Department of Pathology, Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China
| | - Ye Zhu
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Department of gastroenterology, The People' Hospital of Leshan, Leshan 644000, Sichuan Province, People's Republic of China; Precision Pathology Diagnosis for Serious Diseases Key Laboratory of Luzhou, Luzhou 646000, Sichuan Province, People's Republic of China
| | - Tian Xia
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Precision Pathology Diagnosis for Serious Diseases Key Laboratory of Luzhou, Luzhou 646000, Sichuan Province, People's Republic of China
| | - Lizhi Yi
- Department of gastroenterology, The People' Hospital of Leshan, Leshan 644000, Sichuan Province, People's Republic of China
| | - Jianmei Wang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Department of Pathology, Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Precision Pathology Diagnosis for Serious Diseases Key Laboratory of Luzhou, Luzhou 646000, Sichuan Province, People's Republic of China.
| | - Xiaoli Ren
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Department of Pathology, Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Precision Pathology Diagnosis for Serious Diseases Key Laboratory of Luzhou, Luzhou 646000, Sichuan Province, People's Republic of China.
| |
Collapse
|
10
|
Li M, Sun D, Song N, Chen X, Zhang X, Zheng W, Yu Y, Han C. Mutant p53 in head and neck squamous cell carcinoma: Molecular mechanism of gain‑of‑function and targeting therapy (Review). Oncol Rep 2023; 50:162. [PMID: 37449494 PMCID: PMC10394732 DOI: 10.3892/or.2023.8599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most widespread malignancies worldwide. p53, as a transcription factor, can play its role in tumor suppression by activating the expression of numerous target genes. However, p53 is one of the most commonly mutated genes, which frequently harbors missense mutations. These missense mutations are nucleotide substitutions that result in the substitution of an amino acid in the DNA binding domain. Most p53 mutations in HNSCC are missense mutations and the mutation rate of p53 reaches 65‑85%. p53 mutation not only inhibits the tumor suppressive function of p53 but also provides novel functions to facilitate tumor recurrence, called gain‑of‑function (GOF). The present study focused on the prevalence and clinical relevance of p53 mutations in HNSCC, and further described how mutant p53 accumulates. Moreover, mutant p53 in HNSCC can interact with proteins, RNA, and exosomes to exert effects on proliferation, migration, invasion, immunosuppression, and metabolism. Finally, several treatment strategies have been proposed to abolish the tumor‑promoting function of mutant p53; these strategies include reactivation of mutant p53 into wild‑type p53, induction of mutant p53 degradation, enhancement of the synthetic lethality of mutant p53, and treatment with immunotherapy. Due to the high frequency of p53 mutations in HNSCC, a further understanding of the mechanism of mutant p53 may provide potential applications for targeted therapy in patients with HNSCC.
Collapse
Affiliation(s)
- Minmin Li
- School of Stomatology, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Dongyuan Sun
- School of Stomatology, Weifang Medical University, Weifang, Shandong 261000, P.R. China
- Department of Dentistry, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Ning Song
- School of Stomatology, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Xi Chen
- School of Stomatology, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Xinyue Zhang
- School of Stomatology, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Wentian Zheng
- School of Stomatology, Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Yang Yu
- School of Stomatology, Weifang Medical University, Weifang, Shandong 261000, P.R. China
- Department of Dentistry, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Chengbing Han
- Department of Stomatology, First Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
| |
Collapse
|
11
|
Wang Y, Abazid A, Badendieck S, Mustea A, Stope MB. Impact of Non-Invasive Physical Plasma on Heat Shock Protein Functionality in Eukaryotic Cells. Biomedicines 2023; 11:biomedicines11051471. [PMID: 37239142 DOI: 10.3390/biomedicines11051471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/06/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Recently, biomedical research has increasingly investigated physical plasma as an innovative therapeutic approach with a number of therapeutic biomedical effects. It is known from radiation and chemotherapy that these applications can lead to the induction and activation of primarily cytoprotective heat shock proteins (HSP). HSP protect cells and tissues from physical, (bio)chemical, and physiological stress and, ultimately, along with other mechanisms, govern resistance and treatment failure. These mechanisms are well known and comparatively well studied in drug therapy. For therapies in the field of physical plasma medicine, however, extremely little data are available to date. In this review article, we provide an overview of the current studies on the interaction of physical plasma with the cellular HSP system.
Collapse
Affiliation(s)
- Yanqing Wang
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Alexander Abazid
- Department of General, Visceral and Thorax Surgery, Bundeswehr Hospital Berlin, Scharnhorststrasse 13, 10115 Berlin, Germany
| | - Steffen Badendieck
- Department of General, Visceral and Thorax Surgery, Bundeswehr Hospital Berlin, Scharnhorststrasse 13, 10115 Berlin, Germany
| | - Alexander Mustea
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Matthias B Stope
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
12
|
He TQ, Zhao YW, Ning F, Liu Y, Tu L, He J. Development and validation of a prognostic model based on a single-cell RNA-seq in Wilms tumor in children. J Investig Med 2023; 71:173-182. [PMID: 36718830 DOI: 10.1177/10815589221143739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
To analyze the heterogeneity between different cell types in pediatric Wilms tumor (WT) tissue, and identify the differentially expressed genes (DEGs) of malignant tumor cells, thereby establishing a prognostic model. The single-cell sequencing data of pediatric WT tissues were downloaded from the public database. Data filtration and normalization, principal component analysis, and T-distributed stochastic neighbor embedding cluster analysis were performed using the Seurat package of R language. Cells were divided into different clusters, malignant tumor cells were extracted, and DEGs were obtained. Then, the pseudo-time trajectory analysis was performed. Prognostic biomarkers were determined by univariate and multivariate COX regression analyses and LASSO regression analysis. Kaplan-Meier survival analysis and receiver operator characteristic curve analysis were performed. Combined with the prognostic biomarkers and clinical characteristics, a nomogram was generated to predict WT prognosis. The prognostic power was validated in the external datasets. Cells in the WT tissue were divided into 10 clusters. Three prognostic biomarkers that affected the survival time of patients were screened from 215 DEGs in malignant tumor cells, and a nomogram was constructed using the three genes and clinical characteristics. The area under the curve (AUC) values of 3- and 5-year disease-free survival were 0.756 and 0.734, respectively. In the external validation dataset, the AUC value of this nomogram model was 0.826. Based on the single-cell RNA-seq, we recognized cell clusters in the WT tissue of children, identified prognostic biomarkers in malignant tumor cells, and established a comprehensive prognostic model. Our findings might provide new ideas and methods for the diagnosis and treatment of WT.
Collapse
Affiliation(s)
- Tian-Qu He
- Department of Urology, Hunan Children's Hospital, Changsha, China
| | - Yao-Wang Zhao
- Department of Urology, Hunan Children's Hospital, Changsha, China
| | - Feng Ning
- Department of Urology, Hunan Children's Hospital, Changsha, China
| | - Yu Liu
- Department of Urology, Hunan Children's Hospital, Changsha, China
| | - Lei Tu
- Department of Urology, Hunan Children's Hospital, Changsha, China
| | - Jun He
- Department of Urology, Hunan Children's Hospital, Changsha, China
| |
Collapse
|
13
|
Nishikawa S, Iwakuma T. Drugs Targeting p53 Mutations with FDA Approval and in Clinical Trials. Cancers (Basel) 2023; 15:429. [PMID: 36672377 PMCID: PMC9856662 DOI: 10.3390/cancers15020429] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/01/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Mutations in the tumor suppressor p53 (p53) promote cancer progression. This is mainly due to loss of function (LOS) as a tumor suppressor, dominant-negative (DN) activities of missense mutant p53 (mutp53) over wild-type p53 (wtp53), and wtp53-independent oncogenic activities of missense mutp53 by interacting with other tumor suppressors or oncogenes (gain of function: GOF). Since p53 mutations occur in ~50% of human cancers and rarely occur in normal tissues, p53 mutations are cancer-specific and ideal therapeutic targets. Approaches to target p53 mutations include (1) restoration or stabilization of wtp53 conformation from missense mutp53, (2) rescue of p53 nonsense mutations, (3) depletion or degradation of mutp53 proteins, and (4) induction of p53 synthetic lethality or targeting of vulnerabilities imposed by p53 mutations (enhanced YAP/TAZ activities) or deletions (hyperactivated retrotransposons). This review article focuses on clinically available FDA-approved drugs and drugs in clinical trials that target p53 mutations and summarizes their mechanisms of action and activities to suppress cancer progression.
Collapse
Affiliation(s)
- Shigeto Nishikawa
- Department of Pediatrics, Division of Hematology & Oncology, Children’s Mercy Research Institute, Kansas City, MO 64108, USA
| | - Tomoo Iwakuma
- Department of Pediatrics, Division of Hematology & Oncology, Children’s Mercy Research Institute, Kansas City, MO 64108, USA
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
14
|
DNAJA1- and conformational mutant p53-dependent inhibition of cancer cell migration by a novel compound identified through a virtual screen. Cell Death Dis 2022; 8:437. [PMID: 36316326 PMCID: PMC9622836 DOI: 10.1038/s41420-022-01229-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Cancers are frequently addicted to oncogenic missense mutant p53 (mutp53). DNAJA1, a member of heat shock protein 40 (HSP40), also known as J-domain proteins (JDPs), plays a crucial role in the stabilization and oncogenic activity of misfolded or conformational mutp53 by binding to and preventing mutp53 from proteasomal degradation. However, strategies to deplete mutp53 are not well-established, and no HSP40/JDPs inhibitors are clinically available. To identify compounds that bind to DNAJA1 and induce mutp53 degradation, we performed an in silico docking study of ~10 million of compounds from the ZINC database for the J-domain of DNAJA1. A compound 7-3 was identified, and its analogue A11 effectively reduced the levels of DNAJA1 and conformational mutp53 with minimal effects on the levels of wild-type p53 and DNA-contact mutp53. A11 suppressed migration and filopodia formation in a manner dependent on DNAJA1 and conformational mutp53. A mutant DNAJA1 with alanine mutations at predicted amino acids (tyrosine 7, lysine 44, and glutamine 47) failed to bind to A11. Cells expressing the mutant DNAJA1 became insensitive to A11-mediated depletion of DNAJA1 and mutp53 as well as A11-mediated inhibition of cell migration. Thus, A11 is the first HSP40/JDP inhibitor that has not been previously characterized for depleting DNAJA1 and subsequently conformational mutp53, leading to inhibition of cancer cell migration. A11 can be exploited for a novel treatment against cancers expressing conformational mutp53.
Collapse
|
15
|
Roth HE, De Lima Leite A, Palermo NY, Powers R. Leveraging the Structure of DNAJA1 to Discover Novel Potential Pancreatic Cancer Therapies. Biomolecules 2022; 12:1391. [PMID: 36291603 PMCID: PMC9599757 DOI: 10.3390/biom12101391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic cancer remains one of the deadliest forms of cancer with a 5-year survival rate of only 11%. Difficult diagnosis and limited treatment options are the major causes of the poor outcome for pancreatic cancer. The human protein DNAJA1 has been proposed as a potential therapeutic target for pancreatic cancer, but its cellular and biological functions remain unclear. Previous studies have suggested that DNAJA1's cellular activity may be dependent upon its protein binding partners. To further investigate this assertion, the first 107 amino acid structures of DNAJA1 were solved by NMR, which includes the classical J-domain and its associated linker region that is proposed to be vital to DNAJA1 functionality. The DNAJA1 NMR structure was then used to identify both protein and ligand binding sites and potential binding partners that may suggest the intracellular roles of DNAJA1. Virtual drug screenings followed by NMR and isothermal titration calorimetry identified 5 drug-like compounds that bind to two different sites on DNAJA1. A pull-down assay identified 8 potentially novel protein binding partners of DNAJA1. These proteins in conjunction with our previously published metabolomics study support a vital role for DNAJA1 in cellular oncogenesis and pancreatic cancer.
Collapse
Affiliation(s)
- Heidi E. Roth
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Aline De Lima Leite
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Nicolas Y. Palermo
- Computational Chemistry Core Facility, VCR Cores, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Robert Powers
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| |
Collapse
|
16
|
Mutant p53 Depletion by Novel Inhibitors for HSP40/J-Domain Proteins Derived from the Natural Compound Plumbagin. Cancers (Basel) 2022; 14:cancers14174187. [PMID: 36077724 PMCID: PMC9454493 DOI: 10.3390/cancers14174187] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 12/05/2022] Open
Abstract
Simple Summary The tumor suppressor p53 is frequently mutated in human cancer. Accumulation of missense mutant p53 (mutp53) in tumors is crucial for malignant progression, and cancers are often addicted to oncogenic mutp53. However, strategies to deplete mutp53 have not yet been established. Recent studies have shown that misfolded or conformational mutp53 is stabilized by DNAJA1, a member of HSP40, also known as J-domain proteins (JDPs). However, no selective DNAJA1 inhibitor is clinically available. Through a molecular docking study, we identified a potential DNAJA1 inhibitor, called PLTFBH, derived from the natural compound plumbagin, as a compound that bound to and reduced protein levels of DNAJA1 and several other HSP40/JDPs. PLTFBH reduced the levels of conformational mutp53 and inhibited cancer cell migration in a manner dependent on DNAJA1 and mutp53. Abstract Accumulation of missense mutant p53 (mutp53) in cancers promotes malignant progression. DNAJA1, a member of HSP40 (also known as J-domain proteins: JDPs), is shown to prevent misfolded or conformational mutp53 from proteasomal degradation. Given frequent addiction of cancers to oncogenic mutp53, depleting mutp53 by DNAJA1 inhibition is a promising approach for cancer therapy. However, there is no clinically available inhibitor for DNAJA1. Our in silico molecular docking study with a natural compound-derived small molecule library identified a plumbagin derivative, PLIHZ (plumbagin–isoniazid analog), as a potential compound binding to the J domain of DNAJA1. PLIHZ efficiently reduced the levels of DNAJA1 and several conformational mutp53 with minimal impact on DNA contact mutp53 and wild-type p53 (wtp53). An analog, called PLTFBH, which showed a similar activity to PLIHZ in reducing DNAJA1 and mutp53 levels, inhibited migration of cancer cells specifically carrying conformational mutp53, but not DNA contact mutp53, p53 null, and wtp53, which was attenuated by depletion of DNAJA1 or mutp53. Moreover, PLTFBH reduced levels of multiple other HSP40/JDPs with tyrosine 7 (Y7) and/or tyrosine 8 (Y8) but failed to deplete DNAJA1 mutants with alanine substitution of these amino acids. Our study suggests PLTFBH as a potential inhibitor for multiple HSP40/JDPs.
Collapse
|
17
|
DNAJA1 Stabilizes EF1A1 to Promote Cell Proliferation and Metastasis of Liver Cancer Mediated by miR-205-5p. JOURNAL OF ONCOLOGY 2022; 2022:2292481. [PMID: 35586205 PMCID: PMC9110222 DOI: 10.1155/2022/2292481] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/05/2022] [Accepted: 03/22/2022] [Indexed: 11/22/2022]
Abstract
Liver cancer is one of the most common and aggressive malignancies worldwide with poor prognosis. Studies on pathogenesis of liver cancer are urgently demanded to develop better treatment strategy. Here, we found that overexpression of DnaJ heat shock protein family (Hsp40) member A1 (DNAJA1) increased cell proliferation, invasion, and angiogenesis in Huh 7 and HepG2 cells, while depletion of DNAJA1 in MHCC-97H and HCC-M3 showed opposite effects. In vivo functional assays indicated that DNAJA1 promoted tumor growth and pulmonary metastasis in mice. Mechanistically, as a direct target of miR-205-5p, DNAJA1 promoted proliferation and metastasis of liver cancer cells by stabilizing eukaryotic elongation factor 1A1 (EF1A1). Moreover, DNAJA was markedly upregulated in liver cancer tissues (P < 0.05) and was significantly associated with poor prognosis. And its expression was correlated with differentiation (P < 0.001), dissemination (P < 0.001), and serum AFP (P = 0.029). The mRNA levels of miR-205-5p and DNAJA1 were negatively correlated in liver cancer. In conclusion, our study reveals that DNAJA1 acts as an oncogene in liver cancer via miR-205-5p/EF1A1 axis and might be a potential biomarker to predict the prognosis for liver cancer patients.
Collapse
|
18
|
Li G, Zhang S, Wang H, Liang L, Liu Z, Wang Y, Xu B, Zhao H. Differential Expression Characterisation of the Heat Shock Proteins DnaJB6, DnaJshv, DnaJB13, and DnaJB14 in Apis cerana cerana Under Various Stress Conditions. Front Ecol Evol 2022. [DOI: 10.3389/fevo.2022.873791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
As key pollinators, bees are frequently exposed to multiple environmental stresses and have developed crucial mechanisms by which they adapt to these stressors. However, the molecular bases mediated at the gene level remain to be discovered. Here, we found four heat shock protein DnaJB subfamily genes, DnaJB6, DnaJshv, DnaJB13, and DnaJB14, from Apis cerana cerana, that all have J domains in their protein sequences. The expression levels of DnaJB6 and DnaJshv were upregulated by different degrees of heat stress, and the transcript level of DnaJB14 was gradually upregulated as the degree of heat stress increased, while the mRNA level of DnaJB13 was downregulated at multiple time points during heat stress treatment. The mRNA levels of all four DnaJBs were upregulated by cold and UV stress. In addition, the expression levels of DnaJB6, DnaJshv and DnaJB13 were reduced under abamectin, imidacloprid, cypermethrin, bifenthrin, spirodiclofen, and methomyl stresses. The transcript level of DnaJB14 was decreased by imidacloprid, cypermethrin, spirodiclofen, and methomyl exposure but increased by abamectin and bifenthrin exposure. These results indicate that the demand of A. cerana cerana for these four DnaJBs differs under various stress conditions. To further explore the role of DnaJBs in the stress response, we successfully silenced DnaJshv and DnaJB14. The content of protein carbonyl was increased, while the content of VC, the enzymatic activities of CAT, GST, and SOD, the mRNA levels of many antioxidant-related genes, and the total antioxidant capacity were reduced after knockdown of DnaJshv and DnaJB14 in A. cerana cerana. These results indicate that silencing DnaJshv and DnaJB14 increases oxidative damage and decreases the antioxidant ability of A. cerana cerana. Taken together, our results demonstrate that DnaJB6, DnaJshv, DnaJB13, and DnaJB14 are differentially expressed under stress conditions and play crucial roles in response to various stressors, possibly through the antioxidant signalling pathway. These findings will be conducive to understanding the molecular basis of bee responses to environmental stresses and are beneficial for improving bee protection.
Collapse
|
19
|
Kaida A, Iwakuma T. Regulation of p53 and Cancer Signaling by Heat Shock Protein 40/J-Domain Protein Family Members. Int J Mol Sci 2021; 22:13527. [PMID: 34948322 PMCID: PMC8706882 DOI: 10.3390/ijms222413527] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/27/2022] Open
Abstract
Heat shock proteins (HSPs) are molecular chaperones that assist diverse cellular activities including protein folding, intracellular transportation, assembly or disassembly of protein complexes, and stabilization or degradation of misfolded or aggregated proteins. HSP40, also known as J-domain proteins (JDPs), is the largest family with over fifty members and contains highly conserved J domains responsible for binding to HSP70 and stimulation of the ATPase activity as a co-chaperone. Tumor suppressor p53 (p53), the most frequently mutated gene in human cancers, is one of the proteins that functionally interact with HSP40/JDPs. The majority of p53 mutations are missense mutations, resulting in acquirement of unexpected oncogenic activities, referred to as gain of function (GOF), in addition to loss of the tumor suppressive function. Moreover, stability and levels of wild-type p53 (wtp53) and mutant p53 (mutp53) are crucial for their tumor suppressive and oncogenic activities, respectively. However, the regulatory mechanisms of wtp53 and mutp53 are not fully understood. Accumulating reports demonstrate regulation of wtp53 and mutp53 levels and/or activities by HSP40/JDPs. Here, we summarize updated knowledge related to the link of HSP40/JDPs with p53 and cancer signaling to improve our understanding of the regulation of tumor suppressive wtp53 and oncogenic mutp53 GOF activities.
Collapse
Affiliation(s)
- Atsushi Kaida
- Department of Oral Radiation Oncology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan;
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Tomoo Iwakuma
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Pediatrics, Children’s Mercy Research Institute, Kansas City, MO 64108, USA
| |
Collapse
|