1
|
Clune S, Awolade P, Esquer H, Zhou Q, LaBarbera DV. CHD1L in cancer and beyond: structure, oncogenic functions, and therapeutic potential. J Exp Clin Cancer Res 2025; 44:167. [PMID: 40442742 PMCID: PMC12123854 DOI: 10.1186/s13046-025-03428-1] [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: 04/10/2025] [Accepted: 05/25/2025] [Indexed: 06/02/2025] Open
Abstract
Chromodomain Helicase DNA-binding protein 1-Like (CHD1L) is a chromatin remodeling enzyme increasingly recognized as an oncogenic factor promoting tumor progression and metastatic potential by orchestrating transcriptional programs that drive epithelial-mesenchymal transition (EMT), cytoskeletal remodeling, and metastatic dissemination. In parallel, CHD1L has emerged as a master regulator of tumor cell survival by regulating DNA damage response and repair and enforcing G1 cell cycle progression. Furthermore, CHD1L plays a key role in immune evasion pathways by regulating signaling cascades and by suppressing both apoptotic and non-apoptotic cell death. In particular, CHD1L is a key suppressor of PARthanatos, a caspase-independent mechanism triggered by poly(ADP-ribose) (PAR) polymer fragmentation and apoptosis-inducing factor (AIF) activation. By regulating SPOCK1, MDM2, and TCTP, CHD1L further supports survival under cellular stress. Its overexpression correlates with metastasis, therapy resistance, and poor prognosis across many solid tumors. This review covers CHD1L's structure, oncogenic functions, and developmental origins, and highlights emerging therapeutic strategies that target CHD1L as a druggable vulnerability in cancer.
Collapse
Affiliation(s)
- Sophia Clune
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Paul Awolade
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Center for Drug Discovery, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Hector Esquer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Center for Drug Discovery, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Qiong Zhou
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Center for Drug Discovery, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Daniel V LaBarbera
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Center for Drug Discovery, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
2
|
Esquer H, Zhou Q, LaBarbera DV. Targeted Inhibition of CHD1L by OTI-611 Reprograms Chemotherapy and Targeted Therapy-Induced Cell Cycle Arrest and Suppresses Proliferation to Produce Synergistic Antitumor Effects in Breast and Colorectal Cancer. Cells 2025; 14:318. [PMID: 40072047 PMCID: PMC11898988 DOI: 10.3390/cells14050318] [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: 01/06/2025] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 03/15/2025] Open
Abstract
The second and third most frequently diagnosed cancers worldwide are breast (2.3 million new cases) and colorectal (1.9 million new cases), respectively. Although advances in cancer therapies and early detection have improved the overall survival of patients, patients still develop resistance or cancer recurrence. Thus, the development of novel therapies that can affect multiple mechanisms of drug resistance and cell survival is ideal for the treatment of advanced and metastatic cancers. CHD1L is a novel oncogenic protein involved in regulating chromatin remodeling, DNA damage repair, epithelial-mesenchymal transition (EMT), and programmed cell death via PARthanatos. Herein, we assess in real-time how the CHD1L inhibitor (CHD1Li) OTI-611 modulates cell cycle progression in Colo678, SUM149PT, and SW620 cell lines. By utilizing a cell cycle reporter, we tracked the real-time cell cycle progression of cancer cells treated with OTI-611 alone and in combination with standard-of-care (SOC) therapies. Our results indicate that OTI-611 causes G1 phase cell cycle arrest through a CHD1L-mediated mechanism that regulates Cyclin D1 expression and localization. As a result of this mechanism, OTI-611 can reprogram the cell cycle effects of other antitumor agents to modulate and arrest cells in G1 when used in combination, including agents commonly known to arrest cells in the G2/M phase. Therefore, we conclude that OTI-611-induced G1 arrest represents a critical component of its unique mechanism of action, contributing significantly to its anticancer activity.
Collapse
Affiliation(s)
- Hector Esquer
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (H.E.); (Q.Z.)
- The Center for Drug Discovery, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- The University of Colorado Cancer Center, Aurora, CO 80045, USA
| | - Qiong Zhou
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (H.E.); (Q.Z.)
- The Center for Drug Discovery, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- The University of Colorado Cancer Center, Aurora, CO 80045, USA
| | - Daniel V. LaBarbera
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (H.E.); (Q.Z.)
- The Center for Drug Discovery, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- The University of Colorado Cancer Center, Aurora, CO 80045, USA
| |
Collapse
|
3
|
Zhang X, Zhang X, Yin H, Li Q, Fan B, Jiang B, Xie A, Guo D, Hao H, Zhang B. Roles of SPOCK1 in the Formation Mechanisms and Treatment of Non-Small-Cell Lung Cancer and Brain Metastases from Lung Cancer. Onco Targets Ther 2025; 18:35-47. [PMID: 39835273 PMCID: PMC11745074 DOI: 10.2147/ott.s483576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025] Open
Abstract
Lung cancer is a malignant tumor with high morbidity and mortality in China and worldwide. Once it metastasizes to the brain, its prognosis is very poor. Brain metastases are found in about 20% of newly diagnosed non-small-cell lung cancer (NSCLC) patients. About 30% of NSCLC patients develop brain metastases during treatment. NSCLC that is positive for EGFR, ALK, and ROS1 variations is especially likely to metastasize to the brain. SPOCK1 is a proteoglycan with systemic physiological functions. It regulates the self-renewal of brain metastasis-initiating cells, regulates invasion and metastasis from the lung to the brain, plays an important role in tumor progression and treatment resistance, and has higher expression in metastatic tumor tissues than other tissues. Current treatments for NSCLC brain metastases include surgery, whole-brain radiotherapy, stereotactic radiotherapy, targeted therapy, and chemotherapy. SPOCK1 is involved in many signaling pathways, by which it influences a variety of NSCLC treatment methods. In this paper, the progress of research on the treatment of NSCLC brain metastases is reviewed to guide decisions on treatment options in clinical practice.
Collapse
Affiliation(s)
- Xuebing Zhang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Xia Zhang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
- Department of Oncology, Dalian Fifth People’s Hospital, Dalian, Liaoning, People’s Republic of China
| | - Hang Yin
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Qizheng Li
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Buqun Fan
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Bolun Jiang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Anqi Xie
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Dandan Guo
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| | - Huanling Hao
- Department of Oncology, Dandong First Hospital, Dandong, Liaoning, People’s Republic of China
| | - Bin Zhang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, People’s Republic of China
| |
Collapse
|
4
|
Sala R, Esquer H, Kellett T, Clune S, Awolade P, Pike LA, Zhou Q, Messersmith WA, LaBarbera DV. CHD1L Inhibitor OTI-611 Synergizes with Chemotherapy to Enhance Antitumor Efficacy and Prolong Survival in Colorectal Cancer Mouse Models. Int J Mol Sci 2024; 25:13160. [PMID: 39684869 DOI: 10.3390/ijms252313160] [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: 11/02/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent and deadly forms of cancer. It is universally treated with a combination of the DNA damaging chemotherapy drugs irinotecan, 5-Fluorouracil (5-FU), and oxaliplatin. CHD1L is a novel oncogene that plays critical roles in chromatin remodeling and DNA damage repair, as well as the regulation of malignant gene expression. We show that an inhibitor of CHD1L, OTI-611, when combined with chemotherapy significantly increases DNA damage in CRC cell lines. OTI-611 also synergizes with SN-38, 5-FU, and oxaliplatin in killing CRC tumor organoids. We also demonstrate that, as in breast cancer, OTI-611 traps CHD1L, PARP1, and PARP2 onto chromatin. The entrapment of CHD1L causes the deprotection of PAR chains in the nucleus, ultimately resulting in cell death by CHD1Li-mediated PARthanatos, as measured by AIF translocation to the nucleus. Finally, the combination of low doses of OTI-611 with irinotecan significantly reduces tumor volume and extends survival in CRC xenograft mouse models compared to irinotecan alone. The combination of standard of care chemotherapy drugs with CHD1Li represents a promising advancement in future therapeutic strategies for CRC and other cancers driven by CHD1L.
Collapse
Affiliation(s)
- Rita Sala
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hector Esquer
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- The CU Anschutz Center for Drug Discovery, Aurora, CO 80045, USA
- The University of Colorado Cancer Center, Aurora, CO 80045, USA
| | - Timothy Kellett
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sophia Clune
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Paul Awolade
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- The CU Anschutz Center for Drug Discovery, Aurora, CO 80045, USA
| | - Laura A Pike
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Qiong Zhou
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- The CU Anschutz Center for Drug Discovery, Aurora, CO 80045, USA
- The University of Colorado Cancer Center, Aurora, CO 80045, USA
| | - Wells A Messersmith
- The CU Anschutz Center for Drug Discovery, Aurora, CO 80045, USA
- The University of Colorado Cancer Center, Aurora, CO 80045, USA
- Division of Medical Oncology, The School of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Daniel V LaBarbera
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- The CU Anschutz Center for Drug Discovery, Aurora, CO 80045, USA
- The University of Colorado Cancer Center, Aurora, CO 80045, USA
| |
Collapse
|
5
|
Ma J, Yang L, Wu J, Huang Z, Zhang J, Liu M, Li M, Luo J, Wang H. Unraveling the Molecular Mechanisms of SIRT7 in Angiogenesis: Insights from Substrate Clues. Int J Mol Sci 2024; 25:11578. [PMID: 39519130 PMCID: PMC11546391 DOI: 10.3390/ijms252111578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Angiogenesis, a vital physiological or pathological process regulated by complex molecular networks, is widely implicated in organismal development and the pathogenesis of various diseases. SIRT7, a member of the Sirtuin family of nicotinamide adenine dinucleotide + (NAD+) dependent deacetylases, plays crucial roles in cellular processes such as transcriptional regulation, cell metabolism, cell proliferation, and genome stability maintenance. Characterized by its enzymatic activities, SIRT7 targets an array of substrates, several of which exert regulatory effects on angiogenesis. Experimental evidence from in vitro and in vivo studies consistently demonstrates the effects of SIRT7 in modulating angiogenesis, mediated through various molecular mechanisms. Consequently, understanding the regulatory role of SIRT7 in angiogenesis holds significant promise, offering novel avenues for therapeutic interventions targeting either SIRT7 or angiogenesis. This review delineates the putative molecular mechanisms by which SIRT7 regulates angiogenesis, taking its substrates as a clue, endeavoring to elucidate experimental observations by integrating knowledge of SIRT7 substrates and established angiogenenic mechanisms.
Collapse
Affiliation(s)
- Junjie Ma
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
| | - Liqian Yang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
| | - Jiaxing Wu
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
| | - Zhihong Huang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
| | - Jiaqi Zhang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
| | - Minghui Liu
- Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, Beijing 100191, China; (M.L.); (M.L.)
| | - Meiting Li
- Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, Beijing 100191, China; (M.L.); (M.L.)
| | - Jianyuan Luo
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
- Department of Medical Genetics, Center for Medical Genetics, Peking University Health Science Center, Beijing 100191, China; (M.L.); (M.L.)
| | - Haiying Wang
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; (J.M.); (L.Y.); (J.W.); (Z.H.); (J.Z.); (J.L.)
| |
Collapse
|
6
|
Chen S, Fan Y, Sun Y, Li S, Zheng Z, Chu C, Li L, Yin C. Identification and functional characteristics of CHD1L gene variants implicated in human Müllerian duct anomalies. Biol Res 2024; 57:68. [PMID: 39342328 PMCID: PMC11437902 DOI: 10.1186/s40659-024-00550-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Müllerian duct anomalies (MDAs) are congenital developmental disorders that present as a series of abnormalities within the reproductive tracts of females. Genetic factors are linked to MDAs and recent advancements in whole-exome sequencing (WES) provide innovative perspectives in this field. However, relevant mechanism has only been investigated in a restricted manner without clear elucidation of respective observations. METHODS Our previous study reported that 2 of 12 patients with MDAs harbored the CHD1L variant c.348-1G>C. Subsequently, an additional 85 MDAs patients were recruited. Variants in CHD1L were screened through the in-house database of WES performed in the cohort and two cases were identified. One presented with partial septate uterus with left renal agenesis and the other with complete septate uterus, duplicated cervices and longitudinal vaginal septum. The pathogenicity of the discovered variants was further assessed by molecular dynamics simulation and various functional assays. RESULTS Ultimately, two novel heterozygous CHD1L variants, including a missense variant c.956G>A (p.R319Q) and a nonsense variant c.1831C>T (p.R611*) were observed. The variants were absent in 100 controls. Altogether, the contribution yield of CHD1L to MDAs was calculated as 4.12% (4/97). All three variants were assessed as pathogenic through various functional analysis. The splice-site variant c.348-1G>C resulted in a 11 bp sequence skipping in exon 4 of CHD1L and led to nonsense mediated decay of its transcripts. Unlike WT CHD1L, the truncated R611* protein mislocalized to the cytoplasm, abolish the ability of CHD1L to promote cell migration and failed to interact with PARP1 owing to the loss of macro domain. The R319Q variant exhibited conformational disparities and showed abnormal protein recruitment behavior through laser microirradiation comparing with the WT CHD1L. All these variants impaired the CHD1L function in DNA damage repair, thus participating in MDAs. CONCLUSIONS The current study not only expands the mutational spectrum of CHD1L in MDAs but determines three variants as pathogenic according to ACMG guidelines with reliable functional evidence. Additionally, the impairment in DNA damage repair is an underlying mechanism involved in MDAs.
Collapse
Affiliation(s)
- Shuya Chen
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 QiHeLou Street, Dongcheng District, Beijing, 100006, China
| | - Yali Fan
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 QiHeLou Street, Dongcheng District, Beijing, 100006, China
| | - Yujun Sun
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 QiHeLou Street, Dongcheng District, Beijing, 100006, China
| | - Shenghui Li
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 251 YaoJiaYuan Road, Chaoyang District, Beijing, 100026, China
| | - Zhi Zheng
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 QiHeLou Street, Dongcheng District, Beijing, 100006, China
| | - Chunfang Chu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 251 YaoJiaYuan Road, Chaoyang District, Beijing, 100026, China.
| | - Lin Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 QiHeLou Street, Dongcheng District, Beijing, 100006, China.
| | - Chenghong Yin
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, 17 QiHeLou Street, Dongcheng District, Beijing, 100006, China.
| |
Collapse
|
7
|
Sala R, Esquer H, Kellett T, Kearns JT, Awolade P, Zhou Q, LaBarbera DV. CHD1L Regulates Cell Survival in Breast Cancer and Its Inhibition by OTI-611 Impedes the DNA Damage Response and Induces PARthanatos. Int J Mol Sci 2024; 25:8590. [PMID: 39201277 PMCID: PMC11354643 DOI: 10.3390/ijms25168590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
The Chromodomain helicase DNA-binding protein 1-like (CHD1L) is a nucleosome remodeling enzyme, which plays a key role in chromatin relaxation during the DNA damage response. Genome editing has shown that deletion of CHD1L sensitizes cells to PARPi, but the effect of its pharmacological inhibition has not been defined. Triple-negative breast cancer SUM149PT, HCC1937, and MDA-MB-231 cells were used to assess the mechanism of action of the CHD1Li OTI-611. Cytotoxicity as a single agent or in combination with standard-of-care treatments was assessed in tumor organoids. Immunofluorescence was used to assess the translocation of PAR and AIF to the cytoplasm or the nucleus and to study markers of DNA damage or apoptosis. Trapping of PARP1/2 or CHD1L onto chromatin was also assessed by in situ subcellular fractionation and immunofluorescence and validated by Western blot. We show that the inhibition of CHD1L's ATPase activity by OTI-611 is cytotoxic to triple-negative breast cancer tumor organoids and synergizes with PARPi and chemotherapy independently of the BRCA mutation status. The inhibition of the remodeling function blocks the phosphorylation of H2AX, traps CHD1L on chromatin, and leaves PAR chains on PARP1/2 open for hydrolysis. PAR hydrolysis traps PARP1/2 at DNA damage sites and mediates PAR translocation to the cytoplasm, release of AIF from the mitochondria, and induction of PARthanatos. The targeted inhibition of CHD1L's oncogenic function by OTI-611 signifies an innovative therapeutic strategy for breast cancer and other cancers. This approach capitalizes on CHD1L-mediated DNA repair and cell survival vulnerabilities, thereby creating synergy with standard-of-care therapies.
Collapse
Affiliation(s)
- Rita Sala
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO 80045, USA; (R.S.); (H.E.); (T.K.); (J.T.K.); (P.A.); (Q.Z.)
| | - Hector Esquer
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO 80045, USA; (R.S.); (H.E.); (T.K.); (J.T.K.); (P.A.); (Q.Z.)
- The CU Anschutz Center for Drug Discovery, Aurora, CO 80045, USA
- The University of Colorado Cancer Center, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Timothy Kellett
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO 80045, USA; (R.S.); (H.E.); (T.K.); (J.T.K.); (P.A.); (Q.Z.)
| | - Jeffrey T. Kearns
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO 80045, USA; (R.S.); (H.E.); (T.K.); (J.T.K.); (P.A.); (Q.Z.)
| | - Paul Awolade
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO 80045, USA; (R.S.); (H.E.); (T.K.); (J.T.K.); (P.A.); (Q.Z.)
| | - Qiong Zhou
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO 80045, USA; (R.S.); (H.E.); (T.K.); (J.T.K.); (P.A.); (Q.Z.)
- The CU Anschutz Center for Drug Discovery, Aurora, CO 80045, USA
- The University of Colorado Cancer Center, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Daniel V. LaBarbera
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, Aurora, CO 80045, USA; (R.S.); (H.E.); (T.K.); (J.T.K.); (P.A.); (Q.Z.)
- The CU Anschutz Center for Drug Discovery, Aurora, CO 80045, USA
- The University of Colorado Cancer Center, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
8
|
Gao Y, Wu Q, Wang G, Zhang S, Ma W, Shi X, Liu H, Wu L, Tian X, Li X, Ma X. Histomorphic analysis and expression of mRNA and miRNA in embryonic gonadal differentiation in Chinese soft-shelled turtle (Pelodiscus sinensis). Gene 2024; 893:147913. [PMID: 37866663 DOI: 10.1016/j.gene.2023.147913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/07/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
The Chinese soft-shelled turtle (Pelodiscus sinensis) is extensively cultured in Asia for its nutritional and medical value. Gonadal differentiation is fantastic in turtles, whereas morphologic, mRNA, and miRNA expressions were insufficient in the turtle. In this study, ovaries and testes histomorphology analysis of 14-23 stage embryos were performed, and mRNA and miRNA expression profiles were analyzed. Histomorphology analysis revealed that gonads were undifferentiated at embryonic stage 14. Ovarian morphological differentiation became evident from stage 15, which was characterized by the development of the cortical region and degeneration of the medullary region. Concurrently, testicular morphological differentiation was apparent from stage 15, marked by the development of the medullary region and degeneration of the cortical region. qRT-PCR results showed that Cyp19a1 and Foxl2 exhibited female-specific expression at stage 15 and the expression increased throughout most of the embryonic development. Dmrt1, Amh, and Sox9 displayed male-specific expression at stage 15 and tended to increase substantially at later developmental stages. The expression of miR-8356 and miR-3299 in ZZ gonads were significantly higher than that in ZW gonads at stage 15, 17 and 19, and they had the highest expression at stage 15. While the expression of miR-8085 and miR-7982 had the highest expression at stage 19. Furthermore, chromatin remodeler genes showed differential expression in female and male P. sinensis gonads. These results of master sex-differentiation genes and morphological characteristics would provide a reference for the research of sex differentiation and sex reversal in turtles. Additionally, the expression of chromatin remodeler genes indicated they might be involved in gonadal differentiation of P. sinensis.
Collapse
Affiliation(s)
- Yijie Gao
- College of Fisheries, Henan Normal University, Xinxiang 453007, China.
| | - Qisheng Wu
- Fisheries Research Institute of Fujian, Xiamen 361000, China.
| | - Guiyu Wang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China.
| | - Shufang Zhang
- College of Fisheries, Henan Normal University, Xinxiang 453007, China.
| | - Wenge Ma
- College of Fisheries, Henan Normal University, Xinxiang 453007, China.
| | - Xi Shi
- College of Fisheries, Henan Normal University, Xinxiang 453007, China.
| | - Huifen Liu
- College of Fisheries, Henan Normal University, Xinxiang 453007, China.
| | - Limin Wu
- College of Fisheries, Henan Normal University, Xinxiang 453007, China.
| | - Xue Tian
- College of Fisheries, Henan Normal University, Xinxiang 453007, China.
| | - Xuejun Li
- College of Fisheries, Henan Normal University, Xinxiang 453007, China.
| | - Xiao Ma
- College of Fisheries, Henan Normal University, Xinxiang 453007, China.
| |
Collapse
|
9
|
Clune S, Awolade P, Zhou Q, Esquer H, Matter B, Kearns JT, Kellett T, Akintayo DC, Kompella UB, LaBarbera DV. The validation of new CHD1L inhibitors as a therapeutic strategy for cancer. Biomed Pharmacother 2024; 170:116037. [PMID: 38128184 PMCID: PMC10792906 DOI: 10.1016/j.biopha.2023.116037] [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: 10/09/2023] [Revised: 12/04/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Chromodomain helicase DNA-binding protein 1 like (CHD1L) is an oncogene that promotes tumor progression, metastasis, and multidrug resistance. CHD1L expression is indicative of poor outcomes and low survival in cancer patients with various cancer types. Herein, we report a set of CHD1L inhibitors (CHD1Li) discovered from high-throughput screening and evaluated using enzyme inhibition, 3D tumor organoid cytotoxicity and mechanistic assays. The structurally distinct compounds 8-11 emerged as hits with promising bioactivity by targeting CHD1L. CHD1Li were further examined for their stability in human and mouse liver microsomes, which showed compounds 9 and 11 to be the most metabolically stable. Additionally, molecular modeling studies of CHD1Li with the target protein shed light on key pharmacophore features driving CHD1L binding. Taken together, these results expand the chemical space of CHD1Li as a potential targeted therapy for colorectal cancer and other cancers.
Collapse
Affiliation(s)
- Sophia Clune
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Paul Awolade
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA
| | - Qiong Zhou
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA; The CU Cancer Center, USA
| | - Hector Esquer
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA; The CU Cancer Center, USA
| | - Brock Matter
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jeffrey T Kearns
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Timothy Kellett
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Damilola Caleb Akintayo
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Uday B Kompella
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA; The CU Cancer Center, USA
| | - Daniel V LaBarbera
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The University of Colorado (CU) Anschutz Medical Campus, Aurora, CO 80045, USA; The CU Anschutz Center for Drug Discovery, USA; The CU Cancer Center, USA.
| |
Collapse
|
10
|
Soltan MA, Eldeen MA, Eid RA, Alyamani NM, Alqahtani LS, Albogami S, Jafri I, Park MN, Alsharif G, Fayad E, Mohamed G, Osman R, Kim B, Zaki MSA. A pan-cancer analysis reveals CHD1L as a prognostic and immunological biomarker in several human cancers. Front Mol Biosci 2023; 10:1017148. [PMID: 37033447 PMCID: PMC10076660 DOI: 10.3389/fmolb.2023.1017148] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction: Several recent studies pointed out that chromodomain-helicase-DNA-binding protein 1-like (CHD1L) is a putative oncogene in many human tumors. However, up to date, there is no pan-cancer analysis performed to study the different aspects of this gene expression and behavior in tumor tissues. Methods: Here, we applied several bioinformatics tools to make a comprehensive analysis for CHD1L. Firstly we assessed the expression of CHD1L in several types of human tumors and tried to correlate that with the stage and grade of the analyzed tumors. Following that, we performed a survival analysis to study the correlation between CHD1L upregulation in tumors and the clinical outcome. Additionally, we investigated the mutation forms, the correlation with several immune cell infiltration, and the potential molecular mechanisms of CHD1L in the tumor tissue. Result and discussion: The results demonstrated that CHD1L is a highly expressed gene across several types of tumors and that was correlated with a poor prognosis for most cancer patients. Moreover, it was found that CHD1L affects the tumor immune microenvironment by influencing the infiltration level of several immune cells. Collectively, the current study provides a comprehensive overview of the oncogenic roles of CHD1L where our results nominate CHD1L as a potential prognostic biomarker and target for antitumor therapy development.
Collapse
Affiliation(s)
- Mohamed A. Soltan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, Ismailia, Egypt
- *Correspondence: Mohamed A. Soltan, ; Bonglee Kim,
| | - Muhammad Alaa Eldeen
- Cell Biology, Histology and Genetics Division, Zoology Department, Faculty of Science, Zagazig University, Zagazig, Egypt
| | - Refaat A. Eid
- Pathology Department, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Najiah M. Alyamani
- Department of Biology, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Leena S. Alqahtani
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Sarah Albogami
- Department of Biotechnology, College of Sciences, Taif University, Taif, Saudi Arabia
| | - Ibrahim Jafri
- Department of Biotechnology, College of Sciences, Taif University, Taif, Saudi Arabia
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Ghadi Alsharif
- College of Clinical Laboratory Sciences, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Eman Fayad
- Department of Biotechnology, College of Sciences, Taif University, Taif, Saudi Arabia
| | - Gamal Mohamed
- Department of Human Anatomy, Jazan University, Jazan, Kingdom of Saydi Arabia
| | - Rihab Osman
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, South Korea
- *Correspondence: Mohamed A. Soltan, ; Bonglee Kim,
| | - Mohamed Samir A. Zaki
- Anatomy Department, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Department of Histology and Cell Biology, College of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
11
|
Prigaro BJ, Esquer H, Zhou Q, Pike LA, Awolade P, Lai XH, Abraham AD, Abbott JM, Matter B, Kompella UB, Messersmith WA, Gustafson DL, LaBarbera DV. Design, Synthesis, and Biological Evaluation of the First Inhibitors of Oncogenic CHD1L. J Med Chem 2022; 65:3943-3961. [PMID: 35192363 DOI: 10.1021/acs.jmedchem.1c01778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Chromodomain helicase DNA-binding protein 1 like (CHD1L) is an oncogene implicated in tumor progression, multidrug resistance, and metastasis in many types of cancer. In this article, we described the optimization of the first lead CHD1L inhibitors (CHD1Li) through drug design and medicinal chemistry. More than 30 CHD1Li were synthesized and evaluated using a variety of colorectal cancer (CRC) tumor organoid models and functional assays. The results led to the prioritization of six lead CHD1Li analogues with improved potency, antitumor activity, and drug-like properties including metabolic stability and in vivo pharmacokinetics. Furthermore, lead CHD1Li 6.11 proved to be an orally bioavailable antitumor agent, significantly reducing the tumor volume of CRC xenografts generated from isolated quasi mesenchymal cells (M-phenotype), which possess enhanced tumorigenic properties. In conclusion, we reported the optimization of first-in-class inhibitors of oncogenic CHD1L as a novel therapeutic strategy with potential for the treatment of cancer.
Collapse
Affiliation(s)
- Brett J Prigaro
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Hector Esquer
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Qiong Zhou
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Laura A Pike
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Paul Awolade
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Xin-He Lai
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Adedoyin D Abraham
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Joshua M Abbott
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Brock Matter
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Uday B Kompella
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States.,The University of Colorado (CU) Anschutz Medical Campus (AMC) Center for Drug Discovery, The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Wells A Messersmith
- The School of Medicine, Division of Medical Oncology, The CU Cancer Center, Aurora, Colorado 80045, United States.,The University of Colorado (CU) Anschutz Medical Campus (AMC) Center for Drug Discovery, The CU Cancer Center, Aurora, Colorado 80045, United States.,The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Daniel L Gustafson
- Flint Animal Cancer Center and Department of Clinical Sciences, School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado 80523, United States.,The University of Colorado (CU) Anschutz Medical Campus (AMC) Center for Drug Discovery, The CU Cancer Center, Aurora, Colorado 80045, United States.,The CU Cancer Center, Aurora, Colorado 80045, United States
| | - Daniel V LaBarbera
- The Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Pharmaceutical Sciences, The CU Cancer Center, Aurora, Colorado 80045, United States.,The University of Colorado (CU) Anschutz Medical Campus (AMC) Center for Drug Discovery, The CU Cancer Center, Aurora, Colorado 80045, United States.,The CU Cancer Center, Aurora, Colorado 80045, United States
| |
Collapse
|
12
|
Váncza L, Karászi K, Péterfia B, Turiák L, Dezső K, Sebestyén A, Reszegi A, Petővári G, Kiss A, Schaff Z, Baghy K, Kovalszky I. SPOCK1 Promotes the Development of Hepatocellular Carcinoma. Front Oncol 2022; 12:819883. [PMID: 35186754 PMCID: PMC8853618 DOI: 10.3389/fonc.2022.819883] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/10/2022] [Indexed: 12/15/2022] Open
Abstract
The extracellular matrix proteoglycan SPOCK1 is increasingly recognized as a contributor to the development and progression of cancers. Here, we study how SPOCK1, which is present in non-tumorous hepatocytes at low concentrations, promotes the development and progression of malignant hepatocellular tumors. Although SPOCK1 is an extracellular matrix proteoglycan, its concentration increases in the cytoplasm of hepatocytes starting with very low expression in the normal cells and then appearing in much higher quantities in cells of cirrhotic human liver and hepatocellular carcinoma. This observation is similar to that observed after diethylnitrosamine induction of mouse hepatocarcinogenesis. Furthermore, syndecan-1, the major proteoglycan of the liver, and SPOCK1 are in inverse correlation in the course of these events. In hepatoma cell lines, the cytoplasmic SPOCK1 colocalized with mitochondrial markers, such as MitoTracker and TOMM20, a characteristic protein of the outer membrane of the mitochondrion and could be detected in the cell nucleus. SPOCK1 downregulation of hepatoma cell lines by siRNA inhibited cell proliferation, upregulated p21 and p27, and interfered with pAkt and CDK4 expression. A tyrosine kinase array revealed that inhibition of SPOCK1 in the liver cancer cells altered MAPK signaling and downregulated several members of the Sarc family, all related to the aggressivity of the hepatoma cell lines. These studies support the idea that SPOCK1 enhancement in the liver is an active contributor to human and rodent hepatocarcinogenesis and cancer progression. However, its mitochondrial localization raises the possibility that it has a currently unidentified physiological function in normal hepatocytes.
Collapse
Affiliation(s)
- Lóránd Váncza
- 1 Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Katalin Karászi
- 1 Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Bálint Péterfia
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Lilla Turiák
- MS Proteomics Research Group, Research Centre for Natural Sciences, Eötvös Loránd Research Network, Budapest, Hungary
| | - Katalin Dezső
- 1 Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Anna Sebestyén
- 1 Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Andrea Reszegi
- 1 Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Gábor Petővári
- 1 Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - András Kiss
- 2 Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Schaff
- 2 Department of Pathology, Semmelweis University, Budapest, Hungary
| | - Kornélia Baghy
- 1 Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Ilona Kovalszky
- 1 Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
- *Correspondence: Ilona Kovalszky, ;
| |
Collapse
|