1
|
Malak MN, Arafa EA, Abdel-Fattah MM, Khalaf MM, Arab HH, Hamzawy MA. Targeting EGFR/PI3K/AKT/mTOR and Bax/Bcl-2/caspase3 pathways with ivermectin mediates its anticancer effects against urethane-induced non-small cell lung cancer in BALB/c mice. Tissue Cell 2025; 95:102873. [PMID: 40174264 DOI: 10.1016/j.tice.2025.102873] [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: 12/02/2024] [Revised: 02/12/2025] [Accepted: 03/15/2025] [Indexed: 04/04/2025]
Abstract
Lung cancer's mortality is among the highest compared to other cancers globally. However, a recent study has shown that ivermectin, an antiparasitic drug, may have a promising anticancer effect on lung cancer. The present study aimed to investigate the impact of ivermectin on EGFR.3/PI3K4/AKT5/mTOR6 signaling pathway in NSCLC.7 Mice were divided into four groups; (1) normal; (2) oral ivermectin alone (5 mg/kg) daily; (3) NSCLC was induced by urethane (1.5 g/kg, i.p.) at days one and sixty; (4) NSCLC group treated with ivermectin. The effect of ivermectin on macroscopic, microscopic, and lung index was assessed. The antitumor and antiproliferative effects of ivermectin were investigated by CYFRA 21-1 level and Ki-67, respectively. IHC determined the molecular expression of EGFR8, while phosphorylated PI3K, AKT, and mTOR were quantified by Western blotting assay. ELISA assay of active caspase 3, Bcl-29, and BAX10 was used to assess the apoptotic effect of ivermectin. Finally, VEGF11 lung content was measured. Findings showed that ivermectin improved macro and microscopic pathological changes. Ivermectin induced cytotoxic effect as indicated by CYFRA 21-1 suppression besides enhancing BAX/Bcl-2 ratio and active caspase 3. The immunoexpression of Ki-67 and EGFR declined. Ivermectin remarkably reduced p-PI3K, p-AKT, p-mTOR, and VEGF expressions. Overall, the study proposes ivermectin as a promising drug for lung cancer through its orchestral regulation of EGFR/PI3K/AKT/mTOR/VEGF signaling.
Collapse
Affiliation(s)
- Marina N Malak
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Elshaimaa A Arafa
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates; Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates.
| | - Maha M Abdel-Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Marwa M Khalaf
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Hany H Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Mohamed A Hamzawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Fayoum University, Fayoum 63514, Egypt
| |
Collapse
|
2
|
Jia LL, Wu CJ, Ye PW, Zhang Q, Liu H, Li TP, Hu XL. Terrestrosin D promotes autophagy and apoptosis of breast cancer cells through PSMD1-dependent activation of P53 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156883. [PMID: 40412055 DOI: 10.1016/j.phymed.2025.156883] [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: 02/06/2025] [Revised: 03/30/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND AND PURPOSE Breast cancer, particularly triple-negative breast cancer (TNBC), poses a significant threat to women's health. In tumor cells, autophagy and apoptosis are double-edged swords, playing complex roles in cancer progression and treatment. This study aimed to investigate whether Terrestrosin D (TED) exerts antitumor effects on TNBC by modulating autophagy and apoptosis, and to elucidate the underlying molecular mechanisms. METHODS The antiproliferative and pro-apoptotic effects of TED on TNBC cells were assessed using CCK-8, EdU assay, Live/Dead staining, and flow cytometry. Autophagy was monitored through immunofluorescence and confocal microscopy. RNA sequencing was performed to identify the pathways and molecular targets involved. The anti-TNBC effects of TED were further evaluated in vivo using tumor xenograft models. Western blotting was conducted to validate the relationship between PSMD1, P53, and TED-induced antitumor activity. RESULTS TED exhibited significant antitumor effects both in vitro and in vivo. Cellular phenotypic analyses revealed that TED promoted autophagy and apoptosis. Transcriptomic analyses indicated that TED stabilizes P53 expression and activates the P53 signaling pathway by inhibiting the function of PSMD1. CONCLUSION TED exhibits potent antitumor effects on TNBC by promoting autophagy and apoptosis. It achieves this through PSMD1 inhibition, stabilizing P53 expression, and activating the P53 pathway. Notably, this study is the first to demonstrate that TED directly targets PSMD1, a key proteasomal regulator, thereby unveiling a novel mechanism for P53 stabilization in TNBC. These findings provide new insights into the therapeutic modulation of the PSMD1 - P53 axis by natural compounds and support the development of TED as a multi-functional agent for aggressive breast cancers.
Collapse
Affiliation(s)
- Li-Ling Jia
- Department of Anaesthesia, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
| | - Cheng-Jie Wu
- Breast Center, Department of General Surgery, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, 510515, PR China
| | - Pei-Wen Ye
- Breast Center, Department of General Surgery, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, 510515, PR China
| | - Qian Zhang
- Breast Center, Department of General Surgery, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, 510515, PR China
| | - Hua Liu
- Department of Anaesthesia, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
| | - Tu-Ping Li
- Department of Anaesthesia, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, PR China
| | - Xiao-Lei Hu
- Breast Center, Department of General Surgery, Southern Medical University Nanfang Hospital, Guangzhou, Guangdong, 510515, PR China.
| |
Collapse
|
3
|
Chen W, Duan X, Zhu Z, Han Y, Li Y. Histone H1.4K75 acetylation promotes tumor growth and migration by regulating p53 and ERK1/2 pathway in non-small lung cancer. Biochem Biophys Res Commun 2025; 768:151880. [PMID: 40345010 DOI: 10.1016/j.bbrc.2025.151880] [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: 04/12/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
Histone H1.4 is a member of the linker histone H1 family, and its post-translational modifications (PTMs) are essential for its function. However, the role of H1.4 PTM in cancer development is not fully understood. Here, we report the discovery of a previously uncharacterized acetylation site at lysine 75 (K75) of H1.4 in non-small cell lung cancer (NSCLC). Point mutation of K75 with arginine (H1.4K75R) markedly suppressed cellular viability and migration in A549 and H1299 cells, and inhibit tumor growth in xenografts mouse models. Moreover, RNA-sequencing and Western Blot analyses revealed that H1.4K75 acetylation orchestrates dual regulatory effects, potentiating ERK1/2 signaling while repressing the p53 pathway. In summary, our studies show that H1.4K75 acetylation is essential for cell viability, migration and tumor growth in NSCLC, and may be a therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Wenqi Chen
- School of Life Sciences, Anhui University, Hefei, Anhui Province, China
| | - Xinyue Duan
- School of Life Sciences, Anhui University, Hefei, Anhui Province, China
| | - Zhebiao Zhu
- School of Life Sciences, Anhui University, Hefei, Anhui Province, China
| | - Yajing Han
- School of Life Sciences, Anhui University, Hefei, Anhui Province, China
| | - Yong Li
- School of Life Sciences, Anhui University, Hefei, Anhui Province, China.
| |
Collapse
|
4
|
Fernandez-Alarcon J, Cladera MA, Rodriguez-Camenforte N, Sitia G, Guerra-Rebollo M, Borros S, Fornaguera C. Regulation of mitochondrial apoptosis via siRNA-loaded metallo-alginate hydrogels: A localized and synergistic antitumor therapy. Biomaterials 2025; 318:123164. [PMID: 39923537 DOI: 10.1016/j.biomaterials.2025.123164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/13/2025] [Accepted: 02/01/2025] [Indexed: 02/11/2025]
Abstract
Preventing relapse after resection of a primary tumor continues to be an unmet clinical need. Development of adjuvant biomaterials with the capacity to kill residual cancer cells after tumor resection is of clinical importance. Here we developed a library of metallo-alginate hydrogels containing high concentrations of metallic ions such as Ca2+ in combination with Zn2+, Li+, or Mg2+ to disrupt Ca2+ homeostasis in the mitochondria of cancer cells by local hyperthermia. To synergistically kill tumor cells and suppress the growth of rechallenged tumors, we embedded oncogene-silencing nucleic acids (mTOR siRNA) loaded into polymerc nanoparticles (NPs) composed of poly (β-amino esters) in the metallo-alginate hydrogels, targeting cancer cells that activate multi-drug resistance pathways such PI3K/AKT/mTOR. Metabolomic studies showed alterations in the Warburg effect, mitochondrial transport, and the TCA cycle, confirming cancer cell damage. In vivo studies of this targeted therapy in mice demonstrated a sex-dependent effect. Male B16F10-tumor-bearing mice treated with the synergistic therapy showed restrained tumor growth. In contrast, no therapeutic effect was observed in female counterparts. Our results demonstrate that in situ-formed NP-loaded metallo-alginate hydrogels can modulate two distinct immune signaling networks that are relevant for enhancing cancer cell death. On the basis of our findings, this combination therapy emerges as a promising sex-dependent strategy for clinical translation.
Collapse
Affiliation(s)
- Jennifer Fernandez-Alarcon
- Grup d'Enginyeria de Materials (GEMAT), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), 08017, Barcelona, Spain
| | - Margalida Artigues Cladera
- Grup d'Electroquímica i Bioanàlisi (EQBA), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), 08017, Barcelona, Spain
| | - Natalia Rodriguez-Camenforte
- Grup d'Enginyeria de Materials (GEMAT), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), 08017, Barcelona, Spain
| | - Giovanni Sitia
- Experimental Hepatology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Marta Guerra-Rebollo
- Grup d'Enginyeria de Materials (GEMAT), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), 08017, Barcelona, Spain
| | - Salvador Borros
- Grup d'Enginyeria de Materials (GEMAT), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), 08017, Barcelona, Spain
| | - Cristina Fornaguera
- Grup d'Enginyeria de Materials (GEMAT), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), 08017, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig de Lluís Companys 23, 08010, Barcelona, Spain.
| |
Collapse
|
5
|
Nakada T, Koga M, Takeuchi H, Doi K, Sugiyama H, Sakurai H. PP2A adapter protein IER5 induces dephosphorylation and degradation of MDM2, thereby stabilizing p53. Cell Signal 2025; 131:111739. [PMID: 40081547 DOI: 10.1016/j.cellsig.2025.111739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/11/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
The tumor suppressor p53 activates transcription of the IER5 gene, which encodes an adapter protein of protein phosphatase PP2A. IER5 binds to both the B55 regulatory subunit of PP2A and PP2A's target proteins, facilitating PP2A/B55-catalyzed dephosphorylation of these proteins. Here, we show that IER5 functions as a positive regulator of p53 by inhibiting its ubiquitination, thereby increasing cellular p53 levels. Mechanistically, this effect of IER5 requires its nuclear localization and binding to both PP2A/B55 and the p53 ubiquitin E3 ligase MDM2. Importantly, IER5 fails to inhibit p53 ubiquitination in cells treated with the MDM2 inhibitor Nutlin-3. The IER5-PP2A/B55 complex dephosphorylates MDM2 at Ser166, leading to MDM2 ubiquitination and a reduction in nuclear MDM2. Altogether, our data provide evidence that IER5-PP2A/B55 regulates the nuclear balance between MDM2 and p53 via MDM2 dephosphorylation.
Collapse
Affiliation(s)
- Taisei Nakada
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Mayuko Koga
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Hiroto Takeuchi
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Kuriko Doi
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Haruka Sugiyama
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan
| | - Hiroshi Sakurai
- Division of Health Sciences, Graduate School of Medical Science, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa 920-0942, Japan.
| |
Collapse
|
6
|
Kweon J, Lee H, Park J, Hong T, An G, Song G, Lim W, Jeong W. Developmental and organ toxicity of fenpropimorph in zebrafish: Involvement of apoptosis and inflammation. Chem Biol Interact 2025; 415:111512. [PMID: 40239885 DOI: 10.1016/j.cbi.2025.111512] [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: 12/16/2024] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 04/18/2025]
Abstract
Pesticides are increasingly the focus as a prominent factor in environmental pollution. Fenpropimorph, a widely utilized morpholine fungicide, is a significant water pollutant. Because of its extensive usage, fenpropimorph is readily detected in diverse aquatic ecosystems. Despite its well-known toxicity to aquatic organisms, its toxicity to zebrafish development and accompanying mechanics remain unexplored. To assess fenpropimorph's toxicity and potential mechanism, we employed the zebrafish model, a representative tool in toxicological studies. Our results showed that exposure to fenpropimorph reduced embryonic viability during the early stages of development and reduced head and body size. Moreover, fenpropimorph triggered apoptosis, DNA fragmentation, and inflammation. Aberrations in the vascular network were observed in the fli1:eGFP transgenic zebrafish model. Additionally, neurotoxic impacts were further assessed using transgenic olig2:dsRed zebrafish, accompanied by a reduction of liver size and fluorescence intensity of fabp10a:dsRed zebrafish. mRNA expression analysis related to corresponding organ development further supported our data. Overall, our research suggests that fenpropimorph may cause aberrations in aquatic organisms.
Collapse
Affiliation(s)
- Junhun Kweon
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Hojun Lee
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Junho Park
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Taeyeon Hong
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Garam An
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| | - Whasun Lim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Wooyoung Jeong
- Department of Biomedical Sciences, Catholic Kwandong University, Gangneung, 25601, Republic of Korea.
| |
Collapse
|
7
|
Kojja V, Kumar D, Kalavagunta PK, Bhukya B, Tangutur AD, Nayak PK. 2-(Diarylalkyl)aminobenzothiazole derivatives induce autophagy and apoptotic death through SIRT inhibition and P53 activation In MCF7 breast cancer cells. Comput Biol Chem 2025; 116:108395. [PMID: 39987744 DOI: 10.1016/j.compbiolchem.2025.108395] [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: 01/02/2025] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
Sirtuins (SIRTs) are multifunctional proteins that exhibit a wide range of substrate preferences and cellular localizations. They are reliant on NAD+ and are essential for the regulation of several cellular functions. The SIRT proteins play important role towards tumor survival and resistance mechanisms in tumor cells. Therefore, molecules targeting SIRT proteins gained significant recognition in cancer research. In this work, we explored the anticancer property, potential and mode of action of 2-(diarylalkyl)aminobenzothiazole derivatives on MCF7 human breast cancer cells. Our studies established that 2-(diarylalkyl)aminobenzothiazole derivatives 1-((6-chlorobenzo[d]thiazol-2-ylamino)(3,4-dichlorophenyl)methyl)naphthalen-2-ol (7ab) and 1-((6-chlorobenzo[d]thiazol-2-ylamino)(4-bromophenyl)methyl)naphthalen-2-ol (7ba) treatment in a dose dependent manner drastically lowered the cell proliferation in MCF7 cells and the IC50 values of 7ab and 7ba was found to be 11.4 µM and 9.6 µM at 24 hr in these cells. Docking and molecular dynamic simulation studies further revealed that 7ab and 7ba show significant binding with SIRT1 protein. Consistently, treatment with 7ab and 7ba reduced the expression levels of SIRT1 protein while increasing acetylation of p53, a known SIRT protein target in MCF-7 cells. We observed that SIRT1inhibition was associated with activation of p53, an essential protein for apoptotic cell death, in MCF-7 cell lines. Furthermore, 7ab and 7ba treatment induced LC3-II expression and vacuole formation in the cytoplasm leading to autophagic cell death. Our findings together reveal the plausible cellular targets and specificity of these new small molecules as SIRT inhibitors, which increase p53 acetylation and suppress the proliferation of MCF-7 human breast cancer cells by triggering autophagic and apoptotic cell death.
Collapse
Affiliation(s)
- Venkateswarlu Kojja
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India
| | - Dinesh Kumar
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana State 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Praveen Kumar Kalavagunta
- Crop Protection Chemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana State 500007, India
| | - Bhima Bhukya
- Centre for Microbial and Fermentation Technology, Department of Microbiology, University College of Science, Osmania University, Hyderabad, Telangana State 500007, India
| | - Anjana Devi Tangutur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana State 500007, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India.
| | - Prasanta Kumar Nayak
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
8
|
Yin Y, Cao Y, Zhou Y, Xu Z, Luo P, Yang B, He Q, Yan H, Yang X. Downregulation of DDIT4 levels with borneol attenuates hepatotoxicity induced by gilteritinib. Biochem Pharmacol 2025; 236:116869. [PMID: 40081769 DOI: 10.1016/j.bcp.2025.116869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Gilteritinib, a multi-target kinase inhibitor, is currently used as standard therapy for acute myeloid leukemia. However, approximately half of the patients encounter liver-related adverse effects during the treatment with gilteritinib, which limiting its clinical applications. The underlying mechanisms of gilteritinib-induced hepatotoxicity and the development of strategies to prevent this toxicity are not well-reported. In our study, we utilized JC-1 dye, and MitoSOX to demonstrate that gilteritinib treatment leads to hepatocytes undergoing p53-mediated mitochondrial apoptosis. Furthermore, qRT-PCR analysis revealed that DNA damage-inducible transcript 4 (DDIT4), a downstream target of p53, was upregulated following gilteritinib administration and was identified as a key factor in gilteritinib-induced hepatotoxicity. After drug screening and western blot analysis, borneol, a bicyclic monoterpenoid, was found to decrease the protein level of DDIT4. This is the first compound found to downregulate DDIT4 levels and ameliorate hepatic injury caused by gilteritinib. Our findings suggest that high levels of DDIT4 are the primary driver behind gilteritinib-induced liver injury, and that borneol could potentially be a clinically safe and feasible therapeutic strategy by inhibiting DDIT4 levels.
Collapse
Affiliation(s)
- Yiming Yin
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, China
| | - Yashi Cao
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, China
| | - Yourong Zhou
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018 Zhejiang, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, China; School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018 Zhejiang, China; School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, China.
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018 Zhejiang, China; Nanhu Brain-computer Interface Institute, Hangzhou 311100 Zhejiang, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058 Zhejiang, China.
| |
Collapse
|
9
|
She F, Zhang T, Lee TH. Multifaceted role of zipper-interacting protein kinase beyond cell death: Implication of ZIPK dysregulation in neuronal and vascular injuries. Pharmacol Res 2025; 216:107793. [PMID: 40409521 DOI: 10.1016/j.phrs.2025.107793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/15/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
Zipper-interacting protein kinase (ZIPK) belongs to the death-associated protein kinase (DAPK) family and is a serine/threonine kinase. ZIPK is ubiquitously expressed in different types of tissues and cells. ZIPK is involved in many cellular functions, including cell death, smooth muscle contraction, transcriptional regulation, inflammatory signaling and the regulation of angiogenesis. The dysregulation of ZIPK has been shown to be involved in multiple diseases, including cancer, neurological diseases such as stroke, and cardiovascular diseases such as hypertension. The molecular mechanisms by which ZIPK inhibits the development of cancer have been well studied, but less is known about how ZIPK dysregulation is involved in vascular and neurological diseases. In this review, we summarize the current knowledge about the cellular processes in which ZIPK is involved and the pathological relevance of ZIPK dysregulation in diseases, with a focus on the role of ZIPK in vascular and neuronal functions. The molecular mechanisms by which ZIPK dysregulation contributes to cancer and vascular and neuronal diseases are discussed. We also review recent advances in the development of ZIPK modulators and their potential in treating vascular damage and neurological disorders. Multiple findings support that ZIPK has important functions in regulating vascular homeostasis and serves as a novel therapeutic target for alleviating neurological diseases.
Collapse
Affiliation(s)
- Fei She
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China
| | - Tao Zhang
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China.
| | - Tae Ho Lee
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, Institute of Basic Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
10
|
Rafai S, Cimbalo A, Manyes L. Evaluation of Dietary Bioactive Agents Against Aflatoxin B1 and Ochratoxin A-Induced Duodenal Toxicity in Rats. Foods 2025; 14:1793. [PMID: 40428572 PMCID: PMC12111047 DOI: 10.3390/foods14101793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2025] [Revised: 05/09/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Aflatoxin B1 (AFB1) and Ochratoxin A (OTA) are two of the most potent mycotoxins, recognized for their severe toxicity. In recent years, the consumption of bioactive substances has proven to be a valuable ally in combating their harmful effects on human health. For this purpose, this study evaluates the protective effects of fermented whey (FW) and pumpkin (P), as functional ingredients in bread, on duodenum tissue against sub-chronic toxicity induced by AFB1 and OTA. Nine groups of male and female Wistar rats (n = 5 per sex/group) were exposed to different combinations of AFB1, OTA, FW, and P for 28 days. The gene expression of apoptotic and antioxidant markers, including p53, Bax, Hmox1, NF-κB, and occludin, was measured by quantitative real-time PCR (RT-qPCR). AFB1 + OTA exposure led to an increased expression of p53 and NF-κB, with the downregulation of Bax and Hmox1. Occludin expression, which supports tight junction integrity, remained largely unaffected. Supplementation with FW and FW + P modulated gene expression favorably, offering protection against AFB1 and OTA toxicity. These bioactive components effectively mitigated oxidative stress and apoptosis in duodenal tissue. Notably, the results indicate that the protective effects of FW and P are not sex-dependent. These findings highlight the potential of FW and P as functional ingredients in combating the toxic effects of AFB1 and OTA in vivo.
Collapse
Affiliation(s)
| | - Alessandra Cimbalo
- Biotech Agrifood, Faculty of Pharmacy and Food Sciences, Universitat de València, Avda, Vicent Andrés Estellés s/n, 46100 Burjassot, Spain; (S.R.); (L.M.)
| | | |
Collapse
|
11
|
Rossi A, Mazzara S, Salemi D, Zanetti S, Sapienza MR, Orecchioni S, Talarico G, Falvo P, Davini A, Ceccarelli C, Motta G, Melle F, Tabanelli V, Agostinelli C, Trerè D, Penzo M, Corsini C, Baiardi E, Calleri A, Vitolo U, Bertolini F, Zinzani PL, Chiarle R, Tarella C, Pileri S, Derenzini E. Downregulation of rRNA synthesis by BCL-2 induces chemoresistance in diffuse large B cell lymphoma. iScience 2025; 28:112333. [PMID: 40276769 PMCID: PMC12020883 DOI: 10.1016/j.isci.2025.112333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/25/2025] [Accepted: 03/28/2025] [Indexed: 04/26/2025] Open
Abstract
Overexpression of the antiapoptotic oncogene BCL-2 predicts poor prognosis in diffuse large B cell lymphoma (DLBCL) treated with anthracycline-based chemoimmunotherapy. Anthracyclines exert antitumor effects by multiple mechanisms including inhibition of ribosome biogenesis (RiBi) through rRNA synthesis blockade. RiBi inhibitors induce p53 stabilization through the ribosomal proteins-MDM2-p53 pathway, with stabilized p53 levels depending on baseline rRNA synthesis rate. We found that the BH3-mimetic venetoclax could not fully reverse BCL-2-mediated resistance to RiBi inhibitors in DLBCL cells. BCL-2 overexpression was associated with decreased baseline rRNA synthesis rate, attenuating p53 stabilization by RiBi inhibitors. Drugs stabilizing p53 irrespective of RiBi inhibition reversed BCL-2-induced resistance in vitro and in vivo, restoring p53 activation and apoptosis. A small nucleolar size, indicative of low baseline rRNA synthesis, correlated with high BCL-2 levels and poor outcomes in DLBCL patients. These findings uncover alternative BCL-2-dependent chemoresistance mechanisms, providing a rationale for specific combination strategies in BCL-2 positive lymphomas.
Collapse
Affiliation(s)
- Alessandra Rossi
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Saveria Mazzara
- Division of Diagnostic Haematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Computing Sciences and Bocconi Institute for Data Science and Analytics (BIDSA), Bocconi University, Milan, Italy
- AI and Systems Biology, IFOM, ETS, Milan, Italy
| | - Dorotea Salemi
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Simone Zanetti
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Maria Rosaria Sapienza
- Division of Diagnostic Haematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Stefania Orecchioni
- Laboratory of Hematology-Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Giovanna Talarico
- Laboratory of Hematology-Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Paolo Falvo
- Laboratory of Hematology-Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Alessandro Davini
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Claudio Ceccarelli
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giovanna Motta
- Division of Diagnostic Haematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria of Bologna, Bologna, Italy
| | - Federica Melle
- Division of Diagnostic Haematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Valentina Tabanelli
- Division of Diagnostic Haematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Claudio Agostinelli
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
- Haematopathology Unit, IRCCS Azienda Ospedaliero-Universitaria of Bologna, Bologna, Italy
| | - Davide Trerè
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
- Department Program in Laboratory Medicine, IRCCS Azienda Ospedaliero-Universitaria of Bologna, Bologna, Italy
| | - Marianna Penzo
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
- Center for Applied Biomedical Research (CRBA), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Chiara Corsini
- Laboratory of Hematology-Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Elena Baiardi
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Angelica Calleri
- Division of Diagnostic Haematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Umberto Vitolo
- Multidisciplinary Oncology Outpatient Clinic, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Francesco Bertolini
- Laboratory of Hematology-Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Pier Luigi Zinzani
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
- Seràgnoli Hematology Institute, IRCCS AOU (Azienda Ospedaliero-Universitaria) of Bologna, Bologna, Italy
| | - Roberto Chiarle
- Division of Diagnostic Haematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Boston Children’s Hospital, Department of Pathology, Harvard Medical School, Boston, MA, USA
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Corrado Tarella
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Stefano Pileri
- Division of Diagnostic Haematopathology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Enrico Derenzini
- Oncohematology Division, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| |
Collapse
|
12
|
Xiao T, Zhang P, Guo S, Su B, Chen Y, Zhao M, Yuan J, Si S, Zheng R, Li K, Chen M. Peunipyrone A, an Unexpected Highly Oxygenated γ-Pyrone with a 6/6/6/6/6 Pentacyclic Ring System from a Sponge-Derived Fungus Aspergillus peuniceus. Org Lett 2025; 27:4958-4963. [PMID: 40329452 DOI: 10.1021/acs.orglett.5c01274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
A highly oxygenated γ-pyrone with an unprecedented carbon skeleton, peunipyrone A (1), was isolated from the sponge-derived fungus Aspergillus peuniceus. Compound 1 features a unique fused 6/6/6/6/6 pentacyclic ring system. The chemical structure of compound 1 was elucidated by comprehensive spectroscopic techniques and single-crystal X-ray diffraction. The plausible biosynthetic pathway of compound 1 was proposed. Compound 1 promoted leukemic cell apoptosis by increasing p53 expression.
Collapse
Affiliation(s)
- Tongmei Xiao
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development, Institute of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Peitao Zhang
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development, Institute of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Shuyue Guo
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 3K3, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Ontario M5G 1X5, Canada
| | - Bingjie Su
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development, Institute of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Yuchuan Chen
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development, Institute of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Mingxuan Zhao
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development, Institute of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Jian Yuan
- State Key Laboratory of Cardiology and Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, People's Republic of China
| | - Shuyi Si
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development, Institute of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Ruifang Zheng
- Key Laboratory for Uighur Medicine, Institute of Materia Medica of Xinjiang Uygur Autonomous Region, Urumqi 830004, People's Republic of China
| | - Ke Li
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development, Institute of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Minghua Chen
- Beijing Key Laboratory of Technology and Application for Anti-Infective New Drugs Research and Development, Institute of Medicinal Biotechnology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
- Key Laboratory for Uighur Medicine, Institute of Materia Medica of Xinjiang Uygur Autonomous Region, Urumqi 830004, People's Republic of China
| |
Collapse
|
13
|
Sun Z, Wu R, Liang X, Shi T, Zhang Y, Pan Z, Zhang W, Luan X. MLCK inhibition induces synthetic lethality in MYC-driven cancer. Cancer Lett 2025; 625:217803. [PMID: 40381685 DOI: 10.1016/j.canlet.2025.217803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 05/08/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
The dysregulation of MYC is widely implicated in human cancers, yet MYC remains an 'undruggable' target. Here, we performed a CRISPR-based loss-of-function screen focusing on kinases, most of which are 'druggable,' to identify genes essential for MYChigh but not MYClow cells. Using an isogenic pair of nonmalignant cells with and without ectopic MYC expression, we uncovered novel MYC synthetic lethal (MYC-SL) interactions, including Myosin Light-Chain Kinase (MLCK) as the most potent MYC-SL target. Inhibition of MLCK induced MYC-dependent cell death, significantly suppressing tumor growth in MYC-driven xenografts, the ApcMin/+ mouse model of colon cancer, and the MYC-transgenic hepatocellular carcinoma (HCC) model, without apparent toxicity. This cell death is attributed to selective DNA damage and p53-mediated apoptosis. Mechanistically, MYC activation promotes nuclear accumulation of myosin II at stalled replication forks, where it resolves replication stress and supports survival. MLCK inhibition disrupts myosin II activity, leading to unresolved replication stress, DNA damage, and activation of the p53-mediated apoptosis pathway. Our findings suggest that targeting MLCK offers a promising therapeutic strategy for MYC-driven cancers.
Collapse
Affiliation(s)
- Zhe Sun
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Rui Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaohui Liang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tiezhu Shi
- Precise Genome Engineering Centre, School of Life Sciences, Guangzhou University, Guangzhou, 510006, China
| | - Yuan Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Guangdong Pharmaceutical University, Guangdong, 510006, China
| | - Zelin Pan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Weidong Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.
| | - Xin Luan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
14
|
Do SD, Rhee JS. Exposure to short-chain chlorinated paraffins induces mortality in marine medaka larvae through neurotoxicity and oxidative stress. JOURNAL OF HAZARDOUS MATERIALS 2025; 494:138531. [PMID: 40354700 DOI: 10.1016/j.jhazmat.2025.138531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Despite the strict regulation of short-chain chlorinated paraffins (SCCPs), they continue to be detected in marine environments and organisms worldwide. However, their toxicity to marine fish has been scarcely studied. In this study, the effect of different concentrations of SCCPs (0-1000 μg L⁻1) on the early life stages of the marine medaka Oryzias melastigma was evaluated. The impacts of SCCPs on the embryonic stage of O. melastigma were considered negligible, while significant growth retardation was observed during the larval stages after 13 days of exposure, with the median lethal concentration (LC50) determined as 227 μg L⁻1. Exposure to SCCPs for 4 days resulted in melanosome dispersion, immobilization, disruption of feeding activity, and an increase in acetylcholinesterase activity, suggesting that SCCPs induce neurotoxicity in the cholinergic system, leading to mortality through starvation. Dose-dependent DNA damage, fluctuations in mitochondrial respiration, and increases in intracellular reactive oxygen species content and apoptosis were observed after 4 days of exposure, indicating strong induction of oxidative stress. In addition, potential endocrine disruption was observed, as SCCPs significantly decreased cortisol content and modulated the mRNA expressions of genes involved in the hypothalamic-pituitary-interrenal axis. Taken together, exposure to SCCPs resulted in mortality and growth retardation, primarily through neurotoxicity and oxidative stress in marine medaka larvae.
Collapse
Affiliation(s)
- Seong Duk Do
- Department of Marine Science, College of Natural Sciences, Incheon National University, Incheon 22012, Republic of Korea
| | - Jae-Sung Rhee
- Department of Marine Science, College of Natural Sciences, Incheon National University, Incheon 22012, Republic of Korea; Research Institute of Basic Sciences, Core Research Institute, Incheon National University, Incheon 22012, Republic of Korea; Yellow Sea Research Institute, Incheon 22012, Republic of Korea.
| |
Collapse
|
15
|
Chatzilygeroudi T, Karantanos T, Pappa V. Unraveling Venetoclax Resistance: Navigating the Future of HMA/Venetoclax-Refractory AML in the Molecular Era. Cancers (Basel) 2025; 17:1586. [PMID: 40361510 PMCID: PMC12071220 DOI: 10.3390/cancers17091586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
Acute myeloid leukemia (AML) has traditionally been linked to a poor prognosis, particularly in older patients who are ineligible for intensive chemotherapy. The advent of Venetoclax, a powerful oral BH3 mimetic targeting anti-apoptotic protein BCL2, has significantly advanced AML treatment. Its combination with the hypomethylating agent azacitidine (AZA/VEN) has become a standard treatment for this group of AML patients, demonstrating a 65% overall response rate and a median overall survival of 14.7 months, compared to 22% and 8 months with azacitidine monotherapy, respectively. However, resistance and relapses remain common, representing a significant clinical challenge. Recent studies have identified molecular alterations, such as mutations in FLT3-ITD, NRAS/KRAS, TP53, and BAX, as major drivers of resistance. Additionally, other factors, including metabolic changes, anti-apoptotic protein expression, and monocytic or erythroid/megakaryocytic differentiation status, contribute to treatment failure. Clinical trials are exploring strategies to overcome venetoclax resistance, including doublet or triplet therapies targeting IDH and FLT3 mutations; novel epigenetic approaches; menin, XPO1, and MDM2 inhibitors; along with immunotherapies like monoclonal antibodies and antibody-drug conjugates. A deeper understanding of the molecular mechanisms of resistance through single-cell analysis will be crucial for developing future therapeutic strategies.
Collapse
Affiliation(s)
- Theodora Chatzilygeroudi
- Second Department of Internal Medicine and Research Unit, Hematology Unit, National and Kapodistrian University of Athens School of Medicine, Attikon University Hospital, 12462 Athens, Greece;
| | - Theodoros Karantanos
- Division of Hematologic Malignancies and Bone Marrow Transplantation, Department of Medical Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Vasiliki Pappa
- Second Department of Internal Medicine and Research Unit, Hematology Unit, National and Kapodistrian University of Athens School of Medicine, Attikon University Hospital, 12462 Athens, Greece;
| |
Collapse
|
16
|
Zhong M, Pan G, Tan J, Yao J, Liu Y, Huang J, Jiang Y, Zhu D, Zhao J, Xu B, Zha J. Venetoclax confers synthetic lethality to chidamide in preclinical models with transformed follicular lymphoma. Clin Epigenetics 2025; 17:74. [PMID: 40320542 PMCID: PMC12051286 DOI: 10.1186/s13148-025-01878-0] [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: 12/11/2024] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Transformed follicular lymphoma (t-FL) is an aggressive and heterogeneous hematological malignancy with limited treatment success; the development of novel therapeutic approaches is urgently needed for patients with t-FL. Here, we conducted high-throughput screening (HTS) and in vitro experiments using t-FL cell lines and primary samples to assess the synergistic effects of the histone deacetylase inhibitor chidamide and the BCL-2 inhibitor venetoclax. In vivo efficacy was further tested in xenograft models. The combination of venetoclax and chidamide significantly inhibited cell proliferation, induced apoptosis, and arrested the cell cycle in the G0/G1 phase across multiple t-FL cell lines. Furthermore, the combined therapy effectively reduced tumor burden, extended overall survival in xenograft models, and synergistically targeted patient samples, while sparing normal PBMCs. Mechanistically, this combination disrupted mitochondrial membrane potential and modulated the Wnt signaling pathway, as evidenced by decreased protein expression levels of Wnt3a, Wnt5a/b, β-catenin, and phosphorylated GSK3β. Concurrently, the combined regimen enhanced their respective anticancer effects by inhibiting the key genes HDAC10 and BCL-xL. Taken together, venetoclax combined with chidamide presents a potent anticancer strategy in preclinical models of t-FL and merits further exploration in clinical trials to validate its effectiveness and safety for treating t-FL.
Collapse
Affiliation(s)
- Mengya Zhong
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Department of Radiology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Guangchao Pan
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Jinshui Tan
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Department of Gastrointestinal Surgery, School of Medicine, Zhongshan Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Jingwei Yao
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
| | - Yating Liu
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
| | - Jiewen Huang
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Yuelong Jiang
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
| | - Depeng Zhu
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China
| | - Jintao Zhao
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China.
| | - Bing Xu
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China.
| | - Jie Zha
- Department of Hematology, School of Medicine, The First Affiliated Hospital of Xiamen University and Institute of Hematology, Xiamen University, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, No. 55, Shizhen Hai Road, Xiamen, 361003, People's Republic of China.
| |
Collapse
|
17
|
Zhu Y, Chen Z, Niu K, Li M, Deng Y, Zhang J, Wei D, Wang J, Zhao Y. USP33 Regulates DNA Damage Response and Carcinogenesis Through Deubiquitylating and Stabilising p53. Cell Prolif 2025; 58:e13793. [PMID: 39694539 PMCID: PMC12099211 DOI: 10.1111/cpr.13793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
The de-ubiquitinase USP33 has been shown to possess either tumour-promoting or inhibitory effect on human cancer cells. However, all these findings are mainly based on in vitro cell culture models, and the in vivo evidence, which is more plausible to digest the functional role of USP33 in carcinogenic process, is still lacking. Here, we demonstrate that USP33 modulates DNA damage responses including cell cycle arrest and apoptosis induction through associating with p53. It directly interacts with p53 to mediate its de-ubiquitination and further stabilisation under DNA damage condition. Depletion of USP33 induces an enhanced level of p53 ubiquitination, which de-stabilises p53 protein leading to impaired DNA damage responses. Furthermore, USP33 silencing shows either promoted or inhibited effect on cell proliferation in human cancer cells with p53 WT and mutant background, respectively. Consistently, mice with hepatocyte-specific USP33 knockout are more sensitive to nitrosodiethylamine (DEN)-induced hepatocarcinogenesis compared to wild type mice. Thus, our in vitro and in vivo evidences illustrate that USP33 possesses anti-tumour activity via regulating p53 stability and activity.
Collapse
Affiliation(s)
- Yuqi Zhu
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Zixiang Chen
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
| | - Kaifeng Niu
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
| | - Mengge Li
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yuchun Deng
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Ji Zhang
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Di Wei
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
| | - Jiaqi Wang
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - YongLiang Zhao
- China National Center for BioinformationBeijingChina
- Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| |
Collapse
|
18
|
Qiu H, Zhang C, Ma X, Li Y. Molecular insights and treatment innovations: Advancing outcomes in acute myeloid leukemia with myelodysplasia‑related changes (Review). Oncol Rep 2025; 53:54. [PMID: 40116086 DOI: 10.3892/or.2025.8887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/10/2025] [Indexed: 03/23/2025] Open
Abstract
Acute myeloid leukemia, myelodysplasia‑related (AML‑MR), a challenging and aggressive subtype of AML, is characterized by unique genetic abnormalities and molecular features, which contribute to its poor prognosis compared with other AML subtypes. The present review summarizes the current understanding of AML‑MR pathogenesis, highlighting notable advancements in genetic and cytogenetic insights. Critical mutations, such as those in the tumor antigen p53 and additional sex combs like 1 genes, and their role in disease progression and resistance to treatment, are explored. The review further investigates how clonal evolution and cellular microenvironment alterations drive AML‑MR transformation and impact patient outcomes. Despite the poor outlook typically associated with AML‑MR, developments in treatment approaches offer hope. The present review considers the efficacy of novel therapeutic agents, including CPX‑351, hypomethylating agents and targeted molecular therapies. Additionally, innovations in immunotherapy and allogeneic hematopoietic stem cell transplantation are discussed as promising avenues to improve patient survival rates. The challenges of treating AML‑MR, particularly in elderly and pretreated patients, underline the necessity for individualized treatment strategies that consider both the biological complexity of the disease and the overall health profile of the patient. The present review focuses on the mechanisms of AML‑MR transformation, highlighting factors that may offer a crucial theoretical foundation and pave the way for future applications in precision medicine. Future research directions include exploring novel targeted therapies and combination regimens to mitigate the transformation risks and enhance the quality of life of patients with AML‑MR.
Collapse
Affiliation(s)
- Hong Qiu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Chaowei Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Xiaochen Ma
- Department of Public Health, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Ying Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
19
|
Vosselman T, Sahin C, Lane DP, Arsenian Henriksson M, Landreh M, Lama D. Conformational modulation of intrinsically disordered transactivation domains for cancer therapy. PNAS NEXUS 2025; 4:pgaf152. [PMID: 40406608 PMCID: PMC12096364 DOI: 10.1093/pnasnexus/pgaf152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 04/24/2025] [Indexed: 05/26/2025]
Abstract
Intrinsically disordered proteins are implicated in many diseases, but their overrepresentation among transcription factors, whose deregulation can cause disproportionate expression of oncogenes, suggests an important role in cancer. Targeting disordered transcription factors for therapy is considered challenging, as they undergo dynamic transitions and exist as an ensemble of interconverting states. This enables them to interact with multiple downstream partners, often through their transactivation domains (TADs) by the mechanisms of conformational selection, folding-upon-binding, or formation of "fuzzy" complexes. The TAD interfaces, despite falling outside of what is considered "classical" binding pockets, can be conformationally modulated to interfere with their target recruitment and hence represent potentially druggable sites. Here, we discuss the structure-activity relationship of TADs from p53, c-MYC, and the androgen receptor, and the progresses made in modulating their interactions with small molecules. These recent advances highlight the potential of targeting these so far "undruggable" proteins for cancer therapy.
Collapse
Affiliation(s)
- Thibault Vosselman
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-171 65 Stockholm, Sweden
| | - Cagla Sahin
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-171 65 Stockholm, Sweden
- Structural Biology and NMR Laboratory and the Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Ole Maaløes Vej 5, 2200 Copenhagen, Denmark
| | - David P Lane
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-171 65 Stockholm, Sweden
| | - Marie Arsenian Henriksson
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-171 65 Stockholm, Sweden
| | - Michael Landreh
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-171 65 Stockholm, Sweden
- Department of Cell and Molecular Biology, Uppsala University, Husargatan 3, SE-751 24 Uppsala, Sweden
| | - Dilraj Lama
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Biomedicum, Solnavägen 9, SE-171 65 Stockholm, Sweden
| |
Collapse
|
20
|
Choi H, Lee JY, Yoo H, Jeon K. Identification and Verification of Necroptosis-Related Genes in Patients With Sepsis by Bioinformatic Analysis and Molecular Experiments. J Cell Mol Med 2025; 29:e70582. [PMID: 40318009 PMCID: PMC12049152 DOI: 10.1111/jcmm.70582] [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: 09/16/2024] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025] Open
Abstract
Although necroptosis is an emerging mechanism of multiple organ dysfunction in sepsis, data on the mechanistic link between necroptosis and sepsis are scarce. Bioinformatic analysis was performed to compare the gene profiles between the sepsis (n = 133) and healthy control (n = 12) groups and identify necroptosis-related differentially expressed genes (DEGs). The identified necroptosis-related DEGs were verified by three-step molecular experiments: (1) quantitative real-time PCR and enzyme-linked immunosorbent assay; (2) cell culture, transfection and Western blotting; and (3) cytokine array with apoptosis inhibition. Additionally, receiver-operating characteristic curve analyses were performed to evaluate the performance of the corresponding proteins to the necroptosis-related DEGs in diagnosing sepsis and in predicting in-hospital mortality of patients with sepsis. Eight necroptosis-related DEGs, including five upregulated (PYGL, TNF, CYLD, FADD and TLR3) and three downregulated (TP53, FASLG and NLRP6) DEGs, were identified. Moreover, the levels of the corresponding proteins to necroptosis-related DEGs showed excellent or considerable accuracy in diagnosing sepsis and in predicting the mortality of sepsis patients. In cell culture media transfected with plasma from the sepsis and control groups, Western blotting revealed that the levels of the corresponding proteins were increased in the upregulated DEGs and decreased in the downregulated DEGs. The cytokine array revealed cytokines in cell culture media transfected with plasma from patients with sepsis while preventing apoptosis by inhibiting the caspase-8 activity, wherein the transfected cells potentially underwent necroptosis. Eight necroptosis-related DEGs were identified in patients with sepsis by bioinformatic analysis and verified by molecular experiments, implying that necroptosis may be a key mechanism of sepsis.
Collapse
Affiliation(s)
- Hayoung Choi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal MedicineHallym University Kangnam Sacred Heart Hospital, Hallym University College of MedicineSeoulRepublic of Korea
| | - Jin Young Lee
- Division of Pulmonary and Critical Care Medicine, Department of MedicineSamsung Medical Center, Sungkyunkwan University School of MedicineSeoulRepublic of Korea
| | - Hongseok Yoo
- Division of Pulmonary and Critical Care Medicine, Department of MedicineSamsung Medical Center, Sungkyunkwan University School of MedicineSeoulRepublic of Korea
| | - Kyeongman Jeon
- Division of Pulmonary and Critical Care Medicine, Department of MedicineSamsung Medical Center, Sungkyunkwan University School of MedicineSeoulRepublic of Korea
- Department of Health Science TechnologySAIHST, Sungkyunkwan UniversitySeoulRepublic of Korea
| |
Collapse
|
21
|
Song L, He X, Duan Y, Chi Y, Li R, Li C, Liu Y, Yang M, Wei J, Zhao Y, Xu Q. Identification of druggable genetic targets for prostate cancer risk based on mendelian randomization and single-cell RNA sequencing. Int Urol Nephrol 2025:10.1007/s11255-025-04525-y. [PMID: 40304996 DOI: 10.1007/s11255-025-04525-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 04/12/2025] [Indexed: 05/02/2025]
Abstract
PURPOSE This study aimed to identify genetic targets linked to prostate cancer risk using advanced genetic analysis techniques. OBJECTIVE The goal was to conduct a comprehensive analysis using Mendelian Randomization (MR), colocalization, and single-cell RNA sequencing to identify druggable genes as potential therapeutic targets or diagnostic markers. METHODS The study involved selecting 2608 druggable genes by intersecting expression Quantitative Trait Loci (eQTLs) with druggable genome databases. MR analysis using prostate cancer GWAS data identified genes with causal associations to prostate cancer risk. Colocalization analysis confirmed shared genetic variants influencing both the exposure and outcome. Single-cell RNA sequencing assessed gene expression in prostate tumor cell types, while a phenome-wide association study (PheWAS) evaluated potential side effects. RESULTS MR analysis identified 58 genes associated with prostate cancer risk, with 12 validated by colocalization analysis. Five genes (BAK1, ATP1B2, PEMT, TPM3, ZDHHC7) demonstrated strong colocalization, indicating potential as drug targets. Single-cell RNA sequencing revealed their enrichment in prostate tumor T cells and macrophages. PheWAS suggested minimal side effects for most, except BAK1, which was linked to increased platelet counts. CONCLUSION This study identified several genetic targets associated with prostate cancer risk, highlighting the potential for targeted therapy. By integrating Mendelian randomization analysis, colocalization analysis, and single-cell RNA sequencing, the accuracy of target validation was improved, which may provide new directions for targeted therapy in prostate cancer.
Collapse
Affiliation(s)
- Liantai Song
- Chengde Medical University, Chengde, 067000, China
| | - Xinyang He
- Chengde Medical University, Chengde, 067000, China
| | - Yibing Duan
- Chengde Medical University, Chengde, 067000, China
| | - Yifan Chi
- Chengde Medical University, Chengde, 067000, China
| | - Reng Li
- Chengde Medical University, Chengde, 067000, China
| | - Cancan Li
- Chengde Medical University, Chengde, 067000, China
| | - Yutian Liu
- Chengde Medical University, Chengde, 067000, China
| | - Mengxin Yang
- Chengde Medical University, Chengde, 067000, China
| | - Jiameng Wei
- Chengde Medical University, Chengde, 067000, China
| | - Yujia Zhao
- Chengde Medical University, Chengde, 067000, China
| | - Qian Xu
- Chengde Medical University, Chengde, 067000, China.
| |
Collapse
|
22
|
Mfotie Njoya E, Tabakam GT, Chukwuma CI, Makhafola TJ. Buxus natalensis (Oliv.) Hutch (Buxaceae) Exhibits Its Anticancer Potential by Stimulating ROS Production and Caspase-p53-BCL-2-Dependent Apoptosis in Hepatocellular Carcinoma and Prostate Cancer Cell Lines. Int J Mol Sci 2025; 26:4173. [PMID: 40362409 PMCID: PMC12071716 DOI: 10.3390/ijms26094173] [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: 03/31/2025] [Revised: 04/20/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Buxus natalensis is recognized as a rich source of triterpenoidal alkaloids that are known to be effective in fighting different cancer types. Nevertheless, to date, no anticancer potential of B. natalensis extract has been yet described. Here, we investigated the antiproliferative activity of different B. natalensis leaf extracts on eight cancer cell lines (MCF-7, 4T1, Caco-2, HeLa, A549, HepG2, DU145, and LNCaP). Chang liver cell line derived from normal liver tissue, was used as control. B. natalensis hydroethanolic leaf extract (BNHLE) was found to exert significant cytotoxic effect against cancerous cell lines, with the highest efficacy being observed on LNCaP and HepG2 with IC50 values of 47.39 and 78.01 µg/mL, respectively. Interestingly, BNHLE was less cytotoxic towards Chang liver cells with an IC50 value of 334.10 µg/mL, yielding selectivity index (SI) values of 6.96 and 4.22 against LNCaP and HepG2 cells, respectively. The study of mechanism of action revealed that BNHLE exerted its antiproliferative effect by inducing ROS production and caspase -3/-7, and -9 activities in LNCaP and HepG2 cells. Moreover, it was found that BNHLE activated apoptosis in both cancerous cell lines by enhancing the expression levels of p53, while suppressing the expression of NF-κB-p65 and BCL-2 protein levels in a dose-dependent manner. The phytochemical analysis of BNHLE showed the presence of flavonoids (24.45 mgQE/g extract) and phenolics (84.64 mgGAE/g extract), and its LC-MS profiling identified several compounds including robinin and rutin, which are known for their cytotoxic effect against different cancer cell lines, such as hepatocellular carcinoma and prostate cancer cell lines. Several compounds are still unknown from B. natalensis, but the data obtained so far justify the use of B. natalensis as a potential source of bioactive compounds against hepatocellular and prostate cancers.
Collapse
Affiliation(s)
- Emmanuel Mfotie Njoya
- Centre for Quality of Health and Living, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9301, Free State, South Africa; (G.T.T.); (C.I.C.)
| | | | | | - Tshepiso J. Makhafola
- Centre for Quality of Health and Living, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9301, Free State, South Africa; (G.T.T.); (C.I.C.)
| |
Collapse
|
23
|
Farahat YA, El-Sayed NM, Hazem RM, Mehanna ET, Radwan A. Sinapic Acid Ameliorates Cadmium-Induced Hepatotoxicity: Modulation of Oxidative Stress, Inflammation, and Apoptosis. Biomedicines 2025; 13:1065. [PMID: 40426893 PMCID: PMC12109072 DOI: 10.3390/biomedicines13051065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Cadmium (Cd) is a harmful metal commonly used in industry. Numerous clinical diseases, including osteomalacia, testicular damage, renal and hepatic failure, and pulmonary edema, are associated with Cd exposure. The current study evaluated the protective effect of Sinapic acid (SA) against Cd-induced hepatotoxicity by investigating different mechanistic pathways interfering with Cd-related liver injury. Methods: Forty rats were randomly assigned to four groups as follows; group 1 served as negative control and received saline, group 2 received saline for 14 days and CdCl2 (3.5 mg/kg IP) as a single dose on day 14, groups 3 and 4 were treated with SA (20, 40 mg/kg PO), respectively, for 14 days and injected with CdCl2 (3.5 mg/kg IP) on day 14. Serum was collected to evaluate liver function. Liver samples were collected for histopathological examination and the assessment of markers related to oxidative stress, inflammation, and apoptosis. Results: Acute Cd administration elevated liver enzymes and induced pathological changes in liver specimens, with the concurrent release of inflammatory markers and reduced antioxidant capabilities. Pretreatment with SA improved liver function and Cd-induced histopathological changes and elevated the activities of antioxidant enzymes. SA ameliorated inflammation, as evidenced by decreased expression of NF-κB, TNF-α, TLR-4, and COX-2, iNOS, and IL-1β levels along with suppression of mTOR, JNK, ERK, BAX, and Bcl-2. Conclusions: The present data suggest that SA represents a promising protective agent against Cd-induced hepatic injury by attenuating oxidative stress, inflammation, and apoptosis.
Collapse
Affiliation(s)
- Yomna A. Farahat
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (Y.A.F.); (R.M.H.); (A.R.)
| | - Norhan M. El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (Y.A.F.); (R.M.H.); (A.R.)
| | - Reem M. Hazem
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (Y.A.F.); (R.M.H.); (A.R.)
| | - Eman T. Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Asmaa Radwan
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; (Y.A.F.); (R.M.H.); (A.R.)
| |
Collapse
|
24
|
Song Y, Ren X, Xiong J, Wang W, Zhao Q, Chang J, Yu B. Ubiquitin-Specific Protease 7 (USP7) as a Promising Therapeutic Target for Drug Discovery: From Mechanisms to Therapies. J Med Chem 2025; 68:7914-7931. [PMID: 40237780 DOI: 10.1021/acs.jmedchem.5c00102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Protein ubiquitination is a reversible post-translational modification regulated by ubiquitin-conjugating and deubiquitinating enzymes (DUBs). Ubiquitin-specific protease 7 (USP7), a well-characterized DUB, plays multifaceted roles in various cellular processes, making it a promising therapeutic target. The plasticity of its catalytic domain and unique allosteric regulation by substrates or external or intramolecular factors facilitate the identification of highly selective USP7 inhibitors. These inhibitors can engage distinct ubiquitin-binding sites through covalent or non-covalent mechanisms. Despite its therapeutic promise, no USP7 inhibitors have entered clinical trials, underscoring the urgent need for novel therapeutics. Here we provide a crystallographic and functional landscape of USP7's multilayer regulation and analyze the structure-activity relationship of inhibitors by chemotypes. Additionally, we explore USP7's roles in diseases and discuss the challenges in USP7-targeted drug discovery and future directions for therapeutic development. This Perspective aims to provide a systematic overview of USP7, from its regulatory mechanisms to its therapeutic potential.
Collapse
Affiliation(s)
- Yihui Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fujian Medical University, Fuzhou, 350122, China
| | - Xiangli Ren
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jinbo Xiong
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenwen Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Qianyan Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Junbiao Chang
- College of Chemistry, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, China
| | - Bin Yu
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou 450001, China
- College of Chemistry, Pingyuan Laboratory, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
25
|
Gu W. A bibliometric analysis of programmed cell death in oral cancer literature: research patterns and emerging trends (2000-2024). Discov Oncol 2025; 16:585. [PMID: 40261469 PMCID: PMC12014878 DOI: 10.1007/s12672-025-02410-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Programmed cell death (PCD) plays a crucial role in oral cancer pathogenesis and treatment. However, a comprehensive bibliometric analysis of the global research landscape in this field has not been conducted. This study aims to analyze the evolution and current trends of PCD research in oral cancer from 2000 to 2024. METHODS Publications were retrieved from the Web of Science Core Collection database using relevant keywords related to oral cancer and PCD. VOSviewer 1.6.20 and CiteSpace 6.1R6 software were employed to conduct bibliometric analysis, including publication trends, citation analysis, co-authorship networks, keyword co-occurrence, and research hotspots. The time span was set from January 2000 to December 2024. RESULTS A total of 963 publications were identified and analyzed. The annual publication output showed a steady increase, with a significant growth rate after 2010, dividing the study period into three distinct phases. The most productive countries were China (58.42%), South Korea (12.27%), and Japan (10.04%), with China Medical University and Kaohsiung Medical University being the leading institutions. Research hotspots evolved from traditional apoptosis studies to emerging forms of PCD such as autophagy, ferroptosis, and pyroptosis. Keyword analysis revealed three major research clusters: basic molecular mechanisms (centered around ROS and oxidative stress), clinical aspects (including prognosis and cell proliferation), and cell death pathways. Citation burst analysis identified emerging trends in targeting multiple PCD pathways simultaneously for oral cancer therapy, with special focus on treatment resistance and survival. CONCLUSION This bibliometric analysis provides a comprehensive overview of global research trends in PCD and oral cancer over the past two decades. The findings highlight the shift from basic mechanistic studies focusing on apoptosis to more diverse PCD pathways and translational research. Emerging research directions include the exploration of synergistic mechanisms among multiple PCD pathways, development of AI-based personalized treatment plans, investigation of microenvironment regulation of PCD, and application of novel drug delivery systems. These trends demonstrate the field's evolution toward more integrated, personalized approaches in oral cancer treatment. This study offers valuable insights for researchers and funding agencies to identify research gaps and potential collaboration opportunities in this rapidly developing field.
Collapse
Affiliation(s)
- Wenli Gu
- Stomatological Hospital, School of Stomatology, Southern Medical University, S366 Jiangnan Boulevard, Haizhu District, Guangzhou, Guangdong, China.
| |
Collapse
|
26
|
Yang B, Wang C, Chen X, Zhai H, Wu Y, Cui M, Wu J, Li W, Hua B. In silico and animal studies on the anti-cancer mechanisms of Shaoyao Decoction against colitis-associated colorectal cancer. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119444. [PMID: 39929402 DOI: 10.1016/j.jep.2025.119444] [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: 10/25/2024] [Revised: 01/08/2025] [Accepted: 02/03/2025] [Indexed: 03/15/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE It is well known that Shaoyao Decoction (SYD), as a commonly used formula of traditional Chinese medicine (TCM), has a beneficial effect on the treatment of ulcerative colitis (UC). It is found that SYD can also prevent colitis-associated colorectal cancer (CAC). However, its potential anti-cancer mechanism is still waiting to be revealed. AIM OF THE STUDY The aim of this study is to investigate the underlying mechanisms of SYD in inhibiting CAC through silico analysis as well as animal experiment validation. MATERIALS AND METHODS The primary active compounds, potential therapeutic targets and intervening signaling pathways, which SYD might inhibit the CAC process were predicted by network pharmacology analysis combined with our previous research result of high performance liquid chromatography (HPLC). We attempted to validate the acquired hub targets from molecular docking combined with the Gene Expression Profiling Interactive Analysis (GEPIA), the Human Protein Atlas (HPA), and the cBioPortal database comprehensively. Subsequently, an animal model of CAC mice induced by azoxymethane (AOM) and dextran sulfate sodium (DSS) was constructed and treated with SYD for 14 weeks, and tumor-related physical indicators were evaluated after sacrificed. In addition, samples of colon tissues were obtained for histologic and protein level studies to verify the predicted mechanism. RESULTS We obtained 166 active ingredients of SYD and predicted 148 potential targets through network pharmacology analysis, among which quercetin, berberine, kaempferol, wogonin and naringenin were selected as core drug ingredients, and TP53, AKT1, CASP3, PTGS2 and CCND1 were identified and included into the range of core targets. GO and KEGG analyses suggested that the PI3K-Akt signaling pathway might hold a crucial role in CAC prevention and treatment by promoting apoptosis and inhibiting tumor proliferation. In the animal experiment, both SYD and SASP treatments improved the inflammatory condition and pathological damage of the colon tissues in mice. After treatments with SYD and SASP, it was found that decreases of Cyclin D1 and Survivin expression levels and increases of p53 and Cleaved caspase-3 expression levels could be mediated by decreasing the phosphorylation levels of PI3K and Akt proteins in the colon tissues of mice. CONCLUSION The results of our study provide supports that SYD effectively inhibits CAC based on modulating PI3K-Akt signaling pathway to suppress tumor proliferation process as well as to promote tumor apoptosis process.
Collapse
Affiliation(s)
- Bingwei Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chenglei Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Xue Chen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Haoyu Zhai
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ying Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, Shaanxi University of Chinese Medicine, Xianyang, 712046, China
| | - Muyao Cui
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jiahe Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Weidong Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Baojin Hua
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| |
Collapse
|
27
|
Chen T, Hughes J, Gregory A, Conroy J, Loughran P, Song J, Chen W, Billiar T. Single-cell transcriptomic analysis identified resistant MDSCs and a stress-tolerant gene co-expression network as common MDSC features across multiple disease settings. Front Immunol 2025; 16:1565211. [PMID: 40264770 PMCID: PMC12011849 DOI: 10.3389/fimmu.2025.1565211] [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: 01/22/2025] [Accepted: 03/17/2025] [Indexed: 04/24/2025] Open
Abstract
Background Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immunosuppressive myeloid cells. The identification of a molecular signature common to MDSC regardless of tissue source would aid in the classification of cells as MDSCs. Methods Single-cell RNA sequencing (scRNA-seq) was performed on GM-CSF+ IL-6-induced human MDSCs to characterize the extent of heterogeneity within monocytic MDSCs (M-MDSCs). Cytokine-treated PBMCs were also cultured in the absence of serum to include an additional element of cell stress. Independent published bulk and single-cell transcriptomic datasets were used for validation. Findings A cluster of cells with preserved MDSC features was induced by the combination of inflammatory signals and cell stress in the form of serum starvation (resistant MDSCs, rMDSCs). A gene co-expression module (the yellow module) was identified specific to rMDSCs. The genes upregulated in MDSCs can be further classified into stress-tolerant vs. -sensitive features. This yellow module mostly contained stress-tolerant genes and showed excellent separation for distinguishing M-MDSCs from control cells across a range of in vitro and in vivo conditions (ROC AUC = 0.954), a feature not found in the stress-sensitive genes. Importantly, rMDSCs were identified in scRNA-seq datasets of immune cells from multiple human cancer types. Tumor C1Q macrophages, which have been associated with immunosuppression, highly expressed the yellow module gene signature. Interpretation These results demonstrate the importance of the combined roles of inflammation and cellular stress in shaping the features of M-MDSCs and highlight cellular resilience represented by rMDSCs and the role of stress-tolerant features in defining common MDSC features.
Collapse
Affiliation(s)
- Tianmeng Chen
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Julia Hughes
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Alyssa Gregory
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Julia Conroy
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jinming Song
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Wei Chen
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Timothy Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
28
|
Lee D, Jeong HS, Hwang SY, Lee YG, Kang YJ. ABCB1 confers resistance to carboplatin by accumulating stem-like cells in the G2/M phase of the cell cycle in p53 null ovarian cancer. Cell Death Discov 2025; 11:132. [PMID: 40175339 PMCID: PMC11965561 DOI: 10.1038/s41420-025-02435-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 02/19/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
High-grade ovarian serous carcinoma, mostly bearing the various mutations in the TP53 gene, typically relapses within six months after first-line therapy due to chemoresistance, with a median overall survival of less than a year. However, the molecular mechanisms of action behind acquired drug resistance, particularly in relation to different TP53 mutation types, have not been fully elucidated. In this study, we demonstrated that acquired resistance to carboplatin in SKOV3 harboring a p53null mutation, but not in OVCAR3 with a p53R248Q, induces a significant portion of cells accumulated in the G2/M phase of the cell cycle, where cells highly expressed stemness marker with elevated proliferative capacity, which we believe was reversed by ABCB1 inhibition to the levels observed in non-resistant parental cells. ABCB1 suppression re-sensitized carboplatin-resistant cells to additional genotoxic stress and reduced their proliferative ability by recovering DNA repair activity and lowering stemness-like features, especially in the G2/M-distributed fraction. This suggests that high levels of stemness and attenuated DNA repair function exhibited in the G2/M-accumulated portion may be a key contributor of chemoresistance in patients with ovarian cancer bearing a p53null mutation, but not other types of mutations expressing p53. Furthermore, the inhibition of ΔNp73 resulted in the suppression of ABCB1, which consequently restricted cell growth in carboplatin-resistant SKOV3, suggesting that the ΔNp73 may act as an upstream regulator of the ABCB1. Notably, combinatorial treatment of carboplatin with the p53 reactivator, APR-246, proved effective in overcoming chemoresistance in OVCAR3 with the p53R248Q. Our findings suggest that the ΔNp73-ABCB1 axis is a promising molecular target for carboplatin-resistant ovarian cancers harboring p53null mutations, which we uncovered could be utilized to increase the efficacy of conventional anti-cancer therapies, to develop more efficient combinatorial therapeutic interventions directed toward overcoming the chemoresistance and improving the survival rates in patients with ovarian cancer.
Collapse
Affiliation(s)
- Danbi Lee
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, South Korea
| | - Hyun-Seok Jeong
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, South Korea
| | - Sun-Young Hwang
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, South Korea
| | - Yu-Gyeong Lee
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, South Korea
| | - Youn-Jung Kang
- Department of Biochemistry, Research Institute for Basic Medical Science, School of Medicine, CHA University, Seongnam-si, South Korea.
| |
Collapse
|
29
|
Bhattacharya R, Avdieiev SS, Bukkuri A, Whelan CJ, Gatenby RA, Tsai KY, Brown JS. The Hallmarks of Cancer as Eco-Evolutionary Processes. Cancer Discov 2025; 15:685-701. [PMID: 40170539 DOI: 10.1158/2159-8290.cd-24-0861] [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: 06/20/2024] [Revised: 11/19/2024] [Accepted: 01/28/2025] [Indexed: 04/03/2025]
Abstract
SIGNIFICANCE Viewing the hallmarks as a sequence of adaptations captures the "why" behind the "how" of the molecular changes driving cancer. This eco-evolutionary view distils the complexity of cancer progression into logical steps, providing a framework for understanding all existing and emerging hallmarks of cancer and developing therapeutic interventions.
Collapse
Affiliation(s)
- Ranjini Bhattacharya
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Cancer Biology, University of South Florida, Tampa, Florida
| | - Stanislav S Avdieiev
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Anuraag Bukkuri
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher J Whelan
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Robert A Gatenby
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kenneth Y Tsai
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Tumor Microenvironment & Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Joel S Brown
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
30
|
Jin SK, Baek KH. Unraveling the role of deubiquitinating enzymes on cisplatin resistance in several cancers. Biochim Biophys Acta Rev Cancer 2025; 1880:189297. [PMID: 40058507 DOI: 10.1016/j.bbcan.2025.189297] [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: 01/07/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
The use of platinum-based drugs in cancer treatment is one of the most common methods in chemotherapy. Especially, cisplatin induces cell death by interrupting DNA synthesis by binding to the DNA bases, thereby leading to the apoptosis via multiple pathways. However, the major hurdle in chemotherapy is drug resistance. To overcome drug resistance, the ubiquitin-proteasome system (UPS) has emerged as a potential therapeutic target. The UPS is a pivotal signaling pathway that regulates the majority of cellular proteins by attaching ubiquitin to substrates, leading to proteasomal degradation. Conversely, deubiquitinating enzymes (DUBs) remove tagged ubiquitin from the substrate and inhibit degradation, thereby maintaining proteostasis. Recently, studies have been conducted to identify the substrates of DUBs and investigated the cellular mechanisms, and now the development of therapeutics using DUB inhibitors is in clinical trials. However, the mechanism of the DUB response to cisplatin remains still unclear. In this review, we summarize the research reported on the function of DUBs responding to cisplatin.
Collapse
Affiliation(s)
- Sun-Kyu Jin
- Department of Biomedical Science, CHA University, Gyeonggi-Do 13488, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA University, Gyeonggi-Do 13488, Republic of Korea.
| |
Collapse
|
31
|
Wang Y, Wang S, He H, Bai Y, Liu Z, Sabihi SS. Mechanisms of apoptosis-related non-coding RNAs in ovarian cancer: a narrative review. Apoptosis 2025; 30:553-578. [PMID: 39833637 DOI: 10.1007/s10495-024-02074-w] [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] [Accepted: 12/27/2024] [Indexed: 01/22/2025]
Abstract
Ovarian cancer remains a major challenge in oncology due to its complex biology and late-stage diagnosis. Recent advances in molecular biology have highlighted the crucial role of non-coding RNAs (ncRNAs) in regulating apoptosis and cancer progression. NcRNAs, including microRNAs, long non-coding RNAs, and circular RNAs, have emerged as significant players in the molecular networks governing ovarian cancer. Despite these insights, the precise mechanisms by which ncRNAs influence ovarian cancer pathology are not fully understood. This complexity, combined with the heterogeneity of the disease and the development of treatment resistance, poses substantial obstacles to effective therapeutic development. Additionally, the lack of reliable early detection methods further complicates treatment strategies. This manuscript reviews the current state of research on ncRNAs in ovarian cancer, discusses the challenges in translating these findings into clinical applications, and outlines potential future directions. Emphasis is placed on the need for integrated approaches to unravel the intricate roles of ncRNAs, improve early detection, and develop personalized treatment strategies to address the diverse and evolving nature of ovarian cancer. While these findings provide valuable insights, it is crucial to recognize that many results are based on preclinical studies and require further validation to establish their clinical applicability.
Collapse
Affiliation(s)
- Yue Wang
- Department of Obstetrics and Gynecology, Tang Du Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Shirui Wang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710038, China
| | - Haiyan He
- Department of Obstetrics and Gynecology, Tang Du Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Yingying Bai
- Department of Obstetrics and Gynecology, Tang Du Hospital, The Air Force Military Medical University, Xi'an, 710038, China
| | - Zhuo Liu
- Department of Obstetrics and Gynecology, Xi'an International Medical Center Hospital, Xi'an, 710038, China
| | - Sima-Sadat Sabihi
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
32
|
Zuazo-Gaztelu I, Lawrence D, Oikonomidi I, Marsters S, Pechuan-Jorge X, Gaspar CJ, Kan D, Segal E, Clark K, Beresini M, Braun MG, Rudolph J, Modrusan Z, Choi M, Sandoval W, Reichelt M, DeWitt DC, Kujala P, van Dijk S, Klumperman J, Ashkenazi A. A nonenzymatic dependency on inositol-requiring enzyme 1 controls cancer cell cycle progression and tumor growth. PLoS Biol 2025; 23:e3003086. [PMID: 40208872 PMCID: PMC12080931 DOI: 10.1371/journal.pbio.3003086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/15/2025] [Accepted: 02/26/2025] [Indexed: 04/12/2025] Open
Abstract
Endoplasmic-reticulum resident inositol-requiring enzyme 1α (IRE1) supports protein homeostasis via its cytoplasmic kinase-RNase module. Known cancer dependency on IRE1 entails its enzymatic activation of the transcription factor XBP1s and of regulated RNA decay. We discovered surprisingly that some cancer cell lines require IRE1 but not its enzymatic activity. IRE1 knockdown but not enzymatic IRE1 inhibition or XBP1 disruption attenuated cell cycle progression and tumor growth. IRE1 silencing led to activation of TP53 and CDKN1A/p21 in conjunction with increased DNA damage and chromosome instability, while decreasing heterochromatin as well as DNA and histone H3K9me3 methylation. Immunoelectron microscopy detected some endogenous IRE1 protein at the nuclear envelope. Thus, cancer cells co-opt IRE1 either enzymatically or nonenzymatically, which has significant implications for IRE1's biological role and therapeutic targeting.
Collapse
Affiliation(s)
- Iratxe Zuazo-Gaztelu
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - David Lawrence
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - Ioanna Oikonomidi
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - Scot Marsters
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - Ximo Pechuan-Jorge
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - Catarina J. Gaspar
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - David Kan
- Department of In Vivo Pharmacology, Genentech, Inc., South San Francisco, California, United States of America
| | - Ehud Segal
- Department of In Vivo Pharmacology, Genentech, Inc., South San Francisco, California, United States of America
| | - Kevin Clark
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California, United States of America
| | - Maureen Beresini
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California, United States of America
| | - Marie-Gabrielle Braun
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, California, United States of America
| | - Joachim Rudolph
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, California, United States of America
| | - Zora Modrusan
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, California, United States of America
| | - Meena Choi
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, California, United States of America
| | - Wendy Sandoval
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, California, United States of America
| | - Mike Reichelt
- Department of Pathology, Genentech, Inc., South San Francisco, California, United States of America
| | - David C. DeWitt
- Department of Pathology, Genentech, Inc., South San Francisco, California, United States of America
| | - Pekka Kujala
- Center for Molecular Medicine—Cell Biology, University Medical Center, Utrecht, The Netherlands
| | - Suzanne van Dijk
- Center for Molecular Medicine—Cell Biology, University Medical Center, Utrecht, The Netherlands
| | - Judith Klumperman
- Center for Molecular Medicine—Cell Biology, University Medical Center, Utrecht, The Netherlands
| | - Avi Ashkenazi
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| |
Collapse
|
33
|
Quintavalle C, Malapelle U, De Martino M, Rocco D, Fusco A, Pepe F, Bellevicine C, Esposito F, Pallante P. The Interconnection Between UbcH10, p53, and EGFR in Lung Cancer Cells and Their Involvement in Treatment Response. Genes (Basel) 2025; 16:404. [PMID: 40282365 PMCID: PMC12026858 DOI: 10.3390/genes16040404] [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: 01/29/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: The UbcH10 protein plays an important role in a variety of human malignancies, including thyroid, breast, ovarian, and colorectal carcinomas. It has been previously reported that UbcH10 is overexpressed in non-small cell lung cancer (NSCLC) compared to normal lungs and that its expression is directly and inversely correlated with the mutational status of p53 and EGFR, respectively. Methods: We transfected lung cancer cells with wild-type and mutant forms of EGFR, modulated the expression of UbcH10 and p53, and treated these cells with tyrosine kinase inhibitor (TKI) erlotinib. Using Western blotting, we evaluated the expression of UbcH10 induced by EGFR and p53. Finally, we employed immunohistochemistry to assess the levels of UbcH10 expression in a subset of NSCLC patients receiving TKI therapy. Results: We reported a possible modulation of UbcH10 expression by the overexpression of wild-type and mutant EGFR in H460 lung cancer cells, potentially through p53. The enforced expression of UbcH10 in cells transfected with mutant EGFR suggested a potential increase in resistance to erlotinib treatment. Finally, immunohistochemical analysis of samples from NSCLC patients with mutant EGFR indicated a possible connection between UbcH10 expression levels and progression-free survival. Conclusions: In NSCLC, UbcH10 may play a role in the regulation of TKI response via a molecular pathway potentially involving p53 and EGFR. However, further research is needed to fully understand this mechanism.
Collapse
Affiliation(s)
- Cristina Quintavalle
- Institute of Endotypes in Oncology, Metabolism and Immunology (IEOMI) “G. Salvatore”, National Research Council (CNR), S. Pansini 5, 80131 Naples, Italy; (C.Q.); (F.E.)
| | - Umberto Malapelle
- Department of Public Health, University of Naples “Federico II”, S. Pansini 5, 80131 Naples, Italy; (U.M.); (F.P.); (C.B.)
| | - Marco De Martino
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples “Federico II”, S. Pansini 5, 80131 Naples, Italy; (M.D.M.); (A.F.)
| | - Danilo Rocco
- Department of Pulmonary Oncology, “V. Monaldi” Hospital, AORN Ospedali dei Colli, L. Bianchi, 80131 Naples, Italy;
| | - Alfredo Fusco
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples “Federico II”, S. Pansini 5, 80131 Naples, Italy; (M.D.M.); (A.F.)
| | - Francesco Pepe
- Department of Public Health, University of Naples “Federico II”, S. Pansini 5, 80131 Naples, Italy; (U.M.); (F.P.); (C.B.)
| | - Claudio Bellevicine
- Department of Public Health, University of Naples “Federico II”, S. Pansini 5, 80131 Naples, Italy; (U.M.); (F.P.); (C.B.)
| | - Francesco Esposito
- Institute of Endotypes in Oncology, Metabolism and Immunology (IEOMI) “G. Salvatore”, National Research Council (CNR), S. Pansini 5, 80131 Naples, Italy; (C.Q.); (F.E.)
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples “Federico II”, S. Pansini 5, 80131 Naples, Italy; (M.D.M.); (A.F.)
| | - Pierlorenzo Pallante
- Institute of Endotypes in Oncology, Metabolism and Immunology (IEOMI) “G. Salvatore”, National Research Council (CNR), S. Pansini 5, 80131 Naples, Italy; (C.Q.); (F.E.)
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples “Federico II”, S. Pansini 5, 80131 Naples, Italy; (M.D.M.); (A.F.)
| |
Collapse
|
34
|
Yang Y, Du Y, Ma X, Yuan G, Li G, Zhang Q, Zhou S. Transcription factor addictions: exploring the potential Achilles' Heel of endometriosis. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2832-8. [PMID: 40163264 DOI: 10.1007/s11427-024-2832-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/15/2024] [Indexed: 04/02/2025]
Abstract
A considerable number of women of reproductive age suffer from endometriosis worldwide. There is a significant physical, mental, and financial burden on patients affected by this condition in terms of pelvic pain, either continuously or intermittently, dysmenorrhea, infertility, and a higher risk of certain types of cancer. Several treatments available in clinical settings for endometriosis management do not provide adequate efficacy and have undesirable side effects. Transcription factors (TFs) are crucial regulators of key biological processes involved in endometriosis. Here, we elaborated on the research progress regarding the crucial roles of TFs in endometriosis, emphasizing their implications for clinical outcomes and critical therapeutic contributions. By delving into their involvement in key processes, such as cell proliferation and apoptosis, we revealed the multifaceted role of key TFs in disease progression. We aimed to provide a systemic understanding of TFs regulation in endometriosis pathogenesis, establishing a foundation for innovative treatment approaches.
Collapse
Affiliation(s)
- Yang Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Yi Du
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Yuan
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Guobo Li
- Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Qian Zhang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China.
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China.
| |
Collapse
|
35
|
Salari R, Rastegari B, Hashemi A, Farjadfar A, Masoomi MY. P53 Gene Therapy with ZIF-8 Metal-Organic Framework: A Platform in Cancer Gene Therapy. ACS OMEGA 2025; 10:10891-10902. [PMID: 40160747 PMCID: PMC11947847 DOI: 10.1021/acsomega.4c08739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 04/02/2025]
Abstract
Gene therapy holds great promise as a therapeutic approach for combating cancer, with the choice of gene delivery vector being a critical factor in its success. In recent years, metal-organic frameworks (MOFs) have emerged as valuable tools for intracellular plasmid delivery in this field. This study aimed to encapsulate plasmid DNA encoding the TP53 tumor suppressor gene (pEGFP-N1-TP53) within zeolitic imidazolate framework-8 (ZIF-8) MOFs and ZIF-8-PEI. Subsequently, the transfection efficiency and ability to induce cell death were assessed in MDA-MB-231, MCF-7, and HeLa cancer cells. A comparative analysis was conducted to evaluate the induction of cell death by pEGFP-N1-TP53@ZIF-8-PEI, pEGFP-N1-TP53-ZIF-8 nanoparticles, and Lipofectamine in the aforementioned cell lines. Additionally, an optimal condition for loading the plasmid into ZIF-8 was proposed. The findings from cell transfection assays, MTT assay, and flow cytometry revealed that both pEGFP-N1-TP53@ZIF-8-PEI and pEGFP-N1-TP53-ZIF-8 effectively delivered the plasmid to the cells. Notably, pEGFP-N1-TP53@ZIF-8-PEI exhibited significant results, inducing 77% cell death in the HeLa cell line and 73% in the MDA-MB-231 cell line. Our observations indicated that MDA-MB-231 and HeLa cells exhibited heightened responsiveness to TP53 gene therapy when delivered through ZIF-8-PEI and ZIF-8. Based on these findings, further investigation of pEGFP-N1-TP53@ZIF-8-PEI as a potential cancer therapeutic platform in other cancer cell types is warranted.
Collapse
Affiliation(s)
- Roya Salari
- Department
of Medical Biotechnology, Fasa University
of Medical Sciences, Fasa 7461686688, Iran
| | - Banafsheh Rastegari
- Diagnostic
Laboratory Sciences and Technology Research Center, Paramedical School, Shiraz University of Medical Sciences, Shiraz 71348-45794, Iran
| | - Amin Hashemi
- Department
of Cell Biology, Faculty of Basic Sciences, Shahed University, Tehran 3319118651, Iran
| | - Akbar Farjadfar
- Department
of Medical Biotechnology, Fasa University
of Medical Sciences, Fasa 7461686688, Iran
| | | |
Collapse
|
36
|
Vogler M, Braun Y, Smith VM, Westhoff MA, Pereira RS, Pieper NM, Anders M, Callens M, Vervliet T, Abbas M, Macip S, Schmid R, Bultynck G, Dyer MJ. The BCL2 family: from apoptosis mechanisms to new advances in targeted therapy. Signal Transduct Target Ther 2025; 10:91. [PMID: 40113751 PMCID: PMC11926181 DOI: 10.1038/s41392-025-02176-0] [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: 11/07/2024] [Revised: 12/21/2024] [Accepted: 02/10/2025] [Indexed: 03/22/2025] Open
Abstract
The B cell lymphoma 2 (BCL2) protein family critically controls apoptosis by regulating the release of cytochrome c from mitochondria. In this cutting-edge review, we summarize the basic biology regulating the BCL2 family including canonical and non-canonical functions, and highlight milestones from basic research to clinical applications in cancer and other pathophysiological conditions. We review laboratory and clinical development of BH3-mimetics as well as more recent approaches including proteolysis targeting chimeras (PROTACs), antibody-drug conjugates (ADCs) and tools targeting the BH4 domain of BCL2. The first BCL2-selective BH3-mimetic, venetoclax, showed remarkable efficacy with manageable toxicities and has transformed the treatment of several hematologic malignancies. Following its success, several chemically similar BCL2 inhibitors such as sonrotoclax and lisaftoclax are currently under clinical evaluation, alone and in combination. Genetic analysis highlights the importance of BCL-XL and MCL1 across different cancer types and the possible utility of BH3-mimetics targeting these proteins. However, the development of BH3-mimetics targeting BCL-XL or MCL1 has been more challenging, with on-target toxicities including thrombocytopenia for BCL-XL and cardiac toxicities for MCL1 inhibitors precluding clinical development. Tumor-specific BCL-XL or MCL1 inhibition may be achieved by novel targeting approaches using PROTACs or selective drug delivery strategies and would be transformational in many subtypes of malignancy. Taken together, we envision that the targeting of BCL2 proteins, while already a success story of translational research, may in the foreseeable future have broader clinical applicability and improve the treatment of multiple diseases.
Collapse
Affiliation(s)
- Meike Vogler
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK) partner site Frankfurt/Mainz, a partnership between DKFZ and University Hospital Frankfurt, Frankfurt am Main, Germany.
- University Cancer Center Frankfurt (UCT), University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany.
| | - Yannick Braun
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
- Department of Pediatric Surgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Victoria M Smith
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Raquel S Pereira
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Nadja M Pieper
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Marius Anders
- Goethe University Frankfurt, Institute for Experimental Pediatric Hematology and Oncology, Frankfurt am Main, Germany
| | - Manon Callens
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Tim Vervliet
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Maha Abbas
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
| | - Salvador Macip
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- Josep Carreras Leukaemia Research Institute, Badalona, Spain
- FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
| | - Ralf Schmid
- Department of Molecular and Cell Biology, University of Leicester, Leicester, UK
- Institute for Structural and Chemical Biology, University of Leicester, Leicester, UK
| | - Geert Bultynck
- KU Leuven, Lab. Molecular & Cellular Signaling, Dep. Cellular & Molecular Medicine, and Leuven Kankerinstituut (LKI), Leuven, Belgium
| | - Martin Js Dyer
- The Ernest and Helen Scott Haematological Research Institute, Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
37
|
ZHAO SHUANG, WEN HONGYONG, WANG BAIQI, XIONG QINGLIN, LI LANXIN, CHENG AILAN. p53: A player in the tumor microenvironment. Oncol Res 2025; 33:795-810. [PMID: 40191727 PMCID: PMC11964878 DOI: 10.32604/or.2025.057317] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/09/2024] [Indexed: 04/09/2025] Open
Abstract
Approximately half of all cancers have p53 inactivating mutations, in addition to which most malignancies inactivate the p53 pathway by increasing p53 inhibitors, decreasing p53 activators, or inactivating p53 downstream targets. A growing number of researches have demonstrated that p53 can influence tumor progression through the tumor microenvironment (TME). TME is involved in the process of tumor development and metastasis and affects the clinical prognosis of patients. p53 participates in host immunity and engages in the immune landscape of the TME, but the specific mechanisms remain to be investigated. This review briefly explores the interactions between different states of p53 and TME components and their mechanisms, as well as their effects on tumor progression. To understand the progress of drug development and clinical studies related to p53 and tumor microenvironment.
Collapse
Affiliation(s)
- SHUANG ZHAO
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - HONGYONG WEN
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - BAIQI WANG
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - QINGLIN XIONG
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - LANXIN LI
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - AILAN CHENG
- Hunan Engineering Research Center for Early Diagnosis and Treatment of Liver Cancer, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|
38
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
39
|
Sharaky M, El Kiki SM, Effat H, Mansour HH. Effect of palliative radiotherapy and cyclin-dependent kinase 4/6 inhibitor on breast cancer cell lines. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03878-6. [PMID: 40035822 DOI: 10.1007/s00210-025-03878-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/03/2025] [Indexed: 03/06/2025]
Abstract
The most prevalent disease in the world and the main reason for women mortality from cancer is breast cancer. The recommended treatment for hormone receptor-positive metastatic breast cancer (MBC) is cyclin-dependent kinase 4/6 inhibitor (CDK4/6i), Abemaciclib. Radiotherapy (RT) is one of the main options to control breast cancer. This work intended to examine the impact of CDK 4/6i and palliative radiation on human breast cancer cell lines. Breast cancer cell lines (MCF7, MDA-MD-468, and MDA-MD-231) were treated with varying doses of Abemaciclib and left to incubate for 48 h. Different radiation doses were applied to the lines that had the best IC50. The intrinsic treatment objectives for MBC are presented in this study, along with the PI3K/AKT/mTOR pathway; CDK4, CDK6, and the NF-κβ/TGF-β pathway; BAX/BcL2, P53; caspase-3, caspase-6, caspase-7, caspase-8, and caspase-9; cytokeratin 18 (CK18); cycloxygenase-2 (COX2); IL-6; IL1β; matrix metalloproteinases (MMP2 and MMP9); and oxidative stress markers. The biochemical assays revealed that abemaciclib hindered the progression of breast cancer cells MDA-MB-231 and MCF-7 and enhanced RT (10 Gy) by provoking cell cycle arrest throughout the restraint of CDK4 and CDK6 expression and increasing apoptosis, in addition to decreasing the PI3K/AKT/mTOR and NF-κβ/TGF-β pathway expression; inhibiting CK18 and COX2 activity; boosting the protein concentration of BAX and P53; and decreasing Bcl-2, IL-6, IL-1β, MMP2, and MMP9, modulating oxidative stress markers. These results implied potential effects of radiation and CDK4/6i abemaciclib on breast cancer cell lines.
Collapse
Affiliation(s)
- Marwa Sharaky
- Pharmacology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Shereen M El Kiki
- Health Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, P.O. Box 29, Nasr City, Cairo, Egypt
| | - Heba Effat
- Medical Biochemistry and Molecular Biology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Heba H Mansour
- Health Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, P.O. Box 29, Nasr City, Cairo, Egypt.
| |
Collapse
|
40
|
Luo R, Wang B, Luo R. Zearalenone delays tissue regeneration by dysregulating neutrophil balance in zebrafish (Danio rerio) larvae. Comp Biochem Physiol C Toxicol Pharmacol 2025; 289:110105. [PMID: 39701198 DOI: 10.1016/j.cbpc.2024.110105] [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: 08/06/2024] [Revised: 11/23/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024]
Abstract
Zearalenone (ZEA), a common mycotoxin, poses significant environmental and health risks. While its toxicological effects are well-studied, its impact on regeneration remains unclear. This study explored ZEA's effects on zebrafish (Danio rerio) larvae, focusing on developmental toxicity, immunotoxicity, and tissue regeneration. Embryos were exposed to 0, 0.5, 1, and 1.5 μM ZEA from 6 to 72 h post-fertilization (hpf). Although hatching and survival rates remained unaffected, malformations, including body axis bending and pericardial edema, increased dose-dependently, with 4.44 % abnormalities observed at 1.5 μM (p = 0.01). Heart rates also declined significantly at 1.5 μM (75.40 vs. 72.53 beats/30s, p = 0.0054). Immunotoxicity was assessed using Tg(mpx: eGFP) zebrafish to monitor neutrophil responses post-injury. ZEA exposure led to increased neutrophil counts (229.87 vs. 330.80, p < 0.0001) and chemotaxis (21.15 % vs. 34.57 %, p < 0.0001). RNA sequencing of 0 and 1.5 μM groups revealed disrupted redox balance and oxygen transport, with down-regulation of hbae1, hbbe2, and hbae3 and up-regulation of hif1a, indicating hypoxia involvement. Elevated reactive oxygen species (ROS), reduced antioxidant enzyme activity, and increased apoptosis were also observed. Tail fin regeneration assays showed delayed regeneration at 1 and 1.5 μM ZEA, linked to impaired immune function and oxidative stress. These findings highlight ZEA's adverse effects on developmental and regenerative processes, underscoring its environmental and health implications and the need for further research.
Collapse
Affiliation(s)
- Rui Luo
- Department of Pathology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bo Wang
- Department of Medical Laboratory, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Luo
- Department of Pathology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
41
|
Qiu Y, Li Y, Li M, Wang Y, Shen M, Shao J, Zhang F, Xu X, Wang F, Zhang Z, Zheng S. NUMB endocytic adaptor protein (NUMB) mediates the anti-hepatic fibrosis effect of artesunate (ART) by inducing senescence in hepatic stellate cells (HSCs). Chin J Nat Med 2025; 23:322-333. [PMID: 40122662 DOI: 10.1016/s1875-5364(25)60836-3] [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: 12/21/2023] [Revised: 03/13/2024] [Accepted: 03/28/2024] [Indexed: 03/25/2025]
Abstract
Developing and identifying effective medications and targets for treating hepatic fibrosis is an urgent priority. Our previous research demonstrated the efficacy of artesunate (ART) in alleviating liver fibrosis by eliminating activated hepatic stellate cells (HSCs). However, the underlying mechanism remains unclear despite these findings. Notably, endocytic adaptor protein (NUMB) has significant implications for treating hepatic diseases, but current research primarily focuses on liver regeneration and hepatocellular carcinoma. The precise function of NUMB in liver fibrosis, particularly its ability to regulate HSCs, requires further investigation. This study aims to elucidate the role of NUMB in the anti-hepatic fibrosis action of ART in HSCs. We observed that the expression level of NUMB significantly decreased in activated HSCs compared to quiescent HSCs, exhibiting a negative correlation with the progression of liver fibrosis. Additionally, ART induced senescence in activated HSCs through the NUMB/P53 tumor suppressor (P53) axis. We identified NUMB as a crucial regulator of senescence in activated HSCs and as a mediator of ART in determining cell fate. This research examines the specific target of ART in eliminating activated HSCs, providing both theoretical and experimental evidence for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Yangling Qiu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yujia Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mengran Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yingqian Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Min Shen
- Department of Biochemistry and Molecular Biology, Medical College, Yangzhou University, Yangzhou 225009, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xuefen Xu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Feixia Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
42
|
Tabbasam R, Khursid S, Ishaq Y, Farrukh SY. Synergistic Cytotoxic Effects of Doxorubicin Loaded Silver, Gold and Zinc Oxide Nanoparticles in HepG2 Liver Cancer Cells. BIONANOSCIENCE 2025; 15:105. [DOI: 10.1007/s12668-024-01747-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2024] [Indexed: 01/05/2025]
|
43
|
Chen X, Wu Y, Xing Y, Zhong P. CENPF as a prognostic marker of glioma: unraveling the molecular mechanisms. J Cancer Res Clin Oncol 2025; 151:96. [PMID: 40019588 PMCID: PMC11870995 DOI: 10.1007/s00432-025-06144-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/17/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVE Glioma is the dominant primary intracranial malignancy. The roles of CENPF and the CENPF - p53 axis in glioma remain elusive. This study uses bioinformatics and animal experiments to clarify the relationship between CENPF and p53 in glioma. CENPF affects spindle assembly and chromosomal segregation, while p53 is a tumor-suppressor gene. Their dysregulation may interact and impact glioma development. Our research aims to uncover the underlying molecular mechanisms, offering new perspectives for glioma diagnosis and treatment. METHOD Gene expression data from the Gene Expression Omnibus (GEO) database ( http://www.ncbi.nlm.nih.gov/geo/ ) were retrieved, specifically datasets GSE50161, GSE104291, and GSE12249. Volcano plots were generated to visualize differentially expressed genes (DEGs), and intersecting DEGs were identified using Venn diagrams. Weighted gene co-expression network analysis (WGCNA) was employed to construct and analyze the protein-protein interaction (PPI) network. Additionally, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were conducted. Gene set enrichment analysis (GSEA) was utilized for comprehensive GO and KEGG analyses of the entire genome. Comparative Toxicogenomics Database (CTD) analysis was performed, and TargetScan was used to identify miRNAs regulating central DEGs. An animal model of glioma was established and analyzed via Western blot. RESULT A total of 501 differentially expressed genes (DEGs) were identified, from which eight significant modules were generated and ten core genes were extracted. These core genes exhibited differential expression patterns between glioma tumor and non-tumor samples. Expression analysis revealed that the ten core genes associated with glioma (CENPF, PBK, ASPM, KIF2C, KIF20A, CDC20, TOP2A, NUSAP1, TTK, KIF23) were significantly upregulated in tumor tissues (P < 0.05). They are primarily enriched in protein signal transduction, coated membrane structures, AP-type membrane coat adaptor complexes, and chloride channel activity. KEGG pathway analysis indicated that these target genes were mainly involved in nicotine addiction, arginine and proline metabolism, beta-alanine metabolism, and histidine metabolism. The mouse model confirmed that CENPF and CDK-1 were highly expressed in glioma tissues, while p53, p21, and Caspase9 were downregulated, leading to inhibition of the apoptosis pathway and exacerbation of glioma progression. Overexpression of CENPF further suppressed key molecules in the p53-mediated apoptosis pathway. Conversely, low expression of CENPF activated these key molecules, inducing apoptosis in glioma cells. CONCLUSIONS CENPF exhibits elevated expression levels in glioma, potentially inhibiting cell apoptosis via the p53 signaling pathway, consequently contributing to the onset and progression of glioma.
Collapse
Affiliation(s)
- Xiuyang Chen
- Department of Neurological Care Unit, The Affliated Yantai Yuhuangding Hospital of Qingdao University, No. 20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, People's Republic of China
| | - Yiwei Wu
- Department of Orthopaedic Surgery, Yantaishan Hospital, Yantai, 264000, Shandong, People's Republic of China
| | - Yining Xing
- Department of Neurological Care Unit, The Affliated Yantai Yuhuangding Hospital of Qingdao University, No. 20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, People's Republic of China
- Department of Orthopaedic Surgery, Yantaishan Hospital, Yantai, 264000, Shandong, People's Republic of China
| | - Peng Zhong
- Department of Neurological Care Unit, The Affliated Yantai Yuhuangding Hospital of Qingdao University, No. 20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, People's Republic of China.
| |
Collapse
|
44
|
Chang L, Xiang QM, Liao ZT, Zhu JQ, Mu CK, Wang CL, Hou CC. Pt-LIS1 participates nuclear deformation and acrosome formation via regulating Dynein-1 during spermatogenesis in Portunus trituberculatus. Sci Rep 2025; 15:6632. [PMID: 39994263 PMCID: PMC11850704 DOI: 10.1038/s41598-024-83566-2] [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: 06/03/2024] [Accepted: 12/16/2024] [Indexed: 02/26/2025] Open
Abstract
Spermatogenesis involves complex dynamic mechanisms. Dynein-1 is a key carrier in cellular cargo transport, participating in nuclear deformation and acrosome formation during spermatogenesis. However, the regulatory mechanisms of Dynein-1 during cargo transport remain unknown. In this study, we explored the role of Lissencephaly 1 (LIS1) in spermatogenesis and its impact on Dynein-1 cargo transport in Portunus trituberculatus. LIS1 was known as a Dynein-1 regulator, which is a causative gene of anencephaly syndrome. Pt-Lis1 was cloned from the crab testis, and its highly expression was observed in the testis. Pt-LIS1 dynamically localized around the nucleus and acrosome during spermatogenesis, colocalizing with Dynein-1 subunits, microtubules, mitochondrial markers (PHB), and Acrosin. RNA interference reduced Pt-Lis1 expression, leading to decreased expression of Pt-dhc and Pt-dic. During spermatogenesis, the signals of Pt-LIS1, Pt-DHC, Pt-DIC, and α-Tubulin were weakened and showed disorganized distribution. The colocalization of Pt-LIS1 with Pt-DHC and Pt-DIC decreased, while abnormal colocalization significantly increased. In addition, caspase-3 and p53 expression significantly increased after Pt-Lis1 silencing, indicating association with apoptosis in spermatogenic cells. All these results suggest that LIS1 played a crucial role in crustacean spermatogenesis by regulating nuclear deformation and acrosome formation through modulating Dynein-1 transport cargoes along microtubules.
Collapse
Affiliation(s)
- Le Chang
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education and Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, 315832, China
| | - Qiu Meng Xiang
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education and Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, 315832, China
| | - Zai-Tian Liao
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education and Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, 315832, China
| | - Jun-Quan Zhu
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education and Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, 315832, China
| | - Chang-Kao Mu
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education and Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, 315832, China
| | - Chun-Lin Wang
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education and Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, 315832, China
| | - Cong-Cong Hou
- Key Laboratory of Applied Marine Biotechnology By the Ministry of Education and Key Laboratory of Marine Biotechnology of Zhejiang Province, School of Marine Sciences, Ningbo University, Ningbo, 315832, China.
| |
Collapse
|
45
|
Cordes J, Zhao S, Engel CM, Stingele J. Cellular responses to RNA damage. Cell 2025; 188:885-900. [PMID: 39983673 DOI: 10.1016/j.cell.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/26/2024] [Accepted: 01/02/2025] [Indexed: 02/23/2025]
Abstract
RNA plays a central role in protein biosynthesis and performs diverse regulatory and catalytic functions, making it essential for all processes of life. Like DNA, RNA is constantly subjected to damage from endogenous and environmental sources. However, while the DNA damage response has been extensively studied, it was long assumed that RNA lesions are relatively inconsequential due to the transient nature of most RNA molecules. Here, we review recent studies that challenge this view by revealing complex RNA damage responses that determine survival when cells are exposed to nucleic acid-damaging agents and promote the resolution of RNA lesions.
Collapse
Affiliation(s)
- Jacqueline Cordes
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Shubo Zhao
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, 81377 Munich, Germany; College of Basic Medical Sciences, Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Carla M Engel
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Julian Stingele
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, 81377 Munich, Germany.
| |
Collapse
|
46
|
Persiani E, Cecchettini A, Amato S, Ceccherini E, Gisone I, Sgalippa A, Ippolito C, Castelvetro V, Lomonaco T, Vozzi F. Virgin and photo-degraded microplastics induce the activation of human vascular smooth muscle cells. Sci Rep 2025; 15:4263. [PMID: 39905115 PMCID: PMC11794685 DOI: 10.1038/s41598-025-89006-z] [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: 08/27/2024] [Accepted: 02/03/2025] [Indexed: 02/06/2025] Open
Abstract
Microplastics (MPs) are an emerging environmental issue due to their accumulation in ecosystems and living organisms. Increasing evidence shows that MPs impact vascular function, with recent studies finding MPs in atheromas linked to cardiovascular events. Since vascular smooth muscle cells (VSMCs) are crucial to maintaining vascular function, this study examined how MPs activate VSMCs, leading to cardiovascular diseases like atherosclerosis and vascular calcification. The study used polyethylene (PE) and polystyrene (PS), common in food packaging, as "virgin" or photo-degraded to simulate environmental conditions. VSMC viability, apoptosis, cytotoxicity, inflammation, and activation markers were evaluated. PE and PS affected VSMC viability, induced apoptosis, and triggered pathological changes such as altered migration and proliferation. Key markers like RUNX-2 and galectin-3, which regulate cardiovascular pathology, were activated, alongside the inflammasome complex. In conclusion, MPs can induce harmful activation of VSMCs, posing potential health risks through inflammation, cell damage, and phenotypic changes. Understanding these toxic mechanisms may reveal critical pathways for intervention and prevention.
Collapse
MESH Headings
- Humans
- Microplastics/toxicity
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Apoptosis/drug effects
- Cell Survival/drug effects
- Cell Proliferation/drug effects
- Polystyrenes
- Cell Movement/drug effects
- Polyethylene/toxicity
- Cells, Cultured
- Inflammation
Collapse
Affiliation(s)
| | - Antonella Cecchettini
- CNR Institute of Clinical Physiology, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Sofia Amato
- CNR Institute of Clinical Physiology, Pisa, Italy
| | | | | | | | - Chiara Ippolito
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Valter Castelvetro
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | - Tommaso Lomonaco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Pisa, Italy
| | | |
Collapse
|
47
|
Lin Z, Feng Y, Wang J, Men Z, Ma X. Microbiota governs host chenodeoxycholic acid glucuronidation to ameliorate bile acid disorder induced diarrhea. MICROBIOME 2025; 13:36. [PMID: 39905483 PMCID: PMC11792533 DOI: 10.1186/s40168-024-02011-8] [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: 03/08/2024] [Accepted: 12/17/2024] [Indexed: 02/06/2025]
Abstract
BACKGROUND Disorder in bile acid (BA) metabolism is known to be an important factor contributing to diarrhea. However, the pathogenesis of BA disorder-induced diarrhea remains unclear. METHODS The colonic BA pool and microbiota between health piglets and BA disorder-induced diarrheal piglets were compared. Fecal microbiota transplantation and various cell experiments further indicated that chenodeoxycholic acid (CDCA) metabolic disorder produced CDCA-3β-glucuronide, which is the main cause of BA disorder diarrhea. Non-targeted metabolomics uncovered the inhibition of the BA glucuronidation by Lactobacillus reuteri (L. reuteri) is through deriving indole-3-carbinol (I3C). In vitro, important gene involved in the reduction of BA disorder induced-diarrhea were screened by RNA transcriptomics sequencing, and activation pathway of FXR-SIRT1-LKB1 to alleviate BA disorder diarrhea and P53-mediated apoptosis were proposed in vitro by multifarious siRNA interference, CO-IP, immunofluorescence, and so on, which mechanism was also verified in a variety of mouse models. RESULTS Here, we reveal for the first time that core microbiota derived I3C represses gut epithelium glucuronidation, particularly 3β-glucuronic CDCA production, which reaction is mediated by host UDP glucuronosyltransferase family 1 member A4 (UGT1A4) and necessary of BA disorder induced diarrhea. Mechanistically, L. reuteri derived I3C activates aryl hydrocarbon receptor to decrease UGT1A4 transcription and CDCA-3β-glucuronide content, thereby upregulating FXR-SIRT1-LKB1 signal. LKB1 binds with P53 based on protein interaction, ultimately resists to apoptosis and diarrhea. Moreover, I3C assists CDCA to attain the ameliorative effects of FXR activation in BA disorder diarrhea, through reversion of abnormal metabolism pathway, improving the outcomes of CDCA supplement. CONCLUSION These findings uncover the crucial interplay between gut epithelial cells and microbes, highlighting UGT1A4-mediated conversion of CDCA-3β-glucuronide as a key target for ameliorating BA disorder-induced diarrhea. Video Abstract.
Collapse
Affiliation(s)
- Zishen Lin
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Yue Feng
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Jinping Wang
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Zhaoyue Men
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition and Feeding, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, China.
| |
Collapse
|
48
|
Balahura (Stămat) LR, Dinu AI, Lungu A, Herman H, Balta C, Hermenean A, Șerban AI, Dinescu S. Implantable Polymer Scaffolds Loaded with Paclitaxel-Cyclodextrin Complexes for Post-Breast Cancer Tissue Reconstruction. Polymers (Basel) 2025; 17:402. [PMID: 39940603 PMCID: PMC11819909 DOI: 10.3390/polym17030402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
The side effects associated with the chemotherapy of triple-negative breast cancer (TNBC), such as nucleotide-binding oligomerization domain (NOD)-like receptor family (NLR), pyrin domain containing 3 (NLRP3) inflammasome activity, are responsible for the treatment failure and high mortality rates. Therefore, advanced delivery systems have been developed to improve the transport and targeted administration of anti-tumor agents at the tumor sites using tissue engineering approaches. Implantable delivery systems based on biodegradable polymers are an effective alternative due high biocompatibility, porosity, and mechanical strength. Moreover, the use of paclitaxel (PTX)-cyclodextrin complexes increases the solubility and permeability of PTX, enhancing the bioavailability and efficacy of the drug. All of these properties contribute to the efficient encapsulation and controlled release of drugs, preventing the damage of healthy tissues. In the current study, we detailed the synthesis process and evaluation of 3D scaffolds based on gelatin functionalized with methacryloyl groups (GelMA) and pectin loaded with PTX-cyclodextrin inclusion complexes on TNBC pathogenesis in vitro and in vivo. Bio-physio-chemical analysis of the proposed scaffolds revealed favorable mechanical and biological properties for the cellular component. To improve the drug solubility, a host-guest interaction was performed by the complexation of PTX with a cyclodextrin derivative prior to scaffold synthesis. The presence of PTX suppressed the growth of breast tumor cells and promoted caspase-1 activity, the release of interleukin (IL)-1β, and the production of reactive oxygen species (ROS), conditioning the expression levels of the genes and proteins associated with breast tumorigenesis and NLRP3 inflammasome. The in vivo experiments suggested the activation of pyroptosis tumor cell death, confirming the in vitro experiments. In conclusion, the bio-mechanical properties of the GelMA and pectin-based scaffolds as well as the addition of the PTX-cyclodextrin complexes allow for the targeted and efficient delivery of PTX, suppressing the viability of the breast tumor cells via pyroptosis cell death initiation.
Collapse
Affiliation(s)
| | - Andreea Ioana Dinu
- Advanced Polymer Materials Group, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (A.I.D.); (A.L.)
| | - Adriana Lungu
- Advanced Polymer Materials Group, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (A.I.D.); (A.L.)
| | - Hildegard Herman
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310414 Arad, Romania; (H.H.); (C.B.); (A.H.)
| | - Cornel Balta
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310414 Arad, Romania; (H.H.); (C.B.); (A.H.)
| | - Anca Hermenean
- “Aurel Ardelean” Institute of Life Sciences, Vasile Goldis Western University of Arad, 310414 Arad, Romania; (H.H.); (C.B.); (A.H.)
| | - Andreea Iren Șerban
- Department Preclinical Sciences, Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 050097 Bucharest, Romania;
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania;
- Research Institute of the University of Bucharest, 050663 Bucharest, Romania
| |
Collapse
|
49
|
Yang X, Gan Y, Zhang M, Xie S, Lin M, Zhong L, Song M, Wang J, Huang Y. Transcriptome analysis unveils the mechanisms of oxidative stress, immunotoxicity and neurotoxicity induced by benzotriazole UV stabilizer-328 in zebrafish embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117822. [PMID: 39884018 DOI: 10.1016/j.ecoenv.2025.117822] [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: 10/16/2024] [Revised: 01/25/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
As an emerging pollutant, ultraviolet stabilizer-328 (UV-328) has been frequently detected in aquatic environments and attracted great attention. Nevertheless, the toxicity and mechanisms of UV-328 to aquatic organisms are still not fully understood. In particular, the immunotoxicity and neurotoxicity of UV-328 to aquatic organisms and their mechanisms have not been reported yet. In this experiment, the developmental toxicity, oxidative stress, apoptosis, immunotoxicity and neurotoxicity in zebrafish embryos exposed to UV-328 with concentrations of 0.01, 0.1, 1, 10 and 100 µg/L for 120 h were studied. By measuring the growth and developmental indices, production of ROS, enzyme activities, MDA content and expression of genes related to oxidative, immune and nerve, and histopathological analysis, it was found that UV-328 had developmental toxicity to zebrafish larvae, and could induce oxidative stress, immunotoxicity and neurotoxicity to zebrafish larvae even at environmental concentrations with concentration-dependent effects. Moreover, the results of transcriptome analysis and qRT-PCR validation suggested that immune and nerve disorders were caused by UV-328 in zebrafish larvae through regulating the RIG-I-like receptor signaling pathway and neuroactive ligand-receptor interaction, respectively. In addition, transcriptome analysis further revealed that UV-328 could mediate the RIG-I to induce oxidative stress through p38-MAPK/p53 signaling pathway, leading to apoptosis and oxidative damage. In addition, the p38-MAPK signaling pathway enhanced ROS production and activated inflammatory cytokines to induce immunotoxicity. The results of the present work provided important information for understanding the toxicity of UV-328 to aquatic organisms and evaluating its ecological risk in aquatic environment.
Collapse
Affiliation(s)
- Xinlu Yang
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Yijing Gan
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Menghuan Zhang
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Shaolin Xie
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Mingfu Lin
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Lixiang Zhong
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Mengke Song
- College of Natural Resources and Environment, South China Agricultural University, Guangzhou 510642, China
| | - Jun Wang
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China.
| | - Yumei Huang
- University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
50
|
Argirova M, Cherneva E, Mihaylova R, Momekov G, Yancheva D. New metal complexes of 1H-benzimidazole-2-yl hydrazones: Cytostatic, proapoptotic and modulatory activity on kinase signaling pathways. Arch Biochem Biophys 2025; 764:110245. [PMID: 39617120 DOI: 10.1016/j.abb.2024.110245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
The copper complexes of two 1H-benzimidazole-2-yl hydrazones were obtained by complexation with copper chloride. The molecular structure of the complexes was studied by microchemical analysis, SEM-EDX, IR and micro-Raman spectroscopy and DFT calculations. It was found that both ligands form 1:1 complexes with the copper, where the Cu ions are coordinated by N-atom from the benzimidazole ring, N-atom of the azomethine bond, O-atom from the ortho-OH group of the aromatic ring and one chlorine atom. The coordination process significantly affected their cytotoxicity profile. The conversion of 2-(2-hydroxybenzylidene)-1-(1H-benzimidazol-2-yl)hydrazine 1.1. into a Cu complex 2.1. led to a 2.4-fold increase in its antileukemic activity against AR-230 cells and an 8-fold increase in the cytostatic activity against MCF-7 breast cancer cell line. The growth-inhibitory effect of the Cu complex of 2-(2-hydroxy-4-methoxybenzylidene)-1-(1H-benzimidazol-2-yl)hydrazine 2.2. on the MCF-7 cells was comparable to that of the respective ligand, however lacked towards the leukemic AR-230 cell population. Regarding their cytotoxic potential towards CCL-1 cells, both Cu complexes exhibited a weaker selectivity pattern as compared to their ligands. The proapoptotic and modulatory activity of 1.1 and 2.1. on key kinase signaling pathways was further studied in the ER + breast cancer (MCF-7) and bcr-abl + leukemic (AR-230) in vitro tumor models in a comparative manner to the reference drugs tamoxifen and imatinib, respectively. Inhibition of the JAK/STAT signaling pathway was outlined as a prominent mechanism in the antileukemic activity against the Ph + AR-230 in vitro model, whereas recruitment and activation of the extrinsic apoptotic pathway was established in the MCF-7 cells.
Collapse
Affiliation(s)
- Maria Argirova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., build. 9, 1113, Sofia, Bulgaria
| | - Emiliya Cherneva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., build. 9, 1113, Sofia, Bulgaria; Faculty of Pharmacy, Medical University of Sofia, 2 Dunav str., 1000, Sofia, Bulgaria.
| | - Rositsa Mihaylova
- Faculty of Pharmacy, Medical University of Sofia, 2 Dunav str., 1000, Sofia, Bulgaria
| | - Georgi Momekov
- Faculty of Pharmacy, Medical University of Sofia, 2 Dunav str., 1000, Sofia, Bulgaria
| | - Denitsa Yancheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., build. 9, 1113, Sofia, Bulgaria; University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756, Sofia, Bulgaria.
| |
Collapse
|