1
|
Tian B, Wu Y, Du X, Zhang Y. Osteosarcoma stem cells resist chemotherapy by maintaining mitochondrial dynamic stability via DRP1. Int J Mol Med 2025; 55:10. [PMID: 39513621 PMCID: PMC11554380 DOI: 10.3892/ijmm.2024.5451] [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: 07/23/2024] [Accepted: 09/27/2024] [Indexed: 11/15/2024] Open
Abstract
Osteosarcoma malignancy exhibits significant heterogeneity, comprising both osteosarcoma stem cells (OSCs) and non‑OSCs. OSCs demonstrate increased resistance to chemotherapy due to their distinctive cellular and molecular characteristics. Alterations in mitochondrial morphology and homeostasis may enhance chemoresistance by modulating metabolic and regulatory processes. However, the relationship between mitochondrial homeostasis and chemoresistance in OSCs remains to be elucidated. The present study employed high‑resolution microscopy to perform multi‑layered image reconstructions for a quantitative analysis of mitochondrial morphology. The results indicated that OSCs exhibited larger mitochondria in comparison with non‑OSCs. Furthermore, treatment of OSCs with cisplatin (CIS) or doxorubicin (DOX) resulted in preserved mitochondrial morphological stability, which was not observed in non‑OSCs. This finding suggested a potential association between mitochondrial homeostasis and chemoresistance. Further analysis indicated that dynamin‑related protein 1 (DRP1) might play a pivotal role in maintaining the stability of mitochondrial homeostasis in OSCs. Depletion of DRP1 resulted in the disruption of mitochondrial stability when OSCs were treated with CIS or DOX. Additionally, knocking out DRP1 in OSCs led to a reduction in chemoresistance. These findings unveil a novel mechanism underlying chemoresistance in osteosarcoma and suggest that targeting DRP1 could be a promising therapeutic strategy to overcome chemoresistance in OSCs. This provided valuable insights for enhancing treatment outcomes among patients with osteosarcoma.
Collapse
Affiliation(s)
- Boren Tian
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, P.R. China
| | - Yaxuan Wu
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, P.R. China
| | - Xiaoyun Du
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, P.R. China
| | - Yan Zhang
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, P.R. China
| |
Collapse
|
2
|
Mengistu BA, Tsegaw T, Demessie Y, Getnet K, Bitew AB, Kinde MZ, Beirhun AM, Mebratu AS, Mekasha YT, Feleke MG, Fenta MD. Comprehensive review of drug resistance in mammalian cancer stem cells: implications for cancer therapy. Cancer Cell Int 2024; 24:406. [PMID: 39695669 DOI: 10.1186/s12935-024-03558-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cancer remains a significant global challenge, and despite the numerous strategies developed to advance cancer therapy, an effective cure for metastatic cancer remains elusive. A major hurdle in treatment success is the ability of cancer cells, particularly cancer stem cells (CSCs), to resist therapy. These CSCs possess unique abilities, including self-renewal, differentiation, and repair, which drive tumor progression and chemotherapy resistance. The resilience of CSCs is linked to certain signaling pathways. Tumors with pathway-dependent CSCs often develop genetic resistance, whereas those with pathway-independent CSCs undergo epigenetic changes that affect gene regulation. CSCs can evade cytotoxic drugs, radiation, and apoptosis by increasing drug efflux transporter activity and activating survival mechanisms. Future research should prioritize the identification of new biomarkers and signaling molecules to better understand drug resistance. The use of cutting-edge approaches, such as bioinformatics, genomics, proteomics, and nanotechnology, offers potential solutions to this challenge. Key strategies include developing targeted therapies, employing nanocarriers for precise drug delivery, and focusing on CSC-targeted pathways such as the Wnt, Notch, and Hedgehog pathways. Additionally, investigating multitarget inhibitors, immunotherapy, and nanodrug delivery systems is critical for overcoming drug resistance in cancer cells.
Collapse
Affiliation(s)
- Bemrew Admassu Mengistu
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia.
| | - Tirunesh Tsegaw
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yitayew Demessie
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Kalkidan Getnet
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Abebe Belete Bitew
- Department of Veterinary Epidemiology and Public Health, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Mebrie Zemene Kinde
- Department of Biomedical Sciences, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Asnakew Mulaw Beirhun
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Atsede Solomon Mebratu
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Yesuneh Tefera Mekasha
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melaku Getahun Feleke
- Department of Veterinary Pharmacy, College of Veterinary Medicine and Animal Sciences, University of Gondar, Gondar, Ethiopia
| | - Melkie Dagnaw Fenta
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine and Animal Science, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
3
|
Oe S, Kakizaki R, Sakamoto S, Sato T, Hayashi M, Isozaki H, Nonaka M, Iwashita H, Hayashi S, Koike T, Seki-Omura R, Nakano Y, Sato Y, Hirahara Y, Kitada M. MicroRNA-505-5p/-3p Regulates the Proliferation, Invasion, Apoptosis, and Temozolomide Resistance in Mesenchymal Glioma Stem Cells by Targeting AUF1. Mol Carcinog 2024. [PMID: 39513659 DOI: 10.1002/mc.23842] [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: 05/13/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024]
Abstract
Mesenchymal glioma stem cells (MES-GSCs) are a major subtype of GSCs that reside within glioma tissues and contribute to metastasis, therapy resistance, and glioma recurrence. However, the precise molecular mechanisms governing MES-GSC functions remain elusive. Our findings revealed that expression levels of miR-505-5p/-3p are elevated in MES-GSCs compared with those in proneural (PN)-GSCs, glioma cell lines, and normal brain tissue and that miR-505-5p/-3p expression levels are decreased in differentiated MES-GSCs. We assumed that miR-505-5p/-3p would play distinctive roles in MES-GSCs and performed loss-of-function experiments targeting miR-505-5p/-3p. Knockdown of miR-505-5p/-3p increased proliferation and promoted differentiation in MES-GSCs while suppressing invasion, temozolomide resistance, and enhancing apoptosis in MES-GSCs. Mechanistically, miR-505-5p/-3p directly targets the 3' UTR of AUF1, leading to its repression in MES-GSCs. Notably, AUF1 expression levels were significantly lower in MES-GSCs compared with those in PN-GSCs, glioma cell lines, and normal brain tissues. Co-inhibition of AUF1 expression with miR-505-5p/-3p knockdown ameliorated the observed cellular phenotypes caused by miR-505-5p/-3p knockdown in MES-GSCs. These results suggest that miR-505-5p/-3p exerts MES-GSC-specific roles in regulating proliferation, differentiation, invasion, apoptosis, and temozolomide resistance by repressing AUF1 expression.
Collapse
Affiliation(s)
- Souichi Oe
- Department of Anatomy, Kansai Medical University, Osaka, Hirakata, Japan
| | - Rio Kakizaki
- Department of Anatomy, Kansai Medical University, Osaka, Hirakata, Japan
| | - Sumika Sakamoto
- Department of Anatomy, Kansai Medical University, Osaka, Hirakata, Japan
| | - Teruhide Sato
- Department of Anatomy, Kansai Medical University, Osaka, Hirakata, Japan
| | - Mikio Hayashi
- Department of Physiology, Institute of Biomedical Science, Kansai Medical University, Osaka, Hirakata, Japan
| | - Haruna Isozaki
- Department of Neurosurgery, Kansai Medical University, Osaka, Hirakata, Japan
| | - Masahiro Nonaka
- Department of Neurosurgery, Kansai Medical University, Osaka, Hirakata, Japan
| | - Hikaru Iwashita
- Department of Anatomy, Kansai Medical University, Osaka, Hirakata, Japan
| | - Shinichi Hayashi
- Department of Anatomy, Kansai Medical University, Osaka, Hirakata, Japan
| | - Taro Koike
- Department of Anatomy, Kansai Medical University, Osaka, Hirakata, Japan
| | - Ryohei Seki-Omura
- Department of Anatomy, Kansai Medical University, Osaka, Hirakata, Japan
| | - Yousuke Nakano
- Department of Anatomy, Kansai Medical University, Osaka, Hirakata, Japan
| | - Yuki Sato
- Department of Anatomy, Kansai Medical University, Osaka, Hirakata, Japan
| | - Yukie Hirahara
- Department of Anatomy, Kansai Medical University, Osaka, Hirakata, Japan
- Faculty of Nursing, Kansai Medical University, Osaka, Hirakata, Japan
| | - Masaaki Kitada
- Department of Anatomy, Kansai Medical University, Osaka, Hirakata, Japan
| |
Collapse
|
4
|
De Angelis ML, Francescangeli F, Aricò E, Verachi P, Zucchetti M, Matteo C, Petricci E, Pilozzi E, Orienti I, Boe A, Eramo A, Rossi R, Corati T, Macchia D, Pacca AM, Zeuner A, Baiocchi M. A nanoencapsulated oral formulation of fenretinide promotes local and metastatic breast cancer dormancy in HER2/neu transgenic mice. J Exp Clin Cancer Res 2024; 43:296. [PMID: 39497135 PMCID: PMC11536529 DOI: 10.1186/s13046-024-03213-6] [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: 05/14/2024] [Accepted: 10/13/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Prevention and treatment of metastatic breast cancer (BC) is an unmet clinical need. The retinoic acid derivative fenretinide (FeR) was previously evaluated in Phase I-III clinical trials but, despite its excellent tolerability and antitumor activity in preclinical models, showed limited therapeutic efficacy due to poor bioavailability. We recently generated a new micellar formulation of FeR, Bionanofenretinide (Bio-nFeR) showing enhanced bioavailability, low toxicity, and strong antitumor efficacy on human lung cancer, colorectal cancer, and melanoma xenografts. In the present study, we tested the effect of Bio-nFeR on a preclinical model of metastatic BC. METHODS We used BC cell lines for in vitro analyses of cell viability, cell cycle and migratory capacity. For in vivo studies, we used HER2/neu transgenic mice (neuT) as a model of spontaneously metastatic BC. Mice were treated orally with Bio-nFeR and at sacrifice primary and metastatic breast tumors were analyzed by histology and immunohistochemistry. Molecular pathways activated in primary tumors were analyzed by immunoblotting. Stem cell content was assessed by flow cytometry, immunoblotting and functional assays such as colony formation ex vivo and second transplantation assay in immunocompromised mice. RESULTS Bio-nFeR inhibited the proliferation and migration of neuT BC cells in vitro and showed significant efficacy against BC onset in neuT mice. Importantly, Bio-nFeR showed the highest effectiveness against metastatic progression, counteracting both metastasis initiation and expansion. The main mechanism of Bio-nFeR action consists of promoting tumor dormancy through a combined induction of antiproliferative signals and inhibition of the mTOR pathway. CONCLUSION The high effectiveness of Bio-nFeR in the neuT model of mammary carcinogenesis, coupled with its low toxicity, indicates this formulation as a potential candidate for the treatment of metastatic BC and for the adjuvant therapy of BC patients at high risk of developing metastasis.
Collapse
Affiliation(s)
- Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | - Eleonora Aricò
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Paola Verachi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Massimo Zucchetti
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Cristina Matteo
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Elena Petricci
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, Unit of Pathologic Anatomy Morphologic and Molecular, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Isabella Orienti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Alessandra Boe
- Cytometry Unit, Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Adriana Eramo
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Rachele Rossi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Tiberio Corati
- Department of Clinical and Molecular Medicine, Unit of Pathologic Anatomy Morphologic and Molecular, Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Daniele Macchia
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Anna Maria Pacca
- Center of Animal Research and Welfare, Istituto Superiore di Sanità, Rome, Italy
| | - Ann Zeuner
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Marta Baiocchi
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
5
|
Han R, Sun X, Wu Y, Yang YH, Wang QC, Zhang XT, Ding T, Yang JT. Proteomic and Phosphoproteomic Profiling of Matrix Stiffness-Induced Stemness-Dormancy State Transition in Breast Cancer Cells. J Proteome Res 2024; 23:4658-4673. [PMID: 39298182 DOI: 10.1021/acs.jproteome.4c00563] [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] [Indexed: 10/05/2024]
Abstract
The dormancy of cancer stem cells is a major factor leading to drug resistance and a high rate of late recurrence and mortality in estrogen receptor-positive (ER+) breast cancer. Previously, we demonstrated that a stiffer matrix induces tumor cell dormancy and drug resistance, whereas a softened matrix promotes tumor cells to exhibit a stem cell state with high proliferation and migration. In this study, we present a comprehensive analysis of the proteome and phosphoproteome in response to gradient changes in matrix stiffness, elucidating the mechanisms behind cell dormancy-induced drug resistance. Overall, we found that antiapoptotic and membrane transport processes may be involved in the mechanical force-induced dormancy resistance of ER+ breast cancer cells. Our research provides new insights from a holistic proteomic and phosphoproteomic perspective, underscoring the significant role of mechanical forces stemming from the stiffness of the surrounding extracellular matrix as a critical regulatory factor in the tumor microenvironment.
Collapse
Affiliation(s)
- Rong Han
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Xu Sun
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Yue Wu
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Ye-Hong Yang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Qiao-Chu Wang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Xu-Tong Zhang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Tao Ding
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| | - Jun-Tao Yang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College, Beijing 10050, China
| |
Collapse
|
6
|
Khorsandi D, Yang JW, Foster S, Khosravi S, Hoseinzadeh N, Zarei F, Lee YB, Runa F, Gangrade A, Voskanian L, Adnan D, Zhu Y, Wang Z, Jucaud V, Dokmeci MR, Shen X, Bishehsari F, Kelber JA, Khademhosseini A, de Barros NR. Patient-Derived Organoids as Therapy Screening Platforms in Cancer Patients. Adv Healthc Mater 2024; 13:e2302331. [PMID: 38359321 PMCID: PMC11324859 DOI: 10.1002/adhm.202302331] [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: 07/21/2023] [Revised: 11/28/2023] [Indexed: 02/17/2024]
Abstract
Patient-derived organoids (PDOs) developed ex vivo and in vitro are increasingly used for therapeutic screening. They provide a more physiologically relevant model for drug discovery and development compared to traditional cell lines. However, several challenges remain to be addressed to fully realize the potential of PDOs in therapeutic screening. This paper summarizes recent advancements in PDO development and the enhancement of PDO culture models. This is achieved by leveraging materials engineering and microfabrication technologies, including organs-on-a-chip and droplet microfluidics. Additionally, this work discusses the application of PDOs in therapy screening to meet diverse requirements and overcome bottlenecks in cancer treatment. Furthermore, this work introduces tools for data processing and analysis of organoids, along with their microenvironment. These tools aim to achieve enhanced readouts. Finally, this work explores the challenges and future perspectives of using PDOs in drug development and personalized screening for cancer patients.
Collapse
Affiliation(s)
- Danial Khorsandi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Jia-Wei Yang
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Samuel Foster
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Safoora Khosravi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Negar Hoseinzadeh
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Fahimeh Zarei
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Yun Bin Lee
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Farhana Runa
- California State University Northridge, 18111 Nordhoff Street, Northridge, California, USA
| | - Ankit Gangrade
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Leon Voskanian
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Darbaz Adnan
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush Medical College, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Zhaohui Wang
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710 USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Xiling Shen
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Faraz Bishehsari
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush Medical College, Rush University Medical Center, Chicago, IL, 60612, USA
- Division of Digestive Diseases, Rush Center for Integrated Microbiome & Chronobiology Research, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Jonathan A. Kelber
- California State University Northridge, 18111 Nordhoff Street, Northridge, California, USA
- Baylor University, 101 Bagby Ave, Waco, Texas, USA
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| |
Collapse
|
7
|
Baek BS, Park H, Choi JW, Lee EY, Youn JI, Seong SY. Dendritic cells pulsed with penetratin-OLFM4 inhibit the growth and metastasis of melanoma in mice. Biomed Pharmacother 2024; 177:117083. [PMID: 38968793 DOI: 10.1016/j.biopha.2024.117083] [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: 01/11/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024] Open
Abstract
Cancer stem cells (CSCs) can self-renew and differentiate, contributing to tumor heterogeneity, metastasis, and recurrence. Their resistance to therapies, including immunotherapy, underscores the importance of targeting them for complete remission and relapse prevention. Olfactomedin 4 (OLFM4), a marker associated with various cancers such as colorectal cancer, is expressed on CSCs promoting immune evasion and tumorigenesis. However, its potential as a target for CSC-specific immunotherapy remains underexplored. The primary aim of this study is to evaluate the effectiveness of targeting OLFM4 with dendritic cell (DC)-based vaccines in inhibiting tumor growth and metastasis. To improve antigen delivery and immune response, OLFM4 was conjugated with a protein-transduction domain (PTD) from the antennapedia of Drosophila called penetratin, creating a fusion protein (P-OLFM4). The efficacy of DCs pulsed with P-OLFM4 (DCs [P-OLFM4]) was compared to DCs pulsed with OLFM4 (DCs [OLFM4]) and PBS (DCs [PBS]). DCs [P-OLFM4] inhibited tumor growth by 91.2 % and significantly reduced lung metastasis of OLFM4+ melanoma cells by 97 %, compared to the DCs [PBS]. DCs [OLFM4] also demonstrated a reduction in lung metastasis by 59.7 % compared to DCs [PBS]. Immunization with DCs [P-OLFM4] enhanced OLFM4-specific T-cell proliferation, interferon-γ production, and cytotoxic T cell activity in mice. The results indicate that OLFM4 is a viable target for CSC-focused immunotherapy. DC [P-OLFM4] vaccines can elicit robust immune responses, significantly inhibiting tumor growth and metastasis. This strategy holds promise for developing more effective cancer treatments that specifically target CSCs, potentially leading to better patient outcomes by reducing the likelihood of tumor relapse and metastasis.
Collapse
Affiliation(s)
- Bum-Seo Baek
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Gangwon Province, South Korea; Department of Biomedical Sciences, South Korea
| | - Hyunmi Park
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Gangwon Province, South Korea
| | - Ji-Woong Choi
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Gangwon Province, South Korea
| | - Eun-Young Lee
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Gangwon Province, South Korea
| | - Je-In Youn
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Gangwon Province, South Korea; Department of Biomedical Sciences, South Korea
| | - Seung-Yong Seong
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Gangwon Province, South Korea; Department of Biomedical Sciences, South Korea; Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea; Shaperon Inc. Ltd, Seoul, South Korea.
| |
Collapse
|
8
|
Miki K, Oe M, Suzuki K, Miki K, Mu H, Kato Y, Iwatake M, Yukawa H, Baba Y, Ueda Y, Mori Y, Ohe K. Dual-responsive near-infrared turn-on fluorescent probe for cancer stem cell-specific visualization. J Mater Chem B 2024; 12:6959-6967. [PMID: 38913327 DOI: 10.1039/d4tb00897a] [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: 06/25/2024]
Abstract
Aldehyde dehydrogenase 1A1 (ALDH1A1) stands out as one of the most reliable intracellular biomarkers for stem cells because it is expressed in both cancer stem cells (CSCs) and normal somatic stem cells (NSCs). Although several turn-on fluorescent probes for ALDH1A1 have been developed to visualize CSCs in cancer cells, the discrimination of CSCs from NSCs is difficult. We here report an AND-type dual-responsive fluorescent probe, CHO_βgal, the near-infrared fluorescence of which can be turned on after responding to both ALDH1A1 and β-galactosidase. The AND-type dual responsiveness enables CSCs to be clearly visualized, whereas NSCs are non-emissive in microscopy. CSC-positive metastasis model lungs were successfully discriminated from normal lungs in ex vivo staining experiments using CHO_βgal, whereas the single-input ALDH1A1-responsive probe failed to achieve this discrimination owing to pronounced false-positive fluorescence output from lung NSCs. In tissue slice staining experiments, even in the presence of adjacent normal tissues, the peripheral region-specific localization of CSCs was clear. The versatility of CHO_βgal holds promise not only as a fundamental in vitro research tool for visualizing CSCs but also as a valuable asset in practical tissue staining diagnosis, significantly contributing to the assessment of cancer malignancy.
Collapse
Affiliation(s)
- Koji Miki
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| | - Masahiro Oe
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| | - Kanae Suzuki
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| | - Koki Miki
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| | - Huiying Mu
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| | - Yoshimi Kato
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Mayumi Iwatake
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Hiroshi Yukawa
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Institute of Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
- Department of Quantum Life Science, Graduate School of Science, Chiba University, Chiba 265-8522, Japan
| | - Yoshinobu Baba
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
- Institute of Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Yoshifumi Ueda
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Kouichi Ohe
- Department of Energy and Hydrocarbon Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan.
| |
Collapse
|
9
|
Pan T, Xie DK, Li J, Qiang YJ, Fan SY, Wang TT, Han YY, Zang J, Yang Y, Zhao JL, Li SZ, Wu S. Glioma-Stem-Cell-Derived Exosomes Remodeled Glioma-Associated Macrophage via NEAT1/miR-125a/STAT3 Pathway. Cancers (Basel) 2024; 16:2500. [PMID: 39061140 PMCID: PMC11274466 DOI: 10.3390/cancers16142500] [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: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 07/28/2024] Open
Abstract
Glioblastoma (GBM), as the most common primary brain tumor, usually results in an extremely poor prognosis, in which glioma stem cells (GSCs) and their immunosuppressive microenvironment prominently intervene in the resistance to radiotherapy and chemotherapy that directly leads to tumor recurrence and shortened survival time. The specific mechanism through which exosomes generated from GSCs support the creation of an immunosuppressive microenvironment remains unknown, while it is acknowledged to be engaged in intercellular communication and the regulation of the glioma immunosuppressive microenvironment. The elevated expression of LncRNA-NEAT1 was found in glioma cells after radiotherapy, chemotherapy, and DNA damage stimulation, and NEAT1 could promote the malignant biological activities of GSCs. Emerging evidence suggests that lncRNAs may reply to external stimuli or DNA damage by playing a role in modulating different aspects of tumor biology. Our study demonstrated a promotive role of the carried NEAT1 by GSC-derived exosomes in the polarization of M2-like macrophages. Further experiments demonstrated the mediative role of miR-125a and its target gene STAT3 in NEAT1-induced polarization of M2-like macrophages that promote glioma progression. Our findings elucidate the mechanism by which GSCs influence the polarization of M2-like macrophages through exosomes, which may contribute to the formation of immunosuppressive microenvironments. Taken together, our study reveals the miR-125a-STAT3 pathway through which exosomal NEAT1 from treatment-resistant GSCs contributes to M2-like macrophage polarization, indicating the potential of exosomal NEAT1 for treating glioma.
Collapse
Affiliation(s)
- Tong Pan
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi’an 710032, China
| | - Dong-Kun Xie
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an 710069, China; (D.-K.X.); (Y.Y.)
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Air Force Medical University, Xi’an 710032, China;
| | - Juan Li
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
| | - Yu-Jie Qiang
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
| | - Song-Yuan Fan
- Department of Neurosurgery, The Air Force Hospital of Central Theater of PLA, Datong 037000, China;
| | - Ting-Ting Wang
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
| | - Yuan-Yuan Han
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
| | - Jian Zang
- Department of Radiotherapy, Xijing Hospital, Air Force Medical University, Xi’an 710032, China;
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an 710069, China; (D.-K.X.); (Y.Y.)
| | - Jun-Long Zhao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Air Force Medical University, Xi’an 710032, China;
| | - San-Zhong Li
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
| | - Shuang Wu
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an 710032, China; (T.P.); (J.L.); (Y.-J.Q.); (T.-T.W.); (Y.-Y.H.)
- Department of Neurosurgery, The Air Force Hospital of Central Theater of PLA, Datong 037000, China;
| |
Collapse
|
10
|
Fiorentino F, Fabbrizi E, Raucci A, Noce B, Fioravanti R, Valente S, Paolini C, De Maria R, Steinkühler C, Gallinari P, Rotili D, Mai A. Uracil- and Pyridine-Containing HDAC Inhibitors Displayed Cytotoxicity in Colorectal and Glioblastoma Cancer Stem Cells. ChemMedChem 2024; 19:e202300655. [PMID: 38529661 DOI: 10.1002/cmdc.202300655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 03/01/2024] [Accepted: 03/25/2024] [Indexed: 03/27/2024]
Abstract
Cancer stem cells (CSCs) are a niche of highly tumorigenic cells featuring self-renewal, activation of pluripotency genes, multidrug resistance, and ability to cause cancer relapse. Seven HDACi (1-7), showing either hydroxamate or 2'-aminoanilide function, were tested in colorectal cancer (CRC) and glioblastoma multiforme (GBM) CSCs to determine their effects on cell proliferation, H3 acetylation levels and in-cell HDAC activity. Two uracil-based hydroxamates, 5 and 6, which differ in substitution at C5 and C6 positions of the pyrimidine ring, exhibited the greatest cytotoxicity in GBM (5) and CRC (6) CSCs, followed by the pyridine-hydroxamate 2, with 2- to 6-fold higher potency than the positive control SAHA. Finally, increased H3 acetylation as well as HDAC inhibition directly in cells by selected 2'-aminoanilide 4 and hydroxamate 5 confirmed target engagement. Further investigation will be conducted into the broad-spectrum anticancer properties of the most potent derivatives and their effects in combination with approved, conventional anticancer drugs.
Collapse
Affiliation(s)
- Francesco Fiorentino
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo, Moro n. 5, 00185, Rome, Italy
| | - Emanuele Fabbrizi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo, Moro n. 5, 00185, Rome, Italy
| | - Alessia Raucci
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo, Moro n. 5, 00185, Rome, Italy
| | - Beatrice Noce
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo, Moro n. 5, 00185, Rome, Italy
| | - Rossella Fioravanti
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo, Moro n. 5, 00185, Rome, Italy
| | - Sergio Valente
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo, Moro n. 5, 00185, Rome, Italy
| | - Chantal Paolini
- IRBM S.p.A., Via Pontina km 30.600, 00071, Pomezia, Rome, Italy
| | - Ruggero De Maria
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Rome, Italy
| | - Christian Steinkühler
- Research and Development, Italfarmaco Group, Via dei Lavoratori 54, 20092, Cinisello Balsamo, Italy
| | - Paola Gallinari
- Exiris S.r.l., Tecnopolo Castel, Romano, Via Castel Romano 100, 00128, Rome, Italy
| | - Dante Rotili
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo, Moro n. 5, 00185, Rome, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le Aldo, Moro n. 5, 00185, Rome, Italy
- Pasteur Institute, Cenci-Bolognetti Foundation, Sapienza University of Rome, P. de Aldo Moro n. 5, 00185, Rome, Italy
| |
Collapse
|
11
|
Bhattacharya A, Wang K, Penailillo J, Chan CN, Fushimi A, Yamashita N, Daimon T, Haratake N, Ozawa H, Nakashoji A, Shigeta K, Morimoto Y, Miyo M, Kufe DW. MUC1-C regulates NEAT1 lncRNA expression and paraspeckle formation in cancer progression. Oncogene 2024; 43:2199-2214. [PMID: 38802648 PMCID: PMC11226401 DOI: 10.1038/s41388-024-03068-3] [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: 12/06/2023] [Revised: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024]
Abstract
The MUC1 gene evolved in mammals for adaptation of barrier tissues in response to infections and damage. Paraspeckles are nuclear bodies formed on the NEAT1 lncRNA in response to loss of homeostasis. There is no known intersection of MUC1 with NEAT1 or paraspeckles. Here, we demonstrate that the MUC1-C subunit plays an essential role in regulating NEAT1 expression. MUC1-C activates the NEAT1 gene with induction of the NEAT1_1 and NEAT1_2 isoforms by NF-κB- and MYC-mediated mechanisms. MUC1-C/MYC signaling also induces expression of the SFPQ, NONO and FUS RNA binding proteins (RBPs) that associate with NEAT1_2 and are necessary for paraspeckle formation. MUC1-C integrates activation of NEAT1 and RBP-encoding genes by recruiting the PBAF chromatin remodeling complex and increasing chromatin accessibility of their respective regulatory regions. We further demonstrate that MUC1-C and NEAT1 form an auto-inductive pathway that drives common sets of genes conferring responses to inflammation and loss of homeostasis. Of functional significance, we find that the MUC1-C/NEAT1 pathway is of importance for the cancer stem cell (CSC) state and anti-cancer drug resistance. These findings identify a previously unrecognized role for MUC1-C in the regulation of NEAT1, RBPs, and paraspeckles that has been co-opted in promoting cancer progression.
Collapse
Affiliation(s)
| | - Keyi Wang
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Johany Penailillo
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Chi Ngai Chan
- Tissue Technologies Unit, Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Atsushi Fushimi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Nami Yamashita
- Breast Surgical Oncology, Breast Oncology Center, The Cancer Institute Hospital of the JFCR, Tokyo, Japan
| | - Tatsuaki Daimon
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Naoki Haratake
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Hiroki Ozawa
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ayako Nakashoji
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Keisuke Shigeta
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yoshihiro Morimoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Masaaki Miyo
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Donald W Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Waqar MA, Zaman M, Khan R, Shafeeq Ur Rahman M, Majeed I. Navigating the tumor microenvironment: mesenchymal stem cell-mediated delivery of anticancer agents. J Drug Target 2024; 32:624-634. [PMID: 38652480 DOI: 10.1080/1061186x.2024.2347356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/21/2024] [Indexed: 04/25/2024]
Abstract
Scientific knowledge of cancer has advanced greatly throughout the years, with most recent studies findings includes many hallmarks that capture disease's multifaceted character. One of the novel approach utilised for the delivery of anti-cancer agents includes mesenchymal stem cell mediated drug delivery. Mesenchymal stem cells (MSCs) are non-haematopoietic progenitor cells that may be extracted from bone marrow, tooth pulp, adipose tissue and placenta/umbilical cord blood dealing with adult stem cells. MSCs are mostly involved in regeneration of tissue, they have also been shown to preferentially migrate to location of several types of tumour in-vivo. Usage of MSCs ought to improve both effectiveness and safety of anti-cancer drugs by enhancing delivery efficiency of anti-cancer therapies to tumour site. Numerous researches has demonstrated that various drugs, when delivered via mesenchymal stem cell mediated delivery can elicit anti-tumour effect of cells in cancers of breast cells and thyroid cells. MSCs have minimal immunogenicity because to lack of co-stimulatory molecule expression, which means there is no requirement for immunosuppression after allogenic transplantation. This current review elaborates recent advancements of mesenchyma stem cell mediated drug delivery of anti-cancer agents along with its mechanism and previously reported studies of drugs manufactured via this drug delivery system.
Collapse
Affiliation(s)
- Muhammad Ahsan Waqar
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Lahore University of Biological & Applied Sciences, Lahore, Pakistan
| | - Muhammad Zaman
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Rabeel Khan
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Lahore University of Biological & Applied Sciences, Lahore, Pakistan
| | | | - Imtiaz Majeed
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| |
Collapse
|
13
|
Du J, Qin H. Lipid metabolism dynamics in cancer stem cells: potential targets for cancers. Front Pharmacol 2024; 15:1367981. [PMID: 38994204 PMCID: PMC11236562 DOI: 10.3389/fphar.2024.1367981] [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: 01/09/2024] [Accepted: 06/10/2024] [Indexed: 07/13/2024] Open
Abstract
Cancer stem cells (CSCs) represent a small subset of heterogeneous cells within tumors that possess the ability to self-renew and initiate tumorigenesis. They serve as potential drivers for tumor initiation, metastasis, recurrence, and drug resistance. Recent research has demonstrated that the stemness preservation of CSCs is heavily reliant on their unique lipid metabolism alterations, enabling them to maintain their own environmental homeostasis through various mechanisms. The primary objectives involve augmenting intracellular fatty acid (FA) content to bolster energy supply, promoting β-oxidation of FA to optimize energy utilization, and elevating the mevalonate (MVA) pathway for efficient cholesterol synthesis. Additionally, lipid droplets (LDs) can serve as alternative energy sources in the presence of glycolysis blockade in CSCs, thereby safeguarding FA from peroxidation. Furthermore, the interplay between autophagy and lipid metabolism facilitates rapid adaptation of CSCs to the harsh microenvironment induced by chemotherapy. In this review, we comprehensively review recent studies pertaining to lipid metabolism in CSCs and provide a concise overview of the indispensable role played by LDs, FA, cholesterol metabolism, and autophagy in maintaining the stemness of CSCs.
Collapse
Affiliation(s)
- Juan Du
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Hai Qin
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, China
| |
Collapse
|
14
|
Nakashoji A, Haratake N, Bhattacharya A, Mao W, Xu K, Wang K, Daimon T, Ozawa H, Shigeta K, Fushimi A, Yamashita N, Morimoto Y, Shimokawa M, Saito S, Egloff AM, Uppaluri R, Long MD, Kufe D. Identification of MUC1-C as a Target for Suppressing Progression of Head and Neck Squamous Cell Carcinomas. CANCER RESEARCH COMMUNICATIONS 2024; 4:1268-1281. [PMID: 38619287 PMCID: PMC11092937 DOI: 10.1158/2767-9764.crc-24-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/27/2024] [Accepted: 04/02/2024] [Indexed: 04/16/2024]
Abstract
The MUC1-C protein is aberrantly expressed in adenocarcinomas of epithelial barrier tissues and contributes to their progression. Less is known about involvement of MUC1-C in the pathogenesis of squamous cell carcinomas (SCC). Here, we report that the MUC1 gene is upregulated in advanced head and neck SCCs (HNSCC). Studies of HNSCC cell lines demonstrate that the MUC1-C subunit regulates expression of (i) RIG-I and MDA5 pattern recognition receptors, (ii) STAT1 and IFN regulatory factors, and (iii) downstream IFN-stimulated genes. MUC1-C integrates chronic activation of the STAT1 inflammatory pathway with induction of the ∆Np63 and SOX2 genes that are aberrantly expressed in HNSCCs. In extending those dependencies, we demonstrate that MUC1-C is necessary for NOTCH3 expression, self-renewal capacity, and tumorigenicity. The findings that MUC1 associates with ∆Np63, SOX2 and NOTCH3 expression by single-cell RNA sequencing analysis further indicate that MUC1-C drives the HNSCC stem cell state and is a target for suppressing HNSCC progression. SIGNIFICANCE This work reports a previously unrecognized role for MUC1-C in driving STAT1-mediated chronic inflammation with the progression of HNSCC and identifies MUC1-C as a druggable target for advanced HNSCC treatment.
Collapse
Affiliation(s)
- Ayako Nakashoji
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Naoki Haratake
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - Weipu Mao
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kangjie Xu
- Central Laboratory Department, Binhai County People's Hospital, Yancheng, P.R. China
| | - Keyi Wang
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Tatsuaki Daimon
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Hiroki Ozawa
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Keisuke Shigeta
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Atsushi Fushimi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Nami Yamashita
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yoshihiro Morimoto
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Mototsugu Shimokawa
- Department of Biostatistics, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Shin Saito
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ann Marie Egloff
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ravindra Uppaluri
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Mark D. Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Fatma H, Siddique HR. Cancer cell plasticity, stem cell factors, and therapy resistance: how are they linked? Cancer Metastasis Rev 2024; 43:423-440. [PMID: 37796391 DOI: 10.1007/s10555-023-10144-9] [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: 09/06/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023]
Abstract
Cellular plasticity can occur naturally in an organism and is considered an adapting mechanism during the developmental stage. However, abnormal cellular plasticity is observed in different diseased conditions, including cancer. Cancer cell plasticity triggers the stimuli of epithelial-mesenchymal transition (EMT), abnormal epigenetic changes, expression of stem cell factors and implicated signaling pathways, etc., and helps in the maintenance of CSC phenotype. Conversely, CSC maintains the cancer cell plasticity, EMT, and epigenetic plasticity. EMT contributes to increased cell migration and greater diversity within tumors, while epigenetic changes, stem cell factors (OCT4, NANOG, and SOX2), and various signaling pathways allow cancer cells to maintain various phenotypes, giving rise to intra- and inter-tumoral heterogeneity. The intricate relationships between cancer cell plasticity and stem cell factors help the tumor cells adopt drug-tolerant states, evade senescence, and successfully acquire drug resistance with treatment dismissal. Inhibiting molecules/signaling pathways involved in promoting CSCs, cellular plasticity, EMT, and epigenetic plasticity might be helpful for successful cancer therapy management. This review discussed the role of cellular plasticity, EMT, and stem cell factors in tumor initiation, progression, reprogramming, and therapy resistance. Finally, we discussed how the intervention in this axis will help better manage cancers and improve patient survivability.
Collapse
Affiliation(s)
- Homa Fatma
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, UP, 202002, India
| | - Hifzur R Siddique
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, UP, 202002, India.
| |
Collapse
|
16
|
Kim R, Kin T, Beck WT. Impact of Complex Apoptotic Signaling Pathways on Cancer Cell Sensitivity to Therapy. Cancers (Basel) 2024; 16:984. [PMID: 38473345 DOI: 10.3390/cancers16050984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Anticancer drugs induce apoptotic and non-apoptotic cell death in various cancer types. The signaling pathways for anticancer drug-induced apoptotic cell death have been shown to differ between drug-sensitive and drug-resistant cells. In atypical multidrug-resistant leukemia cells, the c-Jun/activator protein 1 (AP-1)/p53 signaling pathway leading to apoptotic death is altered. Cancer cells treated with anticancer drugs undergo c-Jun/AP-1-mediated apoptotic death and are involved in c-Jun N-terminal kinase activation and growth arrest- and DNA damage-inducible gene 153 (Gadd153)/CCAAT/enhancer-binding protein homologous protein pathway induction, regardless of the p53 genotype. Gadd153 induction is associated with mitochondrial membrane permeabilization after anticancer drug treatment and involves a coupled endoplasmic reticulum stress response. The induction of apoptosis by anticancer drugs is mediated by the intrinsic pathway (cytochrome c, Cyt c) and subsequent activation of the caspase cascade via proapoptotic genes (e.g., Bax and Bcl-xS) and their interactions. Anticancer drug-induced apoptosis involves caspase-dependent and caspase-independent pathways and occurs via intrinsic and extrinsic pathways. The targeting of antiapoptotic genes such as Bcl-2 enhances anticancer drug efficacy. The modulation of apoptotic signaling by Bcl-xS transduction increases the sensitivity of multidrug resistance-related protein-overexpressing epidermoid carcinoma cells to anticancer drugs. The significance of autophagy in cancer therapy remains to be elucidated. In this review, we summarize current knowledge of cancer cell death-related signaling pathways and their alterations during anticancer drug treatment and discuss potential strategies to enhance treatment efficacy.
Collapse
Affiliation(s)
- Ryungsa Kim
- Department of Breast Surgery, Hiroshima Mark Clinic, 1-4-3F, 2-Chome Ohte-machi, Naka-ku, Hiroshima 730-0051, Japan
| | - Takanori Kin
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - William T Beck
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
17
|
Saggese P, Pandey A, Alcaraz M, Fung E, Hall A, Yanagawa J, Rodriguez EF, Grogan TR, Giurato G, Nassa G, Salvati A, Shirihai OS, Weisz A, Dubinett SM, Scafoglio C. Glucose Deprivation Promotes Pseudohypoxia and Dedifferentiation in Lung Adenocarcinoma. Cancer Res 2024; 84:305-327. [PMID: 37934116 PMCID: PMC10790128 DOI: 10.1158/0008-5472.can-23-1148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/12/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
Increased utilization of glucose is a hallmark of cancer. Sodium-glucose transporter 2 (SGLT2) is a critical player in glucose uptake in early-stage and well-differentiated lung adenocarcinoma (LUAD). SGLT2 inhibitors, which are FDA approved for diabetes, heart failure, and kidney disease, have been shown to significantly delay LUAD development and prolong survival in murine models and in retrospective studies in diabetic patients, suggesting that they may be repurposed for lung cancer. Despite the antitumor effects of SGLT2 inhibition, tumors eventually escape treatment. Here, we studied the mechanisms of resistance to glucose metabolism-targeting treatments. Glucose restriction in LUAD and other tumors induced cancer cell dedifferentiation, leading to a more aggressive phenotype. Glucose deprivation caused a reduction in alpha-ketoglutarate (αKG), leading to attenuated activity of αKG-dependent histone demethylases and histone hypermethylation. The dedifferentiated phenotype depended on unbalanced EZH2 activity that suppressed prolyl-hydroxylase PHD3 and increased expression of hypoxia-inducible factor 1α (HIF1α), triggering epithelial-to-mesenchymal transition. Finally, a HIF1α-dependent transcriptional signature of genes upregulated by low glucose correlated with prognosis in human LUAD. Overall, this study furthers current knowledge of the relationship between glucose metabolism and cell differentiation in cancer, characterizing the epigenetic adaptation of cancer cells to glucose deprivation and identifying targets to prevent the development of resistance to therapies targeting glucose metabolism. SIGNIFICANCE Epigenetic adaptation allows cancer cells to overcome the tumor-suppressive effects of glucose restriction by inducing dedifferentiation and an aggressive phenotype, which could help design better metabolic treatments.
Collapse
Affiliation(s)
- Pasquale Saggese
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Aparamita Pandey
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Martín Alcaraz
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Eileen Fung
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
- Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Abbie Hall
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Jane Yanagawa
- Department of Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Erika F. Rodriguez
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Tristan R. Grogan
- Division of General Internal Medicine and Health Services Research, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana,’ University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health – CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana,’ University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health – CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
| | - Annamaria Salvati
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana,’ University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health – CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
- Medical Genomics Program and Division of Onco-Hematology, AOU “S. Giovanni di Dio e Ruggi d'Aragona,” University of Salerno, Salerno, Italy
| | - Orian S. Shirihai
- Department of Medicine (Endocrinology), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana,’ University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health – CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
- Medical Genomics Program and Division of Onco-Hematology, AOU “S. Giovanni di Dio e Ruggi d'Aragona,” University of Salerno, Salerno, Italy
| | - Steven M. Dubinett
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Claudio Scafoglio
- Department of Medicine (Pulmonary, Critical Care, and Sleep Medicine), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
18
|
Monti M, Benerini Gatta L, Bugatti M, Pezzali I, Picinoli S, Manfredi M, Lavazza A, Vanella VV, De Giorgis V, Zanatta L, Missale F, Lonardi S, Zanetti B, Bozzoni G, Cadei M, Abate A, Vergani B, Balzarini P, Battocchio S, Facco C, Turri-Zanoni M, Castelnuovo P, Nicolai P, Fonsatti E, Leone BE, Marengo E, Sigala S, Ronca R, Perego M, Lombardi D, Vermi W. Novel cellular systems unveil mucosal melanoma initiating cells and a role for PI3K/Akt/mTOR pathway in mucosal melanoma fitness. J Transl Med 2024; 22:35. [PMID: 38191367 PMCID: PMC10775657 DOI: 10.1186/s12967-023-04784-2] [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: 07/11/2023] [Accepted: 12/04/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Mucosal Melanomas (MM) are highly aggressive neoplasms arising from mucosal melanocytes. Current treatments offer a limited survival benefit for patients with advanced MM; moreover, the lack of pre-clinical cellular systems has significantly limited the understanding of their immunobiology. METHODS Five novel cell lines were obtained from patient-derived biopsies of MM arising in the sino-nasal mucosa and designated as SN-MM1-5. The morphology, ultrastructure and melanocytic identity of SN-MM cell lines were validated by transmission electron microscopy and immunohistochemistry. Moreover, in vivo tumorigenicity of SN-MM1-5 was tested by subcutaneous injection in NOD/SCID mice. Molecular characterization of SN-MM cell lines was performed by a mass-spectrometry proteomic approach, and their sensitivity to PI3K chemical inhibitor LY294002 was validated by Akt activation, measured by pAkt(Ser473) and pAkt(Thr308) in immunoblots, and MTS assay. RESULTS This study reports the validation and functional characterization of five newly generated SN-MM cell lines. Compared to the normal counterpart, the proteomic profile of SN-MM is consistent with transformed melanocytes showing a heterogeneous degree of melanocytic differentiation and activation of cancer-related pathways. All SN-MM cell lines resulted tumorigenic in vivo and display recurrent structural variants according to aCGH analysis. Of relevance, the microscopic analysis of the corresponding xenotransplants allowed the identification of clusters of MITF-/CDH1-/CDH2 + /ZEB1 + /CD271 + cells, supporting the existence of melanoma-initiating cells also in MM, as confirmed in clinical samples. In vitro, SN-MM cell lines were sensitive to cisplatin, but not to temozolomide. Moreover, the proteomic analysis of SN-MM cell lines revealed that RICTOR, a subunit of mTORC2 complex, is the most significantly activated upstream regulator, suggesting a relevant role for the PI3K-Akt-mTOR pathway in these neoplasms. Consistently, phosphorylation of NDRG1 and Akt activation was observed in SN-MM, the latter being constitutive and sustained by PTEN loss in SN-MM2 and SN-MM3. The cell viability impairment induced by LY294002 confirmed a functional role for the PI3K-Akt-mTOR pathway in SN-MM cell lines. CONCLUSIONS Overall, these novel and unique cellular systems represent relevant experimental tools for a better understanding of the biology of these neoplasms and, as an extension, to MM from other sites.
Collapse
Affiliation(s)
- Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luisa Benerini Gatta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Histocompatibility Laboratory "Vittorio Mero", Department of Transfusion Medicine, ASST Spedali Civili Di Brescia, Brescia, Italy
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Irene Pezzali
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sara Picinoli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marcello Manfredi
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Center for Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Antonio Lavazza
- Istituto Zooprofilattico Sperimentale Della Lombardia E Dell'Emilia-Romagna "Bruno Ubertini", Brescia, Italy
| | - Virginia Vita Vanella
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Center for Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Veronica De Giorgis
- Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
- Center for Autoimmune and Allergic Diseases, University of Piemonte Orientale, Novara, Italy
| | - Lucia Zanatta
- Department of Pathology, Treviso Regional Hospital, Treviso, Italy
| | - Francesco Missale
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Department of Head & Neck Oncology & Surgery Otorhinolaryngology, Antoni Van Leeuwenhoek, Nederlands Kanker Instituut, Amsterdam, The Netherlands
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Benedetta Zanetti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giovanni Bozzoni
- Istituto Zooprofilattico Sperimentale Della Lombardia E Dell'Emilia-Romagna "Bruno Ubertini", Brescia, Italy
| | - Moris Cadei
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Andrea Abate
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Barbara Vergani
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Piera Balzarini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Simonetta Battocchio
- Unit of Pathology, Department of Molecular and Translational Medicine, University of Brescia-"ASST Spedali Civili Di Brescia", Brescia, Italy
| | - Carla Facco
- Unit of Pathology, Department of Medicine and Surgery, ASST Sette-Laghi, University of Insubria, Varese, Italy
| | - Mario Turri-Zanoni
- Unit of Otorhinolaryngology and Head & Neck Surgery, Department of Biotechnology and Life Sciences, ASST Sette Laghi, University of Insubria, Varese, Italy
| | - Paolo Castelnuovo
- Unit of Otorhinolaryngology and Head & Neck Surgery, Department of Biotechnology and Life Sciences, ASST Sette Laghi, University of Insubria, Varese, Italy
| | - Piero Nicolai
- Section of Otorhinolaryngology-Head and Neck Surgery, Department of Neurosciences, University of Padova, Padova, Italy
| | - Ester Fonsatti
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | | | - Emilio Marengo
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy
| | - Sandra Sigala
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Davide Lombardi
- Unit of Otorhinolaryngology - Head and Neck Surgery, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, P.Le Spedali Civili 1, 25123, Brescia, Italy.
| |
Collapse
|
19
|
Daimon T, Bhattacharya A, Wang K, Haratake N, Nakashoji A, Ozawa H, Morimoto Y, Yamashita N, Kosaka T, Oya M, Kufe DW. MUC1-C is a target of salinomycin in inducing ferroptosis of cancer stem cells. Cell Death Discov 2024; 10:9. [PMID: 38182558 PMCID: PMC10770371 DOI: 10.1038/s41420-023-01772-9] [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: 05/24/2023] [Revised: 11/22/2023] [Accepted: 12/11/2023] [Indexed: 01/07/2024] Open
Abstract
The oncogenic MUC1-C transmembrane protein is a critical effector of the cancer stem cell (CSC) state. Addiction to MUC1-C for self-renewal in the progression of human cancers has emphasized the need for development of anti-MUC1-C agents. However, there are presently no approved small molecules for targeting MUC1-C-dependent CSCs. In screening for small molecules, we identified salinomycin (SAL), an inducer of ferroptosis, as a potent inhibitor of MUC1-C signaling. We demonstrate that SAL suppresses MUC1-C expression by disrupting a NF-κB/MUC1-C auto-inductive circuit that is necessary for ferroptosis resistance. Our results show that SAL-induced MUC1-C suppression downregulates a MUC1-C→MYC pathway that activates genes encoding (i) glutathione-disulfide reductase (GSR), and (ii) the LDL receptor related protein 8 (LRP8), which inhibit ferroptosis by generating GSH and regulating selenium levels, respectively. GSR and LRP8 contribute to the function of glutathione peroxidase 4 (GPX4), an essential negative regulator of ferroptotic cell death. We demonstrate that targeting MUC1-C genetically or with the GO-203 peptide inhibitor suppresses GPX4 expression and GPX activity in association with the induction of ferroptosis. Studies of CSCs enriched by serial passage as tumorspheres further demonstrate that the effects of SAL are mediated by downregulation of MUC1-C and thereby overcoming resistance to ferroptosis. As confirmation of these results, rescue of MUC1-C downregulation with the MUC1-C cytoplasmic domain (i) reversed the suppression of GSR, LRP8 and GPX4 expression, and (ii) attenuated the induction of ferroptosis. These findings identify SAL as a unique small molecule inhibitor of MUC1-C signaling and demonstrate that MUC1-C is an important effector of resistance to ferroptosis.
Collapse
Affiliation(s)
- Tatsuaki Daimon
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Keyi Wang
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Naoki Haratake
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ayako Nakashoji
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Hiroki Ozawa
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yoshihiro Morimoto
- Department of Gastroenterological Surgery, Kinan Hospital, Wakayama, Japan
| | - Nami Yamashita
- Breast Surgical Oncology, Breast Oncology Center, The Cancer Institute Hospital of the JFCR, Tokyo, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Donald W Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Maleki EH, Bahrami AR, Matin MM. Cancer cell cycle heterogeneity as a critical determinant of therapeutic resistance. Genes Dis 2024; 11:189-204. [PMID: 37588236 PMCID: PMC10425754 DOI: 10.1016/j.gendis.2022.11.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/20/2022] [Accepted: 11/16/2022] [Indexed: 01/15/2023] Open
Abstract
Intra-tumor heterogeneity is now arguably one of the most-studied topics in tumor biology, as it represents a major obstacle to effective cancer treatment. Since tumor cells are highly diverse at genetic, epigenetic, and phenotypic levels, intra-tumor heterogeneity can be assumed as an important contributing factor to the nullification of chemotherapeutic effects, and recurrence of the tumor. Based on the role of heterogeneous subpopulations of cancer cells with varying cell-cycle dynamics and behavior during cancer progression and treatment; herein, we aim to establish a comprehensive definition for adaptation of neoplastic cells against therapy. We discuss two parallel and yet distinct subpopulations of tumor cells that play pivotal roles in reducing the effects of chemotherapy: "resistant" and "tolerant" populations. Furthermore, this review also highlights the impact of the quiescent phase of the cell cycle as a survival mechanism for cancer cells. Beyond understanding the mechanisms underlying the quiescence, it provides an insightful perspective on cancer stem cells (CSCs) and their dual and intertwined functions based on their cell cycle state in response to treatment. Moreover, CSCs, epithelial-mesenchymal transformed cells, circulating tumor cells (CTCs), and disseminated tumor cells (DTCs), which are mostly in a quiescent state of the cell cycle are proved to have multiple biological links and can be implicated in our viewpoint of cell cycle heterogeneity in tumors. Overall, increasing our knowledge of cell cycle heterogeneity is a key to identifying new therapeutic solutions, and this emerging concept may provide us with new opportunities to prevent the dreadful cancer recurrence.
Collapse
Affiliation(s)
- Ebrahim H. Maleki
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, 31-007 Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348 Krakow, Poland
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
| | - Maryam M. Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, 9177948974 Mashhad, Iran
- Stem Cell and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch, 917751376 Mashhad, Iran
| |
Collapse
|
21
|
Zhong C, Wang G, Guo M, Zhu N, Chen X, Yan Y, Li N, Yu W. The Role of Tumor Stem Cells in Colorectal Cancer Drug Resistance. Cancer Control 2024; 31:10732748241274196. [PMID: 39215442 PMCID: PMC11367616 DOI: 10.1177/10732748241274196] [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: 02/24/2024] [Revised: 07/09/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Background: Colorectal cancer is a major cause of mortality among the prevalent malignant tumors of the gastrointestinal tract. Although chemotherapy is a standard treatment for colorectal cancer, its efficacy is limited by chemoresistance. Recent studies have investigated targeting tumor stem cells as a potential new therapeutic approach for addressing chemoresistance in colorectal cancer. Colorectal cancer frequently relapses, with tumor stem cells often representing one of the leading causes of treatment failure. Purpose: Understanding drug resistance in colorectal cancer stem cells is crucial for improving treatment outcomes. By focusing on developing targeted therapies that specifically address drug resistance in colorectal cancer stem cells, there is potential to make significant advancements in the treatment of colorectal cancer.This approach may lead to more effective and lasting outcomes in patients battling colorectal cancer. Research Design: In this review, a comprehensive overview of recent research on colorectal cancer stem cell treatment resistance is presented.Results: Elucidating the key underlying mechanisms. This review also highlights the potential benefits of targeted therapies in overcoming colorectal cancer resistance to treatment. Conclusions: CCSCs are key players in drug resistance of CRC, indicating their potential as targets for effective therapy. Elucidating their role in this process could aid in discovering tailored treatment strategies.The significance of signaling pathways, TME, and miRNA in regulating drug resistance in CCSCs is been highlighted.
Collapse
Affiliation(s)
- Chen Zhong
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Guojuan Wang
- Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Min Guo
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Naicheng Zhu
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Xiudan Chen
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Yuwei Yan
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Nanxin Li
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Wenyan Yu
- Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
22
|
Agüero EI, Belgorosky D, García-Silva JI, Booth R, Lerner B, Pérez MS, Eiján AM. Microdevices for cancer stem cell culture as a predictive chemotherapeutic response platform. J Mol Med (Berl) 2023; 101:1465-1475. [PMID: 37755493 DOI: 10.1007/s00109-023-02375-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 08/20/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023]
Abstract
Microfluidic platforms for clinical use are a promising translational strategy for cancer research specially for drug screening. Identifying cancer stem cells (CSC) using sphere culture techniques in microfluidic devices (MDs) showed to be better reproducing physiological responses than other in vitro models and allow the optimization of samples and reagents. We evaluated individual sphere proliferation and stemness toward chemotherapeutic treatment (CT) with doxorubicin and cisplatin in bladder cancer cell lines (MB49-I and J82) cultured in MDs used as CSC treatment response platform. Our results confirm the usefulness of this device to evaluate the CT effect in sphere-forming efficiency, size, and growth rate from individual spheres within MDs and robust information comparable to conventional culture plates was obtained. The expression of pluripotency genetic markers (Oct4, Sox2, Nanog, and CD44) could be analyzed by qPCR and immunofluorescence in spheres growing directly in MDs. MDs are a suitable platform for sphere isolation from tumor samples and can provide information about CT response. Microfluidic-based CSC studies could provide information about treatment response of cancer patients from small samples and can be a promising tool for CSC-targeted specific treatment with potential in precision medicine. KEY MESSAGES: We have designed a microfluidic platform for CSC enriched culture by tumor sphere formation. Using MDs, we could quantify and determine sphere response after CT using murine and human cell lines as a proof of concept. MDs can be used as a tumor-derived sphere isolation platform to test the effect of antitumoral compounds in sphere proliferation.
Collapse
Affiliation(s)
- Eduardo Imanol Agüero
- Facultad de Ciencias Médicas, Instituto de Oncología "Ángel H. Roffo", Área de Investigación, Universidad de Buenos Aires, C1417DTB, Ciudad Autónoma de Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1425FQB, Ciudad Autónoma de Buenos Aires, Argentina
| | - Denise Belgorosky
- Facultad de Ciencias Médicas, Instituto de Oncología "Ángel H. Roffo", Área de Investigación, Universidad de Buenos Aires, C1417DTB, Ciudad Autónoma de Buenos Aires, Argentina
| | - Julio Israel García-Silva
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1425FQB, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ross Booth
- Roche Sequencing Solutions, Santa Clara, CA, 95050, USA
| | - Betiana Lerner
- Department of Electrical and Computer Engineering, Florida International University (FIU), Miami, FL, 33174, USA
- Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg im Breisgau, Germany
- Universidad Tecnológica Nacional (UTN), Centro IREN, B1706EAH, Buenos Aires, Argentina
- Facultad de Ingeniería, Instituto de Ingeniería Biomédica, Universidad de Buenos Aires, C1063ACV, Ciudad Autónoma de Buenos Aires, Argentina
| | - Maximiliano Sebastián Pérez
- Department of Electrical and Computer Engineering, Florida International University (FIU), Miami, FL, 33174, USA.
- Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg im Breisgau, Germany.
- Universidad Tecnológica Nacional (UTN), Centro IREN, B1706EAH, Buenos Aires, Argentina.
- Facultad de Ingeniería, Instituto de Ingeniería Biomédica, Universidad de Buenos Aires, C1063ACV, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Ana María Eiján
- Facultad de Ciencias Médicas, Instituto de Oncología "Ángel H. Roffo", Área de Investigación, Universidad de Buenos Aires, C1417DTB, Ciudad Autónoma de Buenos Aires, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1425FQB, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
23
|
Brisset M, Mehlen P, Meurette O, Hollande F. Notch receptor/ligand diversity: contribution to colorectal cancer stem cell heterogeneity. Front Cell Dev Biol 2023; 11:1231416. [PMID: 37860822 PMCID: PMC10582728 DOI: 10.3389/fcell.2023.1231416] [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: 05/30/2023] [Accepted: 09/21/2023] [Indexed: 10/21/2023] Open
Abstract
Cancer cell heterogeneity is a key contributor to therapeutic failure and post-treatment recurrence. Targeting cell subpopulations responsible for chemoresistance and recurrence seems to be an attractive approach to improve treatment outcome in cancer patients. However, this remains challenging due to the complexity and incomplete characterization of tumor cell subpopulations. The heterogeneity of cells exhibiting stemness-related features, such as self-renewal and chemoresistance, fuels this complexity. Notch signaling is a known regulator of cancer stem cell (CSC) features in colorectal cancer (CRC), though the effects of its heterogenous signaling on CRC cell stemness are only just emerging. In this review, we discuss how Notch ligand-receptor specificity contributes to regulating stemness, self-renewal, chemoresistance and cancer stem cells heterogeneity in CRC.
Collapse
Affiliation(s)
- Morgan Brisset
- Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, The University of Melbourne, Melbourne, VIC, Australia
- Cancer Cell Death Laboratory, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Patrick Mehlen
- Cancer Cell Death Laboratory, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Olivier Meurette
- Cancer Cell Death Laboratory, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Université de Lyon, Lyon, France
| | - Frédéric Hollande
- Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
24
|
Lasorsa F, Rutigliano M, Milella M, Ferro M, Pandolfo SD, Crocetto F, Autorino R, Battaglia M, Ditonno P, Lucarelli G. Cancer Stem Cells in Renal Cell Carcinoma: Origins and Biomarkers. Int J Mol Sci 2023; 24:13179. [PMID: 37685983 PMCID: PMC10487877 DOI: 10.3390/ijms241713179] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/14/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The term "cancer stem cell" (CSC) refers to a cancer cell with the following features: clonogenic ability, the expression of stem cell markers, differentiation into cells of different lineages, growth in nonadhesive spheroids, and the in vivo ability to generate serially transplantable tumors that reflect the heterogeneity of primary cancers (tumorigenicity). According to this model, CSCs may arise from normal stem cells, progenitor cells, and/or differentiated cells because of striking genetic/epigenetic mutations or from the fusion of tissue-specific stem cells with circulating bone marrow stem cells (BMSCs). CSCs use signaling pathways similar to those controlling cell fate during early embryogenesis (Notch, Wnt, Hedgehog, bone morphogenetic proteins (BMPs), fibroblast growth factors, leukemia inhibitory factor, and transforming growth factor-β). Recent studies identified a subpopulation of CD133+/CD24+ cells from ccRCC specimens that displayed self-renewal ability and clonogenic multipotency. The development of agents targeting CSC signaling-specific pathways and not only surface proteins may ultimately become of utmost importance for patients with RCC.
Collapse
Affiliation(s)
- Francesco Lasorsa
- Department of Precision and Regenerative Medicine and Ionian Area-Urology, Andrology and Kidney Transplantation Unit, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Monica Rutigliano
- Department of Precision and Regenerative Medicine and Ionian Area-Urology, Andrology and Kidney Transplantation Unit, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Martina Milella
- Department of Precision and Regenerative Medicine and Ionian Area-Urology, Andrology and Kidney Transplantation Unit, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Matteo Ferro
- Division of Urology, European Institute of Oncology, IRCCS, 71013 Milan, Italy
| | - Savio Domenico Pandolfo
- Department of Neurosciences and Reproductive Sciences and Odontostomatology, University of Naples “Federico II”, 80131 Naples, Italy
| | - Felice Crocetto
- Department of Neurosciences and Reproductive Sciences and Odontostomatology, University of Naples “Federico II”, 80131 Naples, Italy
| | - Riccardo Autorino
- Department of Urology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Michele Battaglia
- Department of Precision and Regenerative Medicine and Ionian Area-Urology, Andrology and Kidney Transplantation Unit, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Pasquale Ditonno
- Department of Precision and Regenerative Medicine and Ionian Area-Urology, Andrology and Kidney Transplantation Unit, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Giuseppe Lucarelli
- Department of Precision and Regenerative Medicine and Ionian Area-Urology, Andrology and Kidney Transplantation Unit, University of Bari “Aldo Moro”, 70124 Bari, Italy
| |
Collapse
|
25
|
Li C, Qiu S, Liu X, Guo F, Zhai J, Li Z, Deng L, Ge L, Qian H, Yang L, Xu B. Extracellular matrix-derived mechanical force governs breast cancer cell stemness and quiescence transition through integrin-DDR signaling. Signal Transduct Target Ther 2023; 8:247. [PMID: 37369642 DOI: 10.1038/s41392-023-01453-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 04/13/2023] [Accepted: 04/23/2023] [Indexed: 06/29/2023] Open
Abstract
The extracellular matrix (ECM) serves as signals that regulate specific cell states in tumor tissues. Increasing evidence suggests that extracellular biomechanical force signals are critical in tumor progression. In this study, we aimed to explore the influence of ECM-derived biomechanical force on breast cancer cell status. Experiments were conducted using 3D collagen, fibrinogen, and Matrigel matrices to investigate the role of mechanical force in tumor development. Integrin-cytoskeleton-AIRE and DDR-STAT signals were examined using RNA sequencing and western blotting. Data from 1358 patients and 86 clinical specimens were used for ECM signature-prognosis analysis. Our findings revealed that ECM-derived mechanical force regulated tumor stemness and cell quiescence in breast cancer cells. A mechanical force of ~45 Pa derived from the extracellular substrate activated integrin β1/3 receptors, stimulating stem cell signaling pathways through the cytoskeleton/AIRE axis and promoting tumorigenic potential and stem-like phenotypes. However, excessive mechanical force (450 Pa) could drive stem-like cancer cells into a quiescent state, with the removal of mechanical forces leading to vigorous proliferation in quiescent cancer stem cells. Mechanical force facilitated cell cycle arrest to induce quiescence, dependent on DDR2/STAT1/P27 signaling. Therefore, ECM-derived mechanical force governs breast cancer cell status and proliferative characteristics through stiffness alterations. We further established an ECM signature based on the fibrinogen/fibronectin/vitronectin/elastin axis, which efficiently predicts patient prognosis in breast cancer. Our findings highlight the vital role of ECM-derived mechanical force in governing breast cancer cell stemness/quiescence transition and suggest the novel use of ECM signature in predicting the clinical prognosis of breast cancer.
Collapse
Affiliation(s)
- Cong Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shi Qiu
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China
- National Clinical Research Center of Geriatrics, The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610065, China
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland (IOSI), Bellinzona, 6500, Switzerland
| | - Xiaohan Liu
- Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, Liaoning, 110122, China
| | - Fengzhu Guo
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jingtong Zhai
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhijun Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Linghui Deng
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland (IOSI), Bellinzona, 6500, Switzerland
| | - Liming Ge
- Department of Pharmaceutical and Bioengineering, School of Chemical Engineering, Sichuan University, Chengdu, 610065, China
| | - Haili Qian
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Lu Yang
- Department of Urology, Institute of Urology and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
26
|
Wang Y, Yu Y, Yang W, Wu L, Yang Y, Lu Q, Zhou J. SETD4 Confers Cancer Stem Cell Chemoresistance in Nonsmall Cell Lung Cancer Patients via the Epigenetic Regulation of Cellular Quiescence. Stem Cells Int 2023; 2023:7367854. [PMID: 37274024 PMCID: PMC10239305 DOI: 10.1155/2023/7367854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 04/23/2023] [Accepted: 05/08/2023] [Indexed: 06/06/2023] Open
Abstract
Increasing evidence indicates that quiescent cancer stem cells (CSCs) are a root cause of chemoresistance. SET domain-containing protein 4 (SETD4) epigenetically regulates cell quiescence in breast cancer stem cells (BCSCs), and SETD4-positive BCSCs are chemoradioresistant. However, the role of SETD4 in chemoresistance, tumor progression, and prognosis in nonsmall cell lung cancer (NSCLC) patients is unclear. Here, SETD4-positive cells were identified as quiescent lung cancer stem cells (qLCSCs) since they expressed high levels of ALDH1 and CD133 and low levels of Ki67. SETD4 expression was significantly higher in advanced-stage NSCLC tissues than in early-stage NSCLC tissues and significantly higher in samples from the chemoresistant group than in those from the chemosensitive group. Patients with high SETD4 expression had shorter progression-free survival (PFS) times than those with low SETD4 expression. SETD4 facilitated heterochromatin formation via H4K20me3, thereby leading to cell quiescence. RNA-seq analysis showed upregulation of genes involved in cell proliferation, glucose metabolism, and PI3K-AKT signaling in activated qLCSCs (A-qLCSCs) compared with qLCSCs. In addition, SETD4 overexpression facilitated PTEN-mediated inhibition of the PI3K-mTOR pathway. In summary, SETD4 confers chemoresistance, tumor progression, and a poor prognosis by regulating CSCs in NSCLC patients.
Collapse
Affiliation(s)
- Yuehong Wang
- Department of Respiratory Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yuman Yu
- Department of Geriatrics, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Weijun Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Linying Wu
- Department of Respiratory Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yaoshun Yang
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qianyun Lu
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianying Zhou
- Department of Respiratory Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
27
|
Li J, Goh ELK, He J, Li Y, Fan Z, Yu Z, Yuan P, Liu DX. Emerging Intrinsic Therapeutic Targets for Metastatic Breast Cancer. BIOLOGY 2023; 12:697. [PMID: 37237509 PMCID: PMC10215321 DOI: 10.3390/biology12050697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023]
Abstract
Breast cancer is now the most common cancer worldwide, and it is also the main cause of cancer-related death in women. Survival rates for female breast cancer have significantly improved due to early diagnosis and better treatment. Nevertheless, for patients with advanced or metastatic breast cancer, the survival rate is still low, reflecting a need for the development of new therapies. Mechanistic insights into metastatic breast cancer have provided excellent opportunities for developing novel therapeutic strategies. Although high-throughput approaches have identified several therapeutic targets in metastatic disease, some subtypes such as triple-negative breast cancer do not yet have an apparent tumor-specific receptor or pathway to target. Therefore, exploring new druggable targets in metastatic disease is a high clinical priority. In this review, we summarize the emerging intrinsic therapeutic targets for metastatic breast cancer, including cyclin D-dependent kinases CDK4 and CDK6, the PI3K/AKT/mTOR pathway, the insulin/IGF1R pathway, the EGFR/HER family, the JAK/STAT pathway, poly(ADP-ribose) polymerases (PARP), TROP-2, Src kinases, histone modification enzymes, activated growth factor receptors, androgen receptors, breast cancer stem cells, matrix metalloproteinases, and immune checkpoint proteins. We also review the latest development in breast cancer immunotherapy. Drugs that target these molecules/pathways are either already FDA-approved or currently being tested in clinical trials.
Collapse
Affiliation(s)
- Jiawei Li
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand
| | - Eyleen L. K. Goh
- Neuroscience and Mental Health Faculty, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Ji He
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand
| | - Yan Li
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand
| | - Zhimin Fan
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Zhigang Yu
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan 250033, China;
| | - Peng Yuan
- Department of VIP Medical Services, National Cancer Centre/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dong-Xu Liu
- The Centre for Biomedical and Chemical Sciences, School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 1010, New Zealand
| |
Collapse
|
28
|
Lin F, Ke ZB, Chen H, Zheng WC, Dong RN, Cai H, Li XD, Wei Y, Zheng QS, Xue XY, Chen SH, Xu N. Integrative analysis developing and validating potential candidate biomarkers for cancer stemness features of pan-renal cell carcinoma. Cancer Invest 2023:1-17. [PMID: 37129517 DOI: 10.1080/07357907.2023.2209634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
In our study, 49 key genes significantly associated with renal cell carcinoma (RCC) stemness were obtained. Next, we developed a molecular prognostic signature associated with stemness features of pan-RCC. The difference in OS between high-risk and low-risk group was statistically significant (P < 0.05). The area under ROC curve for 1-years OS, 5-years OS and 10-years OS was 0.759, 0.712 and 0.918, respectively. The results of validation in TCGA cohort and ICGC cohort revealed the predictive capability of this signature. Further, we selected three genes and further validation showed that these three hub genes were potential hub biomarkers for pan-RCC stemness features.
Collapse
Affiliation(s)
- Fei Lin
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Zhi-Bin Ke
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Hang Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Wen-Cai Zheng
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Ru-Nan Dong
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Hai Cai
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Xiao-Dong Li
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Qing-Shui Zheng
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Xue-Yi Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Shao-Hao Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Ning Xu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
29
|
Loric S, Denis JA, Desbene C, Sabbah M, Conti M. Extracellular Vesicles in Breast Cancer: From Biology and Function to Clinical Diagnosis and Therapeutic Management. Int J Mol Sci 2023; 24:7208. [PMID: 37108371 PMCID: PMC10139222 DOI: 10.3390/ijms24087208] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/03/2023] [Accepted: 04/09/2023] [Indexed: 04/29/2023] Open
Abstract
Breast cancer (BC) is the first worldwide most frequent cancer in both sexes and the most commonly diagnosed in females. Although BC mortality has been thoroughly declining over the past decades, there are still considerable differences between women diagnosed with early BC and when metastatic BC is diagnosed. BC treatment choice is widely dependent on precise histological and molecular characterization. However, recurrence or distant metastasis still occurs even with the most recent efficient therapies. Thus, a better understanding of the different factors underlying tumor escape is mainly mandatory. Among the leading candidates is the continuous interplay between tumor cells and their microenvironment, where extracellular vesicles play a significant role. Among extracellular vesicles, smaller ones, also called exosomes, can carry biomolecules, such as lipids, proteins, and nucleic acids, and generate signal transmission through an intercellular transfer of their content. This mechanism allows tumor cells to recruit and modify the adjacent and systemic microenvironment to support further invasion and dissemination. By reciprocity, stromal cells can also use exosomes to profoundly modify tumor cell behavior. This review intends to cover the most recent literature on the role of extracellular vesicle production in normal and cancerous breast tissues. Specific attention is paid to the use of extracellular vesicles for early BC diagnosis, follow-up, and prognosis because exosomes are actually under the spotlight of researchers as a high-potential source of liquid biopsies. Extracellular vesicles in BC treatment as new targets for therapy or efficient nanovectors to drive drug delivery are also summarized.
Collapse
Affiliation(s)
- Sylvain Loric
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | | | - Cédric Desbene
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Michèle Sabbah
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
| | - Marc Conti
- INSERM U538, CRSA, Saint-Antoine University Hospital, 75012 Paris, France; (J.A.D.)
- INTEGRACELL SAS, 91160 Longjumeau, France
| |
Collapse
|
30
|
Abstract
Head and neck cancers are a heterogeneous group of highly aggressive tumors and collectively represent the sixth most common cancer worldwide. Most head and neck cancers are squamous cell carcinomas (HNSCCs). Current multimodal treatment concepts combine surgery, chemotherapy, irradiation, immunotherapy, and targeted therapeutics. Recent scientific advancements have enabled a more precise molecular characterization of HNSCC and revealed novel therapeutic targets and prognostic/predictive biomarkers. Notably, HNSCC is characterized by complex relations between stromal, epithelial, and immune cells within the tumor microenvironment (TME). The TME consists of different subsets of immune cells that infiltrate the tumors and interact with the tumor cells or with each other. Understanding multiple pivotal factors in HNSCC tumorigenesis and tumor progression may help define novel targets and develop more effective therapies for patients. This review provides a comprehensive overview of the latest advances in the molecular biology of HNSCC and their effects on clinical oncology; it is meant for a broad readership in the head and neck cancers field.
Collapse
Affiliation(s)
- Subramanya Pandruvada
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, United States.
| | - Remi Kessler
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Ann Thai
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
31
|
Morimoto Y, Yamashita N, Hirose H, Fushimi A, Haratake N, Daimon T, Bhattacharya A, Ahmad R, Suzuki Y, Takahashi H, Kufe DW. MUC1-C is necessary for SHP2 activation and BRAF inhibitor resistance in BRAF(V600E) mutant colorectal cancer. Cancer Lett 2023; 559:216116. [PMID: 36878307 PMCID: PMC10408991 DOI: 10.1016/j.canlet.2023.216116] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/19/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Colorectal cancers (CRCs) harboring the BRAF(V600E) mutation are associated with aggressive disease and resistance to BRAF inhibitors by feedback activation of the receptor tyrosine kinase (RTK)→RAS→MAPK pathway. The oncogenic MUC1-C protein promotes progression of colitis to CRC; whereas there is no known involvement of MUC1-C in BRAF(V600E) CRCs. The present work demonstrates that MUC1 expression is significantly upregulated in BRAF(V600E) vs wild-type CRCs. We show that BRAF(V600E) CRC cells are dependent on MUC1-C for proliferation and BRAF inhibitor (BRAFi) resistance. Mechanistically, MUC1-C integrates induction of MYC in driving cell cycle progression with activation of the SHP2 phosphotyrosine phosphatase, which enhances RTK-mediated RAS→ERK signaling. We demonstrate that targeting MUC1-C genetically and pharmacologically suppresses (i) activation of MYC, (ii) induction of the NOTCH1 stemness factor, and (iii) the capacity for self-renewal. We also show that MUC1-C associates with SHP2 and is required for SHP2 activation in driving BRAFi-induced feedback of ERK signaling. In this way, targeting MUC1-C in BRAFi-resistant BRAF(V600E) CRC tumors inhibits growth and sensitizes to BRAF inhibition. These findings demonstrate that MUC1-C is a target for the treatment of BRAF(V600E) CRCs and for reversing their resistance to BRAF inhibitors by suppressing the feedback MAPK pathway.
Collapse
Affiliation(s)
| | - Nami Yamashita
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Haruka Hirose
- Division of Systems Biology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Atsushi Fushimi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Naoki Haratake
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Tatsuaki Daimon
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Rehan Ahmad
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yozo Suzuki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Hidekazu Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Donald W Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
32
|
Azari M, Bahreini F, Uversky VN, Rezaei N. Current therapeutic approaches and promising perspectives of using bioengineered peptides in fighting chemoresistance in triple-negative breast cancer. Biochem Pharmacol 2023; 210:115459. [PMID: 36813121 DOI: 10.1016/j.bcp.2023.115459] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 02/11/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023]
Abstract
Breast cancer is a collation of malignancies that manifest in the mammary glands at the early stages. Among breast cancer subtypes, triple-negative breast cancer (TNBC) shows the most aggressive behavior, with apparent stemness features. Owing to the lack of response to hormone therapy and specific targeted therapies, chemotherapy remains the first line of the TNBC treatment. However, the acquisition of resistance to chemotherapeutic agents increase therapy failure, and promotes cancer recurrence and distant metastasis. Invasive primary tumors are the birthplace of cancer burden, though metastasis is a key attribute of TNBC-associated morbidity and mortality. Targeting the chemoresistant metastases-initiating cells via specific therapeutic agents with affinity to the upregulated molecular targets is a promising step in the TNBC clinical management. Exploring the capacity of peptides as biocompatible entities with the specificity of action, low immunogenicity, and robust efficacy provides a principle for designing peptide-based drugs capable of increasing the efficacy of current chemotherapy agents for selective targeting of the drug-tolerant TNBC cells. Here, we first focus on the resistance mechanisms that TNBC cells acquire to evade the effect of chemotherapeutic agents. Next, the novel therapeutic approaches employing tumor-targeting peptides to exploit the mechanisms of drug resistance in chemorefractory TNBC are described.
Collapse
Affiliation(s)
- Mandana Azari
- School of Chemical Engineering-Biotechnology, College of Engineering, University of Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farbod Bahreini
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Yamashita N, Morimoto Y, Fushimi A, Ahmad R, Bhattacharya A, Daimon T, Haratake N, Inoue Y, Ishikawa S, Yamamoto M, Hata T, Akiyoshi S, Hu Q, Liu T, Withers H, Liu S, Shapiro GI, Yoshizumi T, Long MD, Kufe D. MUC1-C Dictates PBRM1-Mediated Chronic Induction of Interferon Signaling, DNA Damage Resistance, and Immunosuppression in Triple-Negative Breast Cancer. Mol Cancer Res 2023; 21:274-289. [PMID: 36445328 PMCID: PMC9975675 DOI: 10.1158/1541-7786.mcr-22-0772] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/25/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022]
Abstract
The polybromo-1 (PBRM1) chromatin-targeting subunit of the SWI/SNF PBAF chromatin remodeling complex drives DNA damage resistance and immune evasion in certain cancer cells through mechanisms that remain unclear. STAT1 and IRF1 are essential effectors of type I and II IFN pathways. Here, we report that MUC1-C is necessary for PBRM1 expression and that it forms a nuclear complex with PBRM1 in triple-negative breast cancer (TNBC) cells. Analysis of global transcriptional (RNA-seq) and chromatin accessibility (ATAC-seq) profiles further demonstrated that MUC1-C and PBRM1 drive STAT1 and IRF1 expression by increasing chromatin accessibility of promoter-like signatures (PLS) on their respective genes. We also found that MUC1-C, PBRM1, and IRF1 increase the expression and chromatin accessibility on PLSs of the (i) type II IFN pathway IDO1 and WARS genes and (ii) type I IFN pathway RIG-I, MDA5, and ISG15 genes that collectively contribute to DNA damage resistance and immune evasion. In support of these results, targeting MUC1-C in wild-type BRCA TNBC cells enhanced carboplatin-induced DNA damage and the loss of self-renewal capacity. In addition, MUC1-C was necessary for DNA damage resistance, self-renewal, and tumorigenicity in olaparib-resistant BRCA1-mutant TNBC cells. Analysis of TNBC tumors corroborated that (i) MUC1 and PBRM1 are associated with decreased responsiveness to chemotherapy and (ii) MUC1-C expression is associated with the depletion of tumor-infiltrating lymphocytes (TIL). These findings demonstrate that MUC1-C activates PBRM1, and thereby chromatin remodeling of IFN-stimulated genes that promote chronic inflammation, DNA damage resistance, and immune evasion. IMPLICATIONS MUC1-C is necessary for PBRM1-driven chromatin remodeling in chronic activation of IFN pathway genes that promote DNA damage resistance and immunosuppression.
Collapse
Affiliation(s)
- Nami Yamashita
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts
| | - Yoshihiro Morimoto
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts
| | - Atsushi Fushimi
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts
| | - Rehan Ahmad
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts
| | - Atrayee Bhattacharya
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts
| | - Tatsuaki Daimon
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts
| | - Naoki Haratake
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts
| | - Yuka Inoue
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoshi Ishikawa
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts
| | - Masaaki Yamamoto
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts
| | - Tsuyoshi Hata
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts
| | - Sayuri Akiyoshi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Qiang Hu
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Tao Liu
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Henry Withers
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Song Liu
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Geoffrey I. Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mark D. Long
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Corresponding Authors: Donald Kufe, Dana-Farber Cancer Institute, 450 Brookline Avenue, D830, Boston, MA 02215. E-mail: ; and Mark D. Long,
| | - Donald Kufe
- Department of Medical Oncology, Dana-Farber Cancer Institute Harvard Medical School, Boston, Massachusetts
- Corresponding Authors: Donald Kufe, Dana-Farber Cancer Institute, 450 Brookline Avenue, D830, Boston, MA 02215. E-mail: ; and Mark D. Long,
| |
Collapse
|
34
|
Alhasan B, Mikeladze M, Guzhova I, Margulis B. Autophagy, molecular chaperones, and unfolded protein response as promoters of tumor recurrence. Cancer Metastasis Rev 2023; 42:217-254. [PMID: 36723697 DOI: 10.1007/s10555-023-10085-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/16/2023] [Indexed: 02/02/2023]
Abstract
Tumor recurrence is a paradoxical function of a machinery, whereby a small proportion of the cancer cell population enters a resistant, dormant state, persists long-term in this condition, and then transitions to proliferation. The dormant phenotype is typical of cancer stem cells, tumor-initiating cells, disseminated tumor cells, and drug-tolerant persisters, which all demonstrate similar or even equivalent properties. Cancer cell dormancy and its conversion to repopulation are regulated by several protein signaling systems that inhibit or induce cell proliferation and provide optimal interrelations between cancer cells and their special niche; these systems act in close connection with tumor microenvironment and immune response mechanisms. During dormancy and reawakening periods, cell proteostasis machineries, autophagy, molecular chaperones, and the unfolded protein response are recruited to protect refractory tumor cells from a wide variety of stressors and therapeutic insults. Proteostasis mechanisms functionally or even physically interfere with the main regulators of tumor relapse, and the significance of these interactions and implications in the tumor recurrence phases are discussed in this review.
Collapse
Affiliation(s)
- Bashar Alhasan
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia.
| | - Marina Mikeladze
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| | - Irina Guzhova
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| | - Boris Margulis
- Institute of Cytology, Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064, St. Petersburg, Russia
| |
Collapse
|
35
|
Dormancy, stemness, and therapy resistance: interconnected players in cancer evolution. Cancer Metastasis Rev 2023; 42:197-215. [PMID: 36757577 PMCID: PMC10014678 DOI: 10.1007/s10555-023-10092-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023]
Abstract
The biological complexity of cancer represents a tremendous clinical challenge, resulting in the frequent failure of current treatment protocols. In the rapidly evolving scenario of a growing tumor, anticancer treatments impose a drastic perturbation not only to cancer cells but also to the tumor microenvironment, killing a portion of the cells and inducing a massive stress response in the survivors. Consequently, treatments can act as a double-edged sword by inducing a temporary response while laying the ground for therapy resistance and subsequent disease progression. Cancer cell dormancy (or quiescence) is a central theme in tumor evolution, being tightly linked to the tumor's ability to survive cytotoxic challenges, metastasize, and resist immune-mediated attack. Accordingly, quiescent cancer cells (QCCs) have been detected in virtually all the stages of tumor development. In recent years, an increasing number of studies have focused on the characterization of quiescent/therapy resistant cancer cells, unveiling QCCs core transcriptional programs, metabolic plasticity, and mechanisms of immune escape. At the same time, our partial understanding of tumor quiescence reflects the difficulty to identify stable QCCs biomarkers/therapeutic targets and to control cancer dormancy in clinical settings. This review focuses on recent discoveries in the interrelated fields of dormancy, stemness, and therapy resistance, discussing experimental evidences in the frame of a nonlinear dynamics approach, and exploring the possibility that tumor quiescence may represent not only a peril but also a potential therapeutic resource.
Collapse
|
36
|
Chai C, Ji P, Xu H, Tang H, Wang Z, Zhang H, Zhou W. Targeting cancer drug resistance utilizing organoid technology. Biomed Pharmacother 2023; 158:114098. [PMID: 36528918 DOI: 10.1016/j.biopha.2022.114098] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer organoids generated from 3D in vitro cell cultures have contributed to the study of drug resistance. Maintenance of genomic and transcriptomic similarity between organoids and parental cancer allows organoids to have the ability of accurate prediction in drug resistance testing. Protocols of establishing therapy-sensitive and therapy-resistant organoids are concluded in two aspects, which are generated directly from respective patients' cancer and by induction of anti-cancer drug. Genomic and transcriptomic analyses and gene editing have been applied to organoid studies to identify key targets in drug resistance and FGFR3, KHDRBS3, lnc-RP11-536 K7.3 and FBN1 were found to be key targets. Furthermore, mechanisms contributing to resistance have been identified, including metabolic adaptation, activation of DNA damage response, defects in apoptosis, reduced cellular senescence, cellular plasticity, subpopulation interactions and gene fusions. Additionally, cancer stem cells (CSCs) have been verified to be involved in drug resistance utilizing organoid technology. Reversal of drug resistance can be achieved by targeting key genes and CSCs in cancer organoids. In this review, we summarize applications of organoids to cancer drug resistance research, indicating prospects and limitations.
Collapse
Affiliation(s)
- Changpeng Chai
- The First Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China; The Forth Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Pengfei Ji
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Hao Xu
- The First Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Huan Tang
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Zhengfeng Wang
- The First Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Hui Zhang
- The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China.
| | - Wence Zhou
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou 730000, Gansu, China; The Second Hospital of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu, China.
| |
Collapse
|
37
|
Saggese P, Pandey A, Fung E, Hall A, Yanagawa J, Rodriguez EF, Grogan TR, Giurato G, Nassa G, Salvati A, Weisz A, Dubinett SM, Scafoglio C. Glucose deprivation promotes pseudo-hypoxia and de-differentiation in lung adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526207. [PMID: 36778362 PMCID: PMC9915520 DOI: 10.1101/2023.01.30.526207] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Increased utilization of glucose is a hallmark of cancer. Several studies are investigating the efficacy of glucose restriction by glucose transporter blockade or glycolysis inhibition. However, the adaptations of cancer cells to glucose restriction are unknown. Here, we report the discovery that glucose restriction in lung adenocarcinoma (LUAD) induces cancer cell de-differentiation, leading to a more aggressive phenotype. Glucose deprivation causes a reduction in alpha-ketoglutarate (αKG), leading to attenuated activity of αKG-dependent histone demethylases and histone hypermethylation. We further show that this de-differentiated phenotype depends on unbalanced EZH2 activity, causing inhibition of prolyl-hydroxylase PHD3 and increased expression of hypoxia inducible factor 1α (HIF1α), triggering epithelial to mesenchymal transition. Finally, we identified an HIF1α-dependent transcriptional signature with prognostic significance in human LUAD. Our studies further current knowledge of the relationship between glucose metabolism and cell differentiation in cancer, characterizing the epigenetic adaptation of cancer cells to glucose deprivation and identifying novel targets to prevent the development of resistance to therapies targeting glucose metabolism.
Collapse
Affiliation(s)
- Pasquale Saggese
- Division of Pulmonary Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Aparamita Pandey
- Division of Pulmonary Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Eileen Fung
- Division of Pulmonary Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Division of Thoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Abbie Hall
- Division of Pulmonary Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jane Yanagawa
- Division of Thoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Erika F. Rodriguez
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Tristan R. Grogan
- Division of General Internal Medicine and Health Services Research, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health - CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health - CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
| | - Annamaria Salvati
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health - CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
- Medical Genomics Program and Division of Onco-Hematology, AOU “S. Giovanni di Dio e Ruggi d’Aragona”, University of Salerno, Salerno, Italy
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry ‘Scuola Medica Salernitana’, University of Salerno, Baronissi (SA), Italy
- Genome Research Center for Health - CRGS, Campus of Medicine of the University of Salerno, Baronissi (SA), Italy
- Medical Genomics Program and Division of Onco-Hematology, AOU “S. Giovanni di Dio e Ruggi d’Aragona”, University of Salerno, Salerno, Italy
| | - Steven M. Dubinett
- Division of Pulmonary Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Claudio Scafoglio
- Division of Pulmonary Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
38
|
Seledtsov VI, Darinskas A, Von Delwig A, Seledtsova GV. Inflammation Control and Immunotherapeutic Strategies in Comprehensive Cancer Treatment. Metabolites 2023; 13:metabo13010123. [PMID: 36677048 PMCID: PMC9865335 DOI: 10.3390/metabo13010123] [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: 12/15/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Tumor growth and expansion are determined by the immunological tumor microenvironment (TME). Typically, early tumorigenic stages are characterized by the immune system not responding or weakly responding to the tumor. However, subsequent tumorigenic stages witness the tumor promoting its growth and metastasis by stimulating tumor-protective (pro-tumor) inflammation to suppress anti-tumor immune responses. Here, we propose the pivotal role of inflammation control in a successful anti-cancer immunotherapy strategy, implying that available and novel immunotherapeutic modalities such as inflammation modulation, antibody (Ab)-based immunostimulation, drug-mediated immunomodulation, cancer vaccination as well as adoptive cell immunotherapy and donor leucocyte transfusion could be applied in cancer patients in a synergistic manner to amplify each other's clinical effects and achieve robust anti-tumor immune reactivity. In addition, the anti-tumor effects of immunotherapy could be enhanced by thermal and/or oxygen therapy. Herein, combined immune-based therapy could prove to be beneficial for patients with advanced cancers, as aiming to provide long-term tumor cell/mass dormancy by restraining compensatory proliferation of surviving cancer cells observed after traditional anti-cancer interventions such as surgery, radiotherapy, and metronomic (low-dose) chemotherapy. We propose the Inflammatory Prognostic Score based on the blood levels of C-reactive protein and lactate dehydrogenase as well as the neutrophil-to-lymphocyte ratio to effectively monitor the effectiveness of comprehensive anti-cancer treatment.
Collapse
Affiliation(s)
- Victor Ivanovich Seledtsov
- Innovita Research Company, 06116 Vilnius, Lithuania
- Russian Scientific Center of Surgery Named after Academician B.V. Petrovsky, 119991 Moscow, Russia
- Correspondence: ; Tel.: +7-915-2636027
| | | | | | | |
Collapse
|
39
|
Moore PC, Henderson KW, Classon M. The epigenome and the many facets of cancer drug tolerance. Adv Cancer Res 2023; 158:1-39. [PMID: 36990531 DOI: 10.1016/bs.acr.2022.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The use of chemotherapeutic agents and the development of new cancer therapies over the past few decades has consequently led to the emergence of myriad therapeutic resistance mechanisms. Once thought to be explicitly driven by genetics, the coupling of reversible sensitivity and absence of pre-existing mutations in some tumors opened the way for discovery of drug-tolerant persisters (DTPs): slow-cycling subpopulations of tumor cells that exhibit reversible sensitivity to therapy. These cells confer multi-drug tolerance, to targeted and chemotherapies alike, until the residual disease can establish a stable, drug-resistant state. The DTP state can exploit a multitude of distinct, yet interlaced, mechanisms to survive otherwise lethal drug exposures. Here, we categorize these multi-faceted defense mechanisms into unique Hallmarks of Cancer Drug Tolerance. At the highest level, these are comprised of heterogeneity, signaling plasticity, differentiation, proliferation/metabolism, stress management, genomic integrity, crosstalk with the tumor microenvironment, immune escape, and epigenetic regulatory mechanisms. Of these, epigenetics was both one of the first proposed means of non-genetic resistance and one of the first discovered. As we describe in this review, epigenetic regulatory factors are involved in most facets of DTP biology, positioning this hallmark as an overarching mediator of drug tolerance and a potential avenue to novel therapies.
Collapse
|
40
|
Adebayo AK, Nakshatri H. Modeling Preclinical Cancer Studies under Physioxia to Enhance Clinical Translation. Cancer Res 2022; 82:4313-4321. [PMID: 36169928 PMCID: PMC9722631 DOI: 10.1158/0008-5472.can-22-2311] [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: 07/20/2022] [Revised: 08/31/2022] [Accepted: 09/23/2022] [Indexed: 01/24/2023]
Abstract
Oxygen (O2) plays a key role in cellular homeostasis. O2 levels are tightly regulated in vivo such that each tissue receives an optimal amount to maintain physiologic status. Physiologic O2 levels in various organs range between 2% and 9% in vivo, with the highest levels of 9% in the kidneys and the lowest of 0.5% in parts of the brain. This physiologic range of O2 tensions is disrupted in pathologic conditions such as cancer, where it can reach as low as 0.5%. Regardless of the state, O2 tension in vivo is maintained at significantly lower levels than ambient O2, which is approximately 21%. Yet, routine in vitro cellular manipulations are carried out in ambient air, regardless of whether or not they are eventually transferred to hypoxic conditions for subsequent studies. Even brief exposure of hematopoietic stem cells to ambient air can cause detrimental effects through a mechanism termed extraphysiologic oxygen shock/stress (EPHOSS), leading to reduced engraftment capabilities. Here, we provide an overview of the effects of ambient air exposure on stem and non-stem cell subtypes, with a focus on recent findings that reveal the impact of EPHOSS on cancer cells.
Collapse
Affiliation(s)
- Adedeji K. Adebayo
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana University Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
41
|
Lee-Theilen M, Fadini DD, Hadhoud JR, van Dongen F, Kroll G, Rolle U, Fiegel HC. Hepatoblastoma Cancer Stem Cells Express PD-L1, Reveal Plasticity and Can Emerge upon Chemotherapy. Cancers (Basel) 2022; 14:cancers14235825. [PMID: 36497307 PMCID: PMC9736435 DOI: 10.3390/cancers14235825] [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: 10/26/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
The biology of cancer stem cells (CSCs) of pediatric cancers, such as hepatoblastoma, is sparsely explored. This is mainly due to the very immature nature of these tumors, which complicates the distinction of CSCs from the other tumor cells. Previously, we identified a CSC population in hepatoblastoma cell lines expressing the CSC markers CD34 and CD90, cell surface Vimentin (csVimentin) and binding of OV-6. In this study, we detected the co-expression of the immune escape factor PD-L1 in the CSC population, whereas the other tumor cells remained negative. FACS data revealed that non-CSCs give rise to CSCs, reflecting plasticity of CSCs and non-CSCs in hepatoblastoma as seen in other tumors. When we treated cells with cisplatin and decitabine, a new CD34+/lowOV-6lowCD90+ population emerged that lacked csVimentin and PD-L1 expression. Expression analyses showed that this new CSC subset shared similar pluripotency and EMT features with the already-known CSCs. FACS results further revealed that this subset is also generated from non-CSCs. In conclusion, we showed that hepatoblastoma CSCs express PD-L1 and that the biology of hepatoblastoma CSCs is of a plastic nature. Chemotherapeutic treatment leads to another CSC subset, which is highly chemoresistant and could be responsible for a poor prognosis after postoperative chemotherapy.
Collapse
|
42
|
Liu H, Zhao H, Sun Y. Tumor microenvironment and cellular senescence: Understanding therapeutic resistance and harnessing strategies. Semin Cancer Biol 2022; 86:769-781. [PMID: 34799201 DOI: 10.1016/j.semcancer.2021.11.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/24/2021] [Accepted: 11/08/2021] [Indexed: 01/27/2023]
Abstract
The tumor microenvironment (TME) is a major contributor to cancer malignancy including development of therapeutic resistance, a process mediated in part through intercellular crosstalk. Besides diverse soluble factors responsible for pro-survival pathway activation, immune evasion and extracellular matrix (ECM) remodeling further promote cancer resistance. Importantly, therapy-induced senescence (TIS) of cells in the TME is frequently observed in anticancer regimens, an off-target effect that can generate profound impacts on disease progression. By conferring the resistance and fueling the repopulation of remaining cancerous cells, TIS is responsible for tumor relapse and distant metastasis in posttreatment stage. This pathological trajectory can be substantially driven by the pro-inflammatory feature of senescent cells, termed as the senescence-associated secretory phenotype (SASP). Targeting strategies to selectively and efficiently remove senescent cells before they exert non-autonomous but largely deleterious effects, are emerging as an effective solution to prevent drug resistance acquired from a treatment-remodeled TME. In this review, we summarize the TME composition and key activities that affect tissue homeostasis and support treatment resistance. Promising opportunities that allow TME-manipulation and senescent cell-targeting (senotherapy) are discussed, with translational pipelines to overcome therapeutic barriers in clinical oncology projected.
Collapse
Affiliation(s)
- Hanxin Liu
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Huifang Zhao
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong, 264003, China
| | - Yu Sun
- Department of Pharmacology, Institute of Aging Medicine, Binzhou Medical University, Yantai, Shandong, 264003, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China; Department of Medicine and VAPSHCS, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
43
|
Karagiorgou Z, Fountas PN, Manou D, Knutsen E, Theocharis AD. Proteoglycans Determine the Dynamic Landscape of EMT and Cancer Cell Stemness. Cancers (Basel) 2022; 14:5328. [PMID: 36358747 PMCID: PMC9653992 DOI: 10.3390/cancers14215328] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 03/15/2024] Open
Abstract
Proteoglycans (PGs) are pivotal components of extracellular matrices, involved in a variety of processes such as migration, invasion, morphogenesis, differentiation, drug resistance, and epithelial-to-mesenchymal transition (EMT). Cellular plasticity is a crucial intermediate phenotypic state acquired by cancer cells, which can modulate EMT and the generation of cancer stem cells (CSCs). PGs affect cell plasticity, stemness, and EMT, altering the cellular shape and functions. PGs control these functions, either by direct activation of signaling cascades, acting as co-receptors, or through regulation of the availability of biological compounds such as growth factors and cytokines. Differential expression of microRNAs is also associated with the expression of PGs and their interplay is implicated in the fine tuning of cancer cell phenotype and potential. This review summarizes the involvement of PGs in the regulation of EMT and stemness of cancer cells and highlights the molecular mechanisms.
Collapse
Affiliation(s)
- Zoi Karagiorgou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Panagiotis N. Fountas
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Erik Knutsen
- Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, 9010 Tromsø, Norway
- Centre for Clinical Research and Education, University Hospital of North Norway, 9038 Tromsø, Norway
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
44
|
Nayak A, Warrier NM, Kumar P. Cancer Stem Cells and the Tumor Microenvironment: Targeting the Critical Crosstalk through Nanocarrier Systems. Stem Cell Rev Rep 2022; 18:2209-2233. [PMID: 35876959 PMCID: PMC9489588 DOI: 10.1007/s12015-022-10426-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2022] [Indexed: 11/25/2022]
Abstract
The physiological state of the tumor microenvironment (TME) plays a central role in cancer development due to multiple universal features that transcend heterogeneity and niche specifications, like promoting cancer progression and metastasis. As a result of their preponderant involvement in tumor growth and maintenance through several microsystemic alterations, including hypoxia, oxidative stress, and acidosis, TMEs make for ideal targets in both diagnostic and therapeutic ventures. Correspondingly, methodologies to target TMEs have been investigated this past decade as stratagems of significant potential in the genre of focused cancer treatment. Within targeted oncotherapy, nanomedical derivates-nanocarriers (NCs) especially-have emerged to present notable prospects in enhancing targeting specificity. Yet, one major issue in the application of NCs in microenvironmental directed therapy is that TMEs are too broad a spectrum of targeting possibilities for these carriers to be effectively employed. However, cancer stem cells (CSCs) might portend a solution to the above conundrum: aside from being quite heavily invested in tumorigenesis and therapeutic resistance, CSCs also show self-renewal and fluid clonogenic properties that often define specific TME niches. Further scrutiny of the relationship between CSCs and TMEs also points towards mechanisms that underly tumoral characteristics of metastasis, malignancy, and even resistance. This review summarizes recent advances in NC-enabled targeting of CSCs for more holistic strikes against TMEs and discusses both the current challenges that hinder the clinical application of these strategies as well as the avenues that can further CSC-targeting initiatives. Central role of CSCs in regulation of cellular components within the TME.
Collapse
Affiliation(s)
- Aadya Nayak
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Neerada Meenakshi Warrier
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Praveen Kumar
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
45
|
Constantinescu T, Mihis AG. Two Important Anticancer Mechanisms of Natural and Synthetic Chalcones. Int J Mol Sci 2022; 23:11595. [PMID: 36232899 PMCID: PMC9570335 DOI: 10.3390/ijms231911595] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
ATP-binding cassette subfamily G and tubulin pharmacological mechanisms decrease the effectiveness of anticancer drugs by modulating drug absorption and by creating tubulin assembly through polymerization. A series of natural and synthetic chalcones have been reported to have very good anticancer activity, with a half-maximal inhibitory concentration lower than 1 µM. By modulation, it is observed in case of the first mechanism that methoxy substituents on the aromatic cycle of acetophenone residue and substitution of phenyl nucleus by a heterocycle and by methoxy or hydroxyl groups have a positive impact. To inhibit tubulin, compounds bind to colchicine binding site. Presence of methoxy groups, amino groups or heterocyclic substituents increase activity.
Collapse
Affiliation(s)
- Teodora Constantinescu
- Department of Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University, 400012 Cluj-Napoca, Romania
| | - Alin Grig Mihis
- Advanced Materials and Applied Technologies Laboratory, Institute of Research-Development-Innovation in Applied Natural Sciences, “Babes-Bolyai” University, Fantanele Str. 30, 400294 Cluj-Napoca, Romania
| |
Collapse
|
46
|
Karami Fath M, Babakhaniyan K, Anjomrooz M, Jalalifar M, Alizadeh SD, Pourghasem Z, Abbasi Oshagh P, Azargoonjahromi A, Almasi F, Manzoor HZ, Khalesi B, Pourzardosht N, Khalili S, Payandeh Z. Recent Advances in Glioma Cancer Treatment: Conventional and Epigenetic Realms. Vaccines (Basel) 2022; 10:1448. [PMID: 36146527 PMCID: PMC9501259 DOI: 10.3390/vaccines10091448] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/14/2022] [Accepted: 08/27/2022] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma (GBM) is the most typical and aggressive form of primary brain tumor in adults, with a poor prognosis. Successful glioma treatment is hampered by ineffective medication distribution across the blood-brain barrier (BBB) and the emergence of drug resistance. Although a few FDA-approved multimodal treatments are available for glioblastoma, most patients still have poor prognoses. Targeting epigenetic variables, immunotherapy, gene therapy, and different vaccine- and peptide-based treatments are some innovative approaches to improve anti-glioma treatment efficacy. Following the identification of lymphatics in the central nervous system, immunotherapy offers a potential method with the potency to permeate the blood-brain barrier. This review will discuss the rationale, tactics, benefits, and drawbacks of current glioma therapy options in clinical and preclinical investigations.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran 1571914911, Iran
| | - Kimiya Babakhaniyan
- Department of Medical Surgical Nursing, School of Nursing and Midwifery, Iran University of Medical Sciences, Tehran 1996713883, Iran
| | - Mehran Anjomrooz
- Department of Radiology, Shariati Hospital, Tehran University of Medical Sciences, Tehran 1411713135, Iran
| | | | | | - Zeinab Pourghasem
- Department of Microbiology, Islamic Azad University of Lahijan, Gilan 4416939515, Iran
| | - Parisa Abbasi Oshagh
- Department of Biology, Faculty of Basic Sciences, Malayer University, Malayer 6571995863, Iran
| | - Ali Azargoonjahromi
- Department of Nursing, School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz 7417773539, Iran
| | - Faezeh Almasi
- Pharmaceutical Biotechnology Lab, Department of Microbial Biotechnology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran 1411734115, Iran
| | - Hafza Zahira Manzoor
- Experimental and Translational Medicine, University of Insubria, Via jean Henry Dunant 3, 21100 Varese, Italy
| | - Bahman Khalesi
- Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj 3197619751, Iran
| | - Navid Pourzardosht
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht 4193713111, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran 1678815811, Iran
| | - Zahra Payandeh
- Department of Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, SE-17177 Stockholm, Sweden
| |
Collapse
|
47
|
Mahmoud R, Ordóñez-Morán P, Allegrucci C. Challenges for Triple Negative Breast Cancer Treatment: Defeating Heterogeneity and Cancer Stemness. Cancers (Basel) 2022; 14:cancers14174280. [PMID: 36077812 PMCID: PMC9454775 DOI: 10.3390/cancers14174280] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/12/2022] [Accepted: 08/26/2022] [Indexed: 11/25/2022] Open
Abstract
The Triple Negative Breast Cancer (TNBC) subtype is known to have a more aggressive clinical course compared to other breast cancer subtypes. Targeted therapies for this type of breast cancer are limited and patients are mostly treated with conventional chemo- and radio-therapies which are not specific and do not target resistant cells. Therefore, one of the major clinical challenges is to find compounds that target the drug-resistant cell populations which are responsible for reforming secondary tumours. The molecular profiling of the different TNBC subtypes holds a promise for better defining these resistant cells specific to each tumour. To this end, a better understanding of TNBC heterogeneity and cancer stemness is required, and extensive genomic analysis can help to understand the disease complexity and distinguish new molecular drivers that can be targeted in the clinics. The use of persister cancer cell-targeting therapies combined with other therapies may provide a big advance to improve TNBC patients' survival.
Collapse
Affiliation(s)
- Rinad Mahmoud
- Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Paloma Ordóñez-Morán
- Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
- Translational Medical Sciences Unit, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
- Correspondence: (P.O.-M.); (C.A.)
| | - Cinzia Allegrucci
- Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
- SVMS, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK
- Correspondence: (P.O.-M.); (C.A.)
| |
Collapse
|
48
|
Cuccu A, Francescangeli F, De Angelis ML, Bruselles A, Giuliani A, Zeuner A. Analysis of Dormancy-Associated Transcriptional Networks Reveals a Shared Quiescence Signature in Lung and Colorectal Cancer. Int J Mol Sci 2022; 23:9869. [PMID: 36077264 PMCID: PMC9456317 DOI: 10.3390/ijms23179869] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022] Open
Abstract
Quiescent cancer cells (QCCs) are a common feature of solid tumors, representing a major obstacle to the long-term success of cancer therapies. We isolated QCCs ex vivo from non-small cell lung cancer (NSCLC) and colorectal cancer (CRC) xenografts with a label-retaining strategy and compared QCCs gene expression profiles to identify a shared "quiescence signature". Principal Component Analysis (PCA) revealed a specific component neatly discriminating quiescent and replicative phenotypes in NSCLC and CRC. The discriminating component showed significant overlapping, with 688 genes in common including ZEB2, a master regulator of stem cell plasticity and epithelial-to-mesenchymal transition (EMT). Gene set enrichment analysis showed that QCCs of both NSCLC and CRC had an increased expression of factors related to stemness/self renewal, EMT, TGF-β, morphogenesis, cell adhesion and chemotaxis, whereas proliferating cells overexpressed Myc targets and factors involved in RNA metabolism. Eventually, we analyzed in depth by means of a complex network approach, both the 'morphogenesis module' and the subset of differentially expressed genes shared by NCSLC and CRC. This allowed us to recognize different gene regulation network wiring for quiescent and proliferating cells and to underpin few genes central for network integration that may represent new therapeutic vulnerabilities. Altogether, our results highlight common regulatory pathways in QCCs of lung and colorectal tumors that may be the target of future therapeutic interventions.
Collapse
Affiliation(s)
- Adriano Cuccu
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Federica Francescangeli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Alessandro Bruselles
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Alessandro Giuliani
- Environment and Health Department, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Ann Zeuner
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
49
|
Tang Y, Kwiatkowski DJ, Henske EP. Midkine expression by stem-like tumor cells drives persistence to mTOR inhibition and an immune-suppressive microenvironment. Nat Commun 2022; 13:5018. [PMID: 36028490 PMCID: PMC9418323 DOI: 10.1038/s41467-022-32673-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/11/2022] [Indexed: 11/29/2022] Open
Abstract
mTORC1 is hyperactive in multiple cancer types1,2. Here, we performed integrative analysis of single cell transcriptomic profiling, paired T cell receptor (TCR) sequencing, and spatial transcriptomic profiling on Tuberous Sclerosis Complex (TSC) associated tumors with mTORC1 hyperactivity, and identified a stem-like tumor cell state (SLS) linked to T cell dysfunction via tumor-modulated immunosuppressive macrophages. Rapamycin and its derivatives (rapalogs) are the primary treatments for TSC tumors, and the stem-like tumor cells showed rapamycin resistance in vitro, reminiscent of the cytostatic effects of these drugs in patients. The pro-angiogenic factor midkine (MDK) was highly expressed by the SLS population, and associated with enrichment of endothelial cells in SLS-dominant samples. Inhibition of MDK showed synergistic benefit with rapamycin in reducing the growth of TSC cell lines in vitro and in vivo. In aggregate, this study suggests an autocrine rapamycin resistance mechanism and a paracrine tumor survival mechanism via immune suppression adopted by the stem-like state tumor cells with mTORC1 hyperactivity.
Collapse
Affiliation(s)
- Yan Tang
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David J Kwiatkowski
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Elizabeth P Henske
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
50
|
Properties of Leukemic Stem Cells in Regulating Drug Resistance in Acute and Chronic Myeloid Leukemias. Biomedicines 2022; 10:biomedicines10081841. [PMID: 36009388 PMCID: PMC9405586 DOI: 10.3390/biomedicines10081841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Notoriously known for their capacity to reconstitute hematological malignancies in vivo, leukemic stem cells (LSCs) represent key drivers of therapeutic resistance and disease relapse, posing as a major medical dilemma. Despite having low abundance in the bulk leukemic population, LSCs have developed unique molecular dependencies and intricate signaling networks to enable self-renewal, quiescence, and drug resistance. To illustrate the multi-dimensional landscape of LSC-mediated leukemogenesis, in this review, we present phenotypical characteristics of LSCs, address the LSC-associated leukemic stromal microenvironment, highlight molecular aberrations that occur in the transcriptome, epigenome, proteome, and metabolome of LSCs, and showcase promising novel therapeutic strategies that potentially target the molecular vulnerabilities of LSCs.
Collapse
|