1
|
Lou W, Zhang L, Wang J. Current status of nucleic acid therapy and its new progress in cancer treatment. Int Immunopharmacol 2024; 142:113157. [PMID: 39288629 DOI: 10.1016/j.intimp.2024.113157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/05/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Nucleic acid is an essential biopolymer in all living cells, performing the functions of storing and transmitting genetic information and synthesizing protein. In recent decades, with the progress of science and biotechnology and the continuous exploration of the functions performed by nucleic acid, more and more studies have confirmed that nucleic acid therapy for living organisms has great medical therapeutic potential. Nucleic acid drugs began to become independent therapeutic agents. As a new therapeutic method, nucleic acid therapy plays an important role in the treatment of genetic diseases, viral infections and cancers. There are currently 19 nucleic acid drugs approved by the Food and Drug Administration (FDA). In the following review, we start from principles and advantages of nucleic acid therapy, and briefly describe development history of nucleic acid drugs. And then we give examples of various RNA therapeutic drugs, including antisense oligonucleotides (ASO), mRNA vaccines, small interfering RNA (siRNA) and microRNA (miRNA), aptamers, and small activating RNA (saRNA). In addition, we also focused on the current status of nucleic acid drugs used in cancer therapy and the breakthrough in recent years. Clinical trials of nucleic acid drugs for cancer treatment are under way, conventional radiotherapy and chemotherapy combined with the immunotherapies such as checkpoint inhibitors and nucleic acid drugs may be the main prospects for successful cancer treatment.
Collapse
Affiliation(s)
- Wenting Lou
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Leqi Zhang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Jianwei Wang
- Department of Surgery, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China; Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, 2nd Affiliated Hospital, Zhejiang University School of Medicine, Jiefang Road 88th, Hangzhou 310009, China.
| |
Collapse
|
2
|
Miller PG, Huang E, Fisher R, Shuler ML. Development of a Microphysiological System to Model Human Cancer Metastasis From the Colon to the Liver. Biotechnol Bioeng 2024. [PMID: 39587032 DOI: 10.1002/bit.28890] [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: 07/28/2024] [Revised: 11/05/2024] [Accepted: 11/10/2024] [Indexed: 11/27/2024]
Abstract
We describe a novel device to mimic the metastasis of cancer cells from the colon into the liver in a human model. The colon mimic is connected to the liver model by a gravity-driven recirculating unidirectional flow of a blood surrogate and can mimic the five steps of the metastatic cascade: invasion in the colon, intravasation into the bloodstream, systemic transportation, extravasation into the liver, and colonization in the liver. The colon mimic uses established normal colon epithelial organoid cells (NL) and human umbilical vein endothelial cells (HUVEC) plated on opposite sides of a membrane. To better mimic the colon structure the NL side of the membrane is exposed to air to establish an air-liquid interface. The liver mimic consists of human liver sinusoidal endothelial cells (HHSEC) and epithelial hepatic cells (HepG2 C3A) plated in Matrigel on opposite sides of a membrane. Labeled colorectal cancer cells/clusters (CA) from organoids are introduced into an established normal colon epithelial cell (NL) layer from the same patient before assembly of the system or alternatively NL organoids and fluorescently labeled CA organoids from the same patient were prepared as a ratio of 10:1 NL:CA and established together before assembly of the system. Cell viability is greater than 85% in this system. We demonstrate that over 5 days of operation that the five steps of the metastatic cascade are replicated. This novel device allows an in vitro estimate of metastatic capability (as measured by using percentages of the labeled areas per device through ImageJ) in response to selected variables. In this study, the metastatic capability depends on the source of cancer cells (e.g., the patient), the clumping of cancer cells, glucose concentration, and oxygen levels (hypoxia). For the first time, this new in vitro system mimics all five steps of the metastatic cascade in a single device and provides a new device to probe and observe the process of metastasis in a human-based model in only 5 days. The rapid observation is due to the use of a high concentration of cancer cells in the colon (e.g. 10%) and the absence of the immune system. Our device makes it possible to probe aspects of each step of metastasis and interactions between steps.
Collapse
Affiliation(s)
- Paula G Miller
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Emina Huang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Robert Fisher
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Michael L Shuler
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
3
|
Mayasin YP, Osinnikova MN, Kharisova CB, Kitaeva KV, Filin IY, Gorodilova AV, Kutovoi GI, Solovyeva VV, Golubev AI, Rizvanov AA. Extracellular Matrix as a Target in Melanoma Therapy: From Hypothesis to Clinical Trials. Cells 2024; 13:1917. [PMID: 39594665 PMCID: PMC11592585 DOI: 10.3390/cells13221917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/10/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Melanoma is a malignant, highly metastatic neoplasm showing increasing morbidity and mortality. Tumor invasion and angiogenesis are based on remodeling of the extracellular matrix (ECM). Selective inhibition of functional components of cell-ECM interaction, such as hyaluronic acid (HA), matrix metalloproteinases (MMPs), and integrins, may inhibit tumor progression and enhance the efficacy of combination treatment with immune checkpoint inhibitors (ICIs), chemotherapy, or immunotherapy. In this review, we combine the results of different approaches targeting extracellular matrix elements in melanoma in preclinical and clinical studies. The identified limitations of many approaches, including side effects, low selectivity, and toxicity, indicate the need for further studies to optimize therapy. Nevertheless, significant progress in expanding our understanding of tumor biology and the development of targeted therapies holds great promise for the early approaches developed several decades ago to inhibit metastasis through ECM targeting.
Collapse
Affiliation(s)
- Yuriy P. Mayasin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Y.P.M.); (M.N.O.); (C.B.K.); (K.V.K.); (I.Y.F.); (A.V.G.); (G.I.K.); (V.V.S.); (A.I.G.)
| | - Maria N. Osinnikova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Y.P.M.); (M.N.O.); (C.B.K.); (K.V.K.); (I.Y.F.); (A.V.G.); (G.I.K.); (V.V.S.); (A.I.G.)
| | - Chulpan B. Kharisova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Y.P.M.); (M.N.O.); (C.B.K.); (K.V.K.); (I.Y.F.); (A.V.G.); (G.I.K.); (V.V.S.); (A.I.G.)
| | - Kristina V. Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Y.P.M.); (M.N.O.); (C.B.K.); (K.V.K.); (I.Y.F.); (A.V.G.); (G.I.K.); (V.V.S.); (A.I.G.)
| | - Ivan Y. Filin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Y.P.M.); (M.N.O.); (C.B.K.); (K.V.K.); (I.Y.F.); (A.V.G.); (G.I.K.); (V.V.S.); (A.I.G.)
| | - Anna V. Gorodilova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Y.P.M.); (M.N.O.); (C.B.K.); (K.V.K.); (I.Y.F.); (A.V.G.); (G.I.K.); (V.V.S.); (A.I.G.)
| | - Grigorii I. Kutovoi
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Y.P.M.); (M.N.O.); (C.B.K.); (K.V.K.); (I.Y.F.); (A.V.G.); (G.I.K.); (V.V.S.); (A.I.G.)
| | - Valeriya V. Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Y.P.M.); (M.N.O.); (C.B.K.); (K.V.K.); (I.Y.F.); (A.V.G.); (G.I.K.); (V.V.S.); (A.I.G.)
| | - Anatolii I. Golubev
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Y.P.M.); (M.N.O.); (C.B.K.); (K.V.K.); (I.Y.F.); (A.V.G.); (G.I.K.); (V.V.S.); (A.I.G.)
| | - Albert A. Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (Y.P.M.); (M.N.O.); (C.B.K.); (K.V.K.); (I.Y.F.); (A.V.G.); (G.I.K.); (V.V.S.); (A.I.G.)
- Division of Medical and Biological Sciences, Tatarstan Academy of Sciences, 420111 Kazan, Russia
| |
Collapse
|
4
|
Santiago TSA, Delezuk JAM, Bataglioli RA, Baratti MO, Carvalho HF, Beppu MM. Influence of hyaluronic acid and chitosan molecular weight on the adhesion of circulating tumor cell on multilayer films. Int J Biol Macromol 2024; 281:136180. [PMID: 39357715 DOI: 10.1016/j.ijbiomac.2024.136180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/25/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
CD44 is a cell receptor glycoprotein overexpressed in circulating tumor cells (CTCs), with levels linked to an increase in metastatic capacity of several tumors. Hyaluronic acid (HA), the natural ligand of CD44, has primarily been investigated for tumor cell interaction in self-assembled polyelectrolyte multilayer films, with little attention given to the complementary polycation. In this study, we screened sixteen different polyelectrolyte multilayer assemblies of HA and chitosan (CHI) to identify key assembly parameters and surface properties that control and govern CTCs adhesion. Statistics analysis revealed a major role of CHI molecular weight in the adhesion, followed by its combinatorial response either with HA ionization degree or ionic strength. PM-IRRAS analysis demonstrated a correlation between the orientation of HA carboxyl groups on the film surface and CTCs adhesion, directly impacted by CHI molecular weight. Overall, although CTCs binding onto the surface of multilayer films is primarily driven by HA-CD44 interaction, both chitosan properties and film assembly conditions modulate this interaction. These findings illustrate an alternative to modifying the performance of biomaterials with minimal changes in the composition of multilayer films.
Collapse
Affiliation(s)
- T S A Santiago
- Universidade Estadual de Campinas, School of Chemical Engineering, Department of Materials Engineering and Bioprocesses, 500 Albert Einstein Ave, Campinas 13083-852, Brazil
| | - J A M Delezuk
- Universidade Estadual de Campinas, School of Chemical Engineering, Department of Materials Engineering and Bioprocesses, 500 Albert Einstein Ave, Campinas 13083-852, Brazil
| | - R A Bataglioli
- Universidade Estadual de Campinas, School of Chemical Engineering, Department of Materials Engineering and Bioprocesses, 500 Albert Einstein Ave, Campinas 13083-852, Brazil
| | - M O Baratti
- Universidade Estadual de Campinas, National Institute of Photonics Applied to Cell Biology, Carl Von Linaeus St, Campinas 13083-864, Brazil
| | - H F Carvalho
- Universidade Estadual de Campinas, National Institute of Photonics Applied to Cell Biology, Carl Von Linaeus St, Campinas 13083-864, Brazil; Universidade Estadual de Campinas, Department of Structural and Functional Biology, Bertrand Russel Ave, Campinas 13083-865, Brazil
| | - M M Beppu
- Universidade Estadual de Campinas, School of Chemical Engineering, Department of Materials Engineering and Bioprocesses, 500 Albert Einstein Ave, Campinas 13083-852, Brazil..
| |
Collapse
|
5
|
Riana GM, Pelupessy NU, Qadar S, Miskad U, Zainuddin AA, Madya F, Lukas E. Association of high expression of CD44 in clinicopathological factors of endometrial cancer. Minerva Obstet Gynecol 2024; 76:410-415. [PMID: 36912890 DOI: 10.23736/s2724-606x.23.05279-x] [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: 03/14/2023]
Abstract
BACKGROUND Clinical stages, histologic type, degree of cell differentiation, myometrial invasion, and lymph-vascular space invasion (LVSI) have been identified as clinicopathological factors that are predictive for endometrial cancer, however, further prognostic indicators are still required to account for the heterogeneity of this cancer. Adhesion molecule CD44, affects the invasion, metastasis, and prognosis of many forms of cancer. The purpose of this study is to examine the expression of CD44 in endometrial cancer and its correlation with established prognostic variables. METHODS A cross-sectional study was conducted on 64 samples of endometrial cancer from Wahidin Sudirohusodo Hospital and Hasanuddin University Hospital. Immunohistochemical analysis was used to detect CD44 expression using mouse anti-human CD44 monoclonal antibody. Differences in Histoscore were studied to determine the association between CD44 expression and clinicopathological factors of endometrial cancer. RESULTS Of the overall sample, 46 samples were in the early stage, whereas 18 samples were in the advanced stage. High expression of CD44 was associated with advanced stage compare than early stage (P=0.010), poor differentiation compare than well-moderate differentiation (P=0.001), myometrial invasion ≥50% compare than myometrial invasion <50% (P=0.004), and positive LVSI compare than negative LVSI (P=0.043) in endometrial cancer, but not associated with histological type of endometrial cancer (P=0.178). CONCLUSIONS High expression of CD44 may be considered as a poor prognostic marker and predictive marker for targeted therapy in endometrial cancer.
Collapse
Affiliation(s)
- Gina M Riana
- Department of Obstetrics and Gynecology, Hasanuddin University, Makassar, Indonesia
| | - Nugraha U Pelupessy
- Department of Obstetrics and Gynecology, Hasanuddin University, Makassar, Indonesia -
| | - Sriwijaya Qadar
- Department of Obstetrics and Gynecology, Hasanuddin University, Makassar, Indonesia
| | - Upik Miskad
- Department of Pathology Anatomy, Hasanuddin University, Makassar, Indonesia
| | - Andi A Zainuddin
- Department of Public Health, Hasanuddin University, Makassar, Indonesia
| | - Fatmawati Madya
- Department of Obstetrics and Gynecology, Hasanuddin University, Makassar, Indonesia
| | - Efendi Lukas
- Department of Obstetrics and Gynecology, Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
6
|
Awuah WA, Ben-Jaafar A, Karkhanis S, Nkrumah-Boateng PA, Kong JSH, Mannan KM, Shet V, Imran S, Bone M, Boye ANA, Ranganathan S, Shah MH, Abdul-Rahman T, Atallah O. Cancer stem cells in meningiomas: novel insights and therapeutic implications. Clin Transl Oncol 2024:10.1007/s12094-024-03728-6. [PMID: 39316249 DOI: 10.1007/s12094-024-03728-6] [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: 08/21/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024]
Abstract
Meningiomas (MGs), which arise from meningothelial cells of the dura mater, represent a significant proportion of primary tumours of the central nervous system (CNS). Despite advances in treatment, the management of malignant meningioma (MMG) remains challenging due to diagnostic, surgical, and resection limitations. Cancer stem cells (CSCs), a subpopulation within tumours capable of self-renewal and differentiation, are highlighted as key markers of tumour growth, metastasis, and treatment resistance. Identifying additional CSC-related markers enhances the precision of malignancy evaluations, enabling advancements in personalised medicine. The review discusses key CSC biomarkers that are associated with high levels of expression, aggressive tumour behaviour, and poor outcomes. Recent molecular research has identified CSC-related biomarkers, including Oct-4, Sox2, NANOG, and CD133, which help maintain cellular renewal, proliferation, and drug resistance in MGs. This study highlights new therapeutic strategies that could improve patient prognosis with more durable tumour regression. The use of combination therapies, such as hydroxyurea alongside diltiazem, suggests more efficient and effective MG management compared to monotherapy. Signalling pathways such as NOTCH and hedgehog also offer additional avenues for therapeutic development. CRISPR/Cas9 technology has also been employed to create meningioma models, uncovering pathways related to cell growth and proliferation. Since the efficacy of traditional therapies is limited in most cases due to resistance mechanisms in CSCs, further studies on the biology of CSCs are warranted to develop therapeutic interventions that are likely to be effective in MG. Consequently, improved diagnostic approaches may lead to personalised treatment plans tailored to the specific needs of each patient.
Collapse
Affiliation(s)
| | - Adam Ben-Jaafar
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | - Jonathan Sing Huk Kong
- School of Medicine, College of Medical & Veterinary Life Sciences, University of Glasgow, Glasgow, UK
| | - Krishitha Meenu Mannan
- School of Medicine, Queen's University Belfast, Dentistry & Biomedical Sciences, Belfast, UK
| | - Vallabh Shet
- University of Connecticut New Britain Program, New Britain, Connecticut, USA
| | - Shahzeb Imran
- School of Medicine, Queen's University Belfast, Dentistry & Biomedical Sciences, Belfast, UK
| | - Matan Bone
- Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford, UK
| | | | | | | | | | - Oday Atallah
- Department of Neurosurgery, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| |
Collapse
|
7
|
Alimohammadvand S, Kaveh Zenjanab M, Mashinchian M, Shayegh J, Jahanban-Esfahlan R. Recent advances in biomimetic cell membrane-camouflaged nanoparticles for cancer therapy. Biomed Pharmacother 2024; 177:116951. [PMID: 38901207 DOI: 10.1016/j.biopha.2024.116951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024] Open
Abstract
The emerging strategy of biomimetic nanoparticles (NPs) via cellular membrane camouflage holds great promise in cancer therapy. This scholarly review explores the utilization of cellular membranes derived from diverse cellular entities; blood cells, immune cells, cancer cells, stem cells, and bacterial cells as examples of NP coatings. The camouflaging strategy endows NPs with nuanced tumor-targeting abilities such as self-recognition, homotypic targeting, and long-lasting circulation, thus also improving tumor therapy efficacy overall. The comprehensive examination encompasses a variety of cell membrane camouflaged NPs (CMCNPs), elucidating their underlying targeted therapy mechanisms and delineating diverse strategies for anti-cancer applications. Furthermore, the review systematically presents the synthesis of source materials and methodologies employed in order to construct and characterize these CMCNPs, with a specific emphasis on their use in cancer treatment.
Collapse
Affiliation(s)
- Sajjad Alimohammadvand
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoumeh Kaveh Zenjanab
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Mashinchian
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Shayegh
- Department of Microbiology, Faculty of Veterinary and Agriculture, Islamic Azad University, Shabestar branch, Shabestar, Iran
| | - Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
8
|
Vasilevska J, Cheng PF, Lehmann J, Ramelyte E, Gómez JM, Dimitriou F, Sella F, Ferretti D, Salas-Bastos A, Jordaan WS, Levesque MP, Dummer R, Sommer L. Monitoring melanoma patients on treatment reveals a distinct macrophage population driving targeted therapy resistance. Cell Rep Med 2024; 5:101611. [PMID: 38942020 PMCID: PMC11293307 DOI: 10.1016/j.xcrm.2024.101611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 04/09/2024] [Accepted: 05/20/2024] [Indexed: 06/30/2024]
Abstract
Resistance to targeted therapy remains a major clinical challenge in melanoma. To uncover resistance mechanisms, we perform single-cell RNA sequencing on fine-needle aspirates from resistant and responding tumors of patients undergoing BRAFi/MEKi treatment. Among the genes most prominently expressed in resistant tumors is POSTN, predicted to signal to a macrophage population associated with targeted therapy resistance (TTR). Accordingly, tumors from patients with fast disease progression after therapy exhibit high POSTN expression levels and high numbers of TTR macrophages. POSTN polarizes human macrophages toward a TTR phenotype and promotes resistance to targeted therapy in a melanoma mouse model, which is associated with a phenotype change in intratumoral macrophages. Finally, polarized TTR macrophages directly protect human melanoma cells from MEKi-induced killing via CD44 receptor expression on melanoma cells. Thus, interfering with the protective activity of TTR macrophages may offer a strategy to overcome resistance to targeted therapy in melanoma.
Collapse
Affiliation(s)
- Jelena Vasilevska
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland
| | - Phil Fang Cheng
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Julia Lehmann
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Julia Martínez Gómez
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Florentia Dimitriou
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Federica Sella
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Daria Ferretti
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland
| | | | | | - Mitchell Paul Levesque
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University of Zurich Hospital and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Lukas Sommer
- Institute of Anatomy, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
9
|
Kildal W, Cyll K, Kalsnes J, Islam R, Julbø FM, Pradhan M, Ersvær E, Shepherd N, Vlatkovic L, Tekpli X, Garred Ø, Kristensen GB, Askautrud HA, Hveem TS, Danielsen HE. Deep learning for automated scoring of immunohistochemically stained tumour tissue sections - Validation across tumour types based on patient outcomes. Heliyon 2024; 10:e32529. [PMID: 39040241 PMCID: PMC11261074 DOI: 10.1016/j.heliyon.2024.e32529] [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: 05/26/2024] [Accepted: 06/05/2024] [Indexed: 07/24/2024] Open
Abstract
We aimed to develop deep learning (DL) models to detect protein expression in immunohistochemically (IHC) stained tissue-sections, and to compare their accuracy and performance with manually scored clinically relevant proteins in common cancer types. Five cancer patient cohorts (colon, two prostate, breast, and endometrial) were included. We developed separate DL models for scoring IHC-stained tissue-sections with nuclear, cytoplasmic, and membranous staining patterns. For training, we used images with annotations of cells with positive and negative staining from the colon cohort stained for Ki-67 and PMS2 (nuclear model), the prostate cohort 1 stained for PTEN (cytoplasmic model) and β-catenin (membranous model). The nuclear DL model was validated for MSH6 in the colon, MSH6 and PMS2 in the endometrium, Ki-67 and CyclinB1 in prostate, and oestrogen and progesterone receptors in the breast cancer cohorts. The cytoplasmic DL model was validated for PTEN and Mapre2, and the membranous DL model for CD44 and Flotillin1, all in prostate cohorts. When comparing the results of manual and DL scores in the validation sets, using manual scores as the ground truth, we observed an average correct classification rate of 91.5 % (76.9-98.5 %) for the nuclear model, 85.6 % (73.3-96.6 %) for the cytoplasmic model, and 78.4 % (75.5-84.3 %) for the membranous model. In survival analyses, manual and DL scores showed similar prognostic impact, with similar hazard ratios and p-values for all DL models. Our findings demonstrate that DL models offer a promising alternative to manual IHC scoring, providing efficiency and reproducibility across various data sources and markers.
Collapse
Affiliation(s)
- Wanja Kildal
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Karolina Cyll
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Joakim Kalsnes
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Rakibul Islam
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Frida M. Julbø
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Manohar Pradhan
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Elin Ersvær
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Neil Shepherd
- Gloucestershire Cellular Pathology Laboratory, Gloucester, GL53 7AN, UK
| | - Ljiljana Vlatkovic
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - OSBREAC
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
- Gloucestershire Cellular Pathology Laboratory, Gloucester, GL53 7AN, UK
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital, NO-0450, Oslo, Norway
- Department of Pathology, Oslo University Hospital, NO-0424, Oslo, Norway
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, OX3 9DU, UK
| | - Xavier Tekpli
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital, NO-0450, Oslo, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Gunnar B. Kristensen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Hanne A. Askautrud
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Tarjei S. Hveem
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Håvard E. Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, OX3 9DU, UK
| |
Collapse
|
10
|
Ziranu P, Pretta A, Aimola V, Cau F, Mariani S, D’Agata AP, Codipietro C, Rizzo D, Dell’Utri V, Sanna G, Moledda G, Cadoni A, Lai E, Puzzoni M, Pusceddu V, Castagnola M, Scartozzi M, Faa G. CD44: A New Prognostic Marker in Colorectal Cancer? Cancers (Basel) 2024; 16:1569. [PMID: 38672650 PMCID: PMC11048923 DOI: 10.3390/cancers16081569] [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: 01/18/2024] [Revised: 03/19/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Cluster of differentiation 44 (CD44) is a non-kinase cell surface glycoprotein. It is overexpressed in several cell types, including cancer stem cells (CSCs). Cells overexpressing CD44 exhibit several CSC traits, such as self-renewal, epithelial-mesenchymal transition (EMT) capability, and resistance to chemo- and radiotherapy. The role of CD44 in maintaining stemness and the CSC function in tumor progression is accomplished by binding to its main ligand, hyaluronan (HA). The HA-CD44 complex activates several signaling pathways that lead to cell proliferation, adhesion, migration, and invasion. The CD44 gene regularly undergoes alternative splicing, resulting in the standard (CD44s) and variant (CD44v) isoforms. The different functional roles of CD44s and specific CD44v isoforms still need to be fully understood. The clinicopathological impact of CD44 and its isoforms in promoting tumorigenesis suggests that CD44 could be a molecular target for cancer therapy. Furthermore, the recent association observed between CD44 and KRAS-dependent carcinomas and the potential correlations between CD44 and tumor mutational burden (TMB) and microsatellite instability (MSI) open new research scenarios for developing new strategies in cancer treatment. This review summarises current research regarding the different CD44 isoform structures, their roles, and functions in supporting tumorigenesis and discusses its therapeutic implications.
Collapse
Affiliation(s)
- Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Valentina Aimola
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (V.A.); (F.C.)
| | - Flaviana Cau
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (V.A.); (F.C.)
| | - Stefano Mariani
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Alessandra Pia D’Agata
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Claudia Codipietro
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Daiana Rizzo
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Veronica Dell’Utri
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Giorgia Sanna
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Giusy Moledda
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Andrea Cadoni
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Eleonora Lai
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Marco Puzzoni
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Valeria Pusceddu
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Massimo Castagnola
- Proteomics Laboratory, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, 00013 Rome, Italy;
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, SS 554 km 4500 Bivio per Sestu, Monserrato, 09042 Cagliari, Italy; (A.P.); (S.M.); (A.P.D.); (C.C.); (D.R.); (V.D.); (G.S.); (G.M.); (A.C.); (E.L.); (M.P.); (V.P.); (M.S.)
| | - Gavino Faa
- Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy;
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
11
|
Bartkowski M, Bincoletto V, Salaroglio IC, Ceccone G, Arenal R, Nervo S, Rolando B, Riganti C, Arpicco S, Giordani S. Enhancing pancreatic ductal adenocarcinoma (PDAC) therapy with targeted carbon nano-onion (CNO)-mediated delivery of gemcitabine (GEM)-derived prodrugs. J Colloid Interface Sci 2024; 659:339-354. [PMID: 38176243 DOI: 10.1016/j.jcis.2023.12.166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
Nanotechnology's potential in revolutionising cancer treatments is evident in targeted drug delivery systems (DDSs) engineered to optimise therapeutic efficacy and minimise toxicity. This study examines a novel nanocarrier constructed with carbon nano-onions (CNOs), engineered and evaluated for its ability to selectively target cancer cells overexpressing the hyaluronic acid receptor; CD44. Our results highlighted that the CNO-based nanocarrier coupled with hyaluronic acid as the targeting agent demonstrated effective uptake by CD44+ PANC-1 and MIA PaCa-2 cells, while avoiding CD44- Capan-1 cells. The CNO-based nanocarrier also exhibited excellent biocompatibility in all tested pancreatic ductal adenocarcinoma (PDAC) cells, as well as healthy cells. Notably, the CNO-based nanocarrier was successfully loaded with chemotherapeutic 4-(N)-acyl- sidechain-containing prodrugs derived from gemcitabine (GEM). These prodrugs alone exhibited remarkable efficacy in killing PDAC cells which are known to be GEM resistant, and their efficacy was amplified when combined with the CNO-based nanocarrier, particularly in targeting GEM-resistant CD44+ PDAC cells. These findings demonstrate the potential of CNOs as promising scaffolds in advancing targeted DDSs, signifying the translational potential of carbon nanoparticles for cancer therapy.
Collapse
Affiliation(s)
- Michał Bartkowski
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Valeria Bincoletto
- Department of Drug Science and Technology, University of Torino, Via P. Giuria 9, Torino, Italy
| | | | | | - Raul Arenal
- Instituto de Nanociencia y Materiales de Aragon (INMA), CSIC-U. de Zaragoza, 50009 Zaragoza, Spain; Laboratorio de Microscopias Avanzadas (LMA), Universidad de Zaragoza, 50018 Zaragoza, Spain; ARAID Foundation, 50018 Zaragoza, Spain
| | - Sara Nervo
- Department of Drug Science and Technology, University of Torino, Via P. Giuria 9, Torino, Italy
| | - Barbara Rolando
- Department of Drug Science and Technology, University of Torino, Via P. Giuria 9, Torino, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Via Nizza 44, Torino, Italy; Molecular Biotechnology Center "Guido Tarone", University of Torino, Italy
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Torino, Via P. Giuria 9, Torino, Italy
| | - Silvia Giordani
- School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland.
| |
Collapse
|
12
|
Zhang Q, Wang X, Liu Y, Xu H, Ye C. Pan-cancer and single-cell analyses identify CD44 as an immunotherapy response predictor and regulating macrophage polarization and tumor progression in colorectal cancer. Front Oncol 2024; 14:1380821. [PMID: 38590654 PMCID: PMC10999581 DOI: 10.3389/fonc.2024.1380821] [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: 02/02/2024] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
Introduction Cluster of differentiation (CD) 44 is a non-kinase cell surface transmembrane glycoprotein critical for tumor maintenance and progression. Methods We conducted a systematic analysis of the expression profile and genomic alteration profile of CD44 in 33 types of cancer. The immune characteristics of CD44 were comprehensively explored by TIMER2.0 and CIBERSORT. In addition, the CD44 transcriptional landscape was examined at the single-cell level. Then, Pseudotime trajectory analysis of CD44 gene expression was performed using Monocle 2, and CellChat was utilized to compare the crosstalk differences between CD44+monocytes and CD44- monocytes. Tumor immune dysfunction and exclusion (TIDE) was used to evaluate the predictive ability of CD44 for immune checkpoint blockade (ICB) responses. The effects of CD44 on colorectal cancer (CRC) and macrophage polarization were investigated by knocking down the expression of CD44 in HCT-116 cell and macrophages in vitro. Results The expression of CD44 elevated in most cancers, predicting unfavorable prognosis. In addditon, CD44 was correlation with immune cell infiltration and key immune regulators. CD44+ monocytes had a higher information flow intensity than CD44- monocytes. CD44 had good predictive ability for immune checkpoint blockade responses. Knockdown of CD44 inhibited the proliferation, migration, and invasion of HCT-116 cell in vitro. Knockdown of CD44 inhibited M2 macrophage polarization. Discussion These findings suggest that CD44 is involved in regulating tumor development, macrophage polarization, and has certain predictive value for patient clinical prognosis and response to immunotherapy.
Collapse
Affiliation(s)
- Qian Zhang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, China
- Department of General Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Xinyu Wang
- Department of General Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Yang Liu
- Department of Pharmacy, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Hao Xu
- Department of General Surgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Chun Ye
- Department of General Surgery, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
13
|
De Fazio E, Pittarello M, Gans A, Ghosh B, Slika H, Alimonti P, Tyler B. Intrinsic and Microenvironmental Drivers of Glioblastoma Invasion. Int J Mol Sci 2024; 25:2563. [PMID: 38473812 PMCID: PMC10932253 DOI: 10.3390/ijms25052563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/07/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Gliomas are diffusely infiltrating brain tumors whose prognosis is strongly influenced by their extent of invasion into the surrounding brain tissue. While lower-grade gliomas present more circumscribed borders, high-grade gliomas are aggressive tumors with widespread brain infiltration and dissemination. Glioblastoma (GBM) is known for its high invasiveness and association with poor prognosis. Its low survival rate is due to the certainty of its recurrence, caused by microscopic brain infiltration which makes surgical eradication unattainable. New insights into GBM biology at the single-cell level have enabled the identification of mechanisms exploited by glioma cells for brain invasion. In this review, we explore the current understanding of several molecular pathways and mechanisms used by tumor cells to invade normal brain tissue. We address the intrinsic biological drivers of tumor cell invasion, by tackling how tumor cells interact with each other and with the tumor microenvironment (TME). We focus on the recently discovered neuronal niche in the TME, including local as well as distant neurons, contributing to glioma growth and invasion. We then address the mechanisms of invasion promoted by astrocytes and immune cells. Finally, we review the current literature on the therapeutic targeting of the molecular mechanisms of invasion.
Collapse
Affiliation(s)
- Emerson De Fazio
- Department of Medicine, Vita-Salute San Raffaele University School of Medicine, 20132 Milan, Italy; (E.D.F.); (P.A.)
| | - Matilde Pittarello
- Department of Medicine, Humanitas University School of Medicine, 20089 Rozzano, Italy;
| | - Alessandro Gans
- Department of Neurology, University of Milan, 20122 Milan, Italy;
| | - Bikona Ghosh
- School of Medicine and Surgery, Dhaka Medical College, Dhaka 1000, Bangladesh;
| | - Hasan Slika
- Hunterian Neurosurgical Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| | - Paolo Alimonti
- Department of Medicine, Vita-Salute San Raffaele University School of Medicine, 20132 Milan, Italy; (E.D.F.); (P.A.)
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Betty Tyler
- Hunterian Neurosurgical Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| |
Collapse
|
14
|
Gui J, Yang L, Liu J, Li Y, Zou M, Sun C, Huang L, Zhu X, Huang K. Identifying the prognosis implication, immunotherapy response prediction value, and potential targeted compound inhibitors of integrin subunit α3 (ITGA3) in human cancers. Heliyon 2024; 10:e24236. [PMID: 38293430 PMCID: PMC10825359 DOI: 10.1016/j.heliyon.2024.e24236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 11/30/2023] [Accepted: 01/04/2024] [Indexed: 02/01/2024] Open
Abstract
The integrin subunit α3 (ITGA3) is a member of the integrin alpha chain protein family, which could promote progression, metastasis, and invasion in some cancers. Still, its function in the tumor microenvironment (TME), cancer prognosis, and immunotherapy remains unclear. A multifaceted analysis of ITGA3 in pan-cancer utilizing various databases and online web tools revealed ITGA3 was aberrantly expressed in tumor tissues and upregulated in most cancers, which may be related to ITGA3 genomic alterations and methylation modification. In addition, ITGA3 was significantly correlated with the poor or better prognosis of cancer patients, immune-related pathways in hallmark, immune infiltration, and immune checkpoints, revealing a biological function of ITGA3 in the tumor progression, tumor microenvironment, and tumor immunity. We also found that ITGA3 could predict the response to tumor immunotherapy based on cytokine-treated samples and immunotherapy cohorts. ITGA3 may participate in shaping and regulating the tumor microenvironment to affect the tumor immune response, which was a promising immunotherapy response predictive biomarker and potential therapeutic target to work synergistically with cancer immunotherapy to boost the response and efficacy. Finally, potential targeted compound inhibitors and sensitive drugs were screened using databases ConnectivityMap (CMap) and CellMiner, and AutoDock Tools was used for molecular docking.
Collapse
Affiliation(s)
- Jiawei Gui
- Department of Neurosurgery, The 2 Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
- HuanKui Academy, Jiangxi Medical College, Nanchang University, Nanchang 330031, PR China
| | - Lufei Yang
- Department of Neurosurgery, The 2 Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, PR China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, 330006, Nanchang, PR China
| | - Junzhe Liu
- Department of Neurosurgery, The 2 Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, PR China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, 330006, Nanchang, PR China
| | - Yishuang Li
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, PR China
| | - Mi Zou
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang 330031, PR China
| | - Chengpeng Sun
- HuanKui Academy, Jiangxi Medical College, Nanchang University, Nanchang 330031, PR China
| | - Le Huang
- HuanKui Academy, Jiangxi Medical College, Nanchang University, Nanchang 330031, PR China
| | - Xingen Zhu
- Department of Neurosurgery, The 2 Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, PR China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, 330006, Nanchang, PR China
| | - Kai Huang
- Department of Neurosurgery, The 2 Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
- Institute of Neuroscience, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, PR China
- Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, PR China
- JXHC Key Laboratory of Neurological Medicine, Jiangxi, 330006, Nanchang, PR China
| |
Collapse
|
15
|
Boman BM, Viswanathan V, Facey COB, Fields JZ, Stave JW. The v8-10 variant isoform of CD44 is selectively expressed in the normal human colonic stem cell niche and frequently is overexpressed in colon carcinomas during tumor development. Cancer Biol Ther 2023; 24:2195363. [PMID: 37005380 PMCID: PMC10072056 DOI: 10.1080/15384047.2023.2195363] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/02/2023] [Accepted: 03/20/2023] [Indexed: 04/04/2023] Open
Abstract
CD44 protein and its variant isoforms are expressed in cancer stem cells (CSCs), and various CD44 isoforms can have different functional roles in cells. Our goal was to investigate how different CD44 isoforms contribute to the emergence of stem cell (SC) overpopulation that drives colorectal cancer (CRC) development. Specific CD44 variant isoforms are selectively expressed in normal colonic SCs and become overexpressed in CRCs during tumor development. We created a unique panel of anti-CD44 rabbit genomic antibodies to 16 specific epitopes that span the entire length of the CD44 molecule. Our panel was used to comprehensively investigate the expression of different CD44 isoforms in matched pairs (n = 10) of malignant colonic tissue and adjacent normal mucosa, using two (IHC & IF) immunostaining approaches. We found that: i) CD44v8-10 is selectively expressed in the normal human colonic SC niche; ii) CD44v8-10 is co-expressed with the SC markers ALDH1 and LGR5 in normal and malignant colon tissues; iii) colon carcinoma tissues frequently (80%) stain for CD44v8-10 while staining for CD44v6 was less frequent (40%). Given that CD44v8-10 expression is restricted to cells in the normal human colonic SC niche and CD44v8-10 expression progressively increases during CRC development, CD44v8-10 expression likely contributes to the SC overpopulation that drives the development and growth of colon cancers. Since the CD44 variant v8-10 epitope is located on CD44's extracellular region, it offers great promise for targeted anti-CSC treatment approaches.
Collapse
Affiliation(s)
- Bruce M. Boman
- Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE, USA
- Department of Biologic Sciences, University of Delaware, Newark, DE, USA
- Jefferson Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Vignesh Viswanathan
- Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE, USA
- Department of Biologic Sciences, University of Delaware, Newark, DE, USA
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Caroline O. B. Facey
- Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE, USA
| | - Jeremy Z. Fields
- Center for Translational Cancer Research, Helen F. Graham Cancer Center & Research Institute, Newark, DE, USA
- Department of Cancer Research and Innovation, CA*TX Inc, Princeton, NJ, USA
| | - James W. Stave
- Department of Cancer Research and Innovation, Strategic Diagnostics Inc, Newark, DE, USA
| |
Collapse
|
16
|
Zhang X, Yi Y, Jiang Y, Liao J, Yang R, Deng X, Zhang L. Targeted Therapy of Acute Liver Injury via Cryptotanshinone-Loaded Biomimetic Nanoparticles Derived from Mesenchymal Stromal Cells Driven by Homing. Pharmaceutics 2023; 15:2764. [PMID: 38140104 PMCID: PMC10747007 DOI: 10.3390/pharmaceutics15122764] [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: 10/24/2023] [Revised: 11/16/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Acute liver injury (ALI) has the potential to compromise hepatic function rapidly, with severe cases posing a considerable threat to human health and wellbeing. Conventional treatments, such as the oral administration of antioxidants, can inadvertently lead to liver toxicity and other unwanted side effects. Mesenchymal stromal cells (MSCs) can target therapeutic agents directly to inflammatory sites owing to their homing effect, and they offer a promising avenue for the treatment of ALI. However, the efficacy and feasibility of these live cell products are hampered by challenges associated with delivery pathways and safety concerns. Therefore, in this work, MSC membranes were ingeniously harnessed as protective shells to encapsulate synthesized PLGA nanoparticle cores (PLGA/MSCs). This strategic approach enabled nanoparticles to simulate endogenous substances and yielded a core-shell nano-biomimetic structure. The biomimetic nanocarrier remarkably maintained the homing ability of MSCs to inflammatory sites. In this study, cryptotanshinone (CPT)-loaded PLGA/MSCs (CPT@PLGA/MSC) were prepared. These nanoparticles can be effectively internalized by LO2 cells. They reduced cellular oxidative stress and elevated inflammatory levels. In vivo results suggested that, after intravenous administration, CPT@PLGA/MSCs significantly reduced uptake by the reticuloendothelial system and immune recognition compared to PLGA nanoparticles without MSC membrane coatings, subsequently resulting in their targeted and enhanced accumulation in the liver. The effectiveness of CPT@PLGA/MSCs in alleviating carbon tetrachloride-induced oxidative stress and inflammation in a mouse model was unequivocally demonstrated through comprehensive histological examination and liver function tests. This study introduces a pioneering strategy with substantial potential for ALI treatment.
Collapse
Affiliation(s)
- Xin Zhang
- College of Science, Sichuan Agricultural University, Ya’an 625014, China; (X.Z.); (Y.Y.); (Y.J.); (X.D.)
| | - Yao Yi
- College of Science, Sichuan Agricultural University, Ya’an 625014, China; (X.Z.); (Y.Y.); (Y.J.); (X.D.)
| | - Yuanyuan Jiang
- College of Science, Sichuan Agricultural University, Ya’an 625014, China; (X.Z.); (Y.Y.); (Y.J.); (X.D.)
| | - Jinqiu Liao
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.L.); (R.Y.)
| | - Ruiwu Yang
- College of Life Science, Sichuan Agricultural University, Ya’an 625014, China; (J.L.); (R.Y.)
| | - Xuexue Deng
- College of Science, Sichuan Agricultural University, Ya’an 625014, China; (X.Z.); (Y.Y.); (Y.J.); (X.D.)
| | - Li Zhang
- College of Science, Sichuan Agricultural University, Ya’an 625014, China; (X.Z.); (Y.Y.); (Y.J.); (X.D.)
| |
Collapse
|
17
|
Inoue A, Ohnishi T, Nishikawa M, Ohtsuka Y, Kusakabe K, Yano H, Tanaka J, Kunieda T. A Narrative Review on CD44's Role in Glioblastoma Invasion, Proliferation, and Tumor Recurrence. Cancers (Basel) 2023; 15:4898. [PMID: 37835592 PMCID: PMC10572085 DOI: 10.3390/cancers15194898] [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: 09/11/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023] Open
Abstract
High invasiveness is a characteristic of glioblastoma (GBM), making radical resection almost impossible, and thus, resulting in a tumor with inevitable recurrence. GBM recurrence may be caused by glioma stem-like cells (GSCs) that survive many kinds of therapy. GSCs with high expression levels of CD44 are highly invasive and resistant to radio-chemotherapy. CD44 is a multifunctional molecule that promotes the invasion and proliferation of tumor cells via various signaling pathways. Among these, paired pathways reciprocally activate invasion and proliferation under different hypoxic conditions. Severe hypoxia (0.5-2.5% O2) upregulates hypoxia-inducible factor (HIF)-1α, which then activates target genes, including CD44, TGF-β, and cMET, all of which are related to tumor migration and invasion. In contrast, moderate hypoxia (2.5-5% O2) upregulates HIF-2α, which activates target genes, such as vascular endothelial growth factor (VEGF)/VEGFR2, cMYC, and cyclin D1. All these genes are related to tumor proliferation. Oxygen environments around GBM can change before and after tumor resection. Before resection, the oxygen concentration at the tumor periphery is severely hypoxic. In the reparative stage after resection, the resection cavity shows moderate hypoxia. These observations suggest that upregulated CD44 under severe hypoxia may promote the migration and invasion of tumor cells. Conversely, when tumor resection leads to moderate hypoxia, upregulated HIF-2α activates HIF-2α target genes. The phenotypic transition regulated by CD44, leading to a dichotomy between invasion and proliferation according to hypoxic conditions, may play a crucial role in GBM recurrence.
Collapse
Affiliation(s)
- Akihiro Inoue
- Department of Neurosurgery, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (M.N.); (Y.O.); (K.K.); (T.K.)
| | - Takanori Ohnishi
- Department of Neurosurgery, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (M.N.); (Y.O.); (K.K.); (T.K.)
- Department of Neurosurgery, Advanced Brain Disease Center, Washoukai Sadamoto Hospital, 1-6-1 Takehara, Matsuyama 790-0052, Ehime, Japan
| | - Masahiro Nishikawa
- Department of Neurosurgery, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (M.N.); (Y.O.); (K.K.); (T.K.)
| | - Yoshihiro Ohtsuka
- Department of Neurosurgery, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (M.N.); (Y.O.); (K.K.); (T.K.)
| | - Kosuke Kusakabe
- Department of Neurosurgery, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (M.N.); (Y.O.); (K.K.); (T.K.)
| | - Hajime Yano
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicene, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (H.Y.); (J.T.)
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicene, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (H.Y.); (J.T.)
| | - Takeharu Kunieda
- Department of Neurosurgery, Ehime University Graduate School of Medicine, 454 Shitsukawa, Toon 791-0295, Ehime, Japan; (M.N.); (Y.O.); (K.K.); (T.K.)
| |
Collapse
|
18
|
Qadri MM. Targeting CD44 Receptor Pathways in Degenerative Joint Diseases: Involvement of Proteoglycan-4 (PRG4). Pharmaceuticals (Basel) 2023; 16:1425. [PMID: 37895896 PMCID: PMC10609794 DOI: 10.3390/ph16101425] [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: 09/07/2023] [Revised: 10/02/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Rheumatoid arthritis (RA), osteoarthritis (OA), and gout are the most prevalent degenerative joint diseases (DJDs). The pathogenesis underlying joint disease in DJDs remains unclear. Considering the severe toxicities reported with anti-inflammatory and disease-modifying agents, there is a clear need to develop new treatments that are specific in their effect while not being associated with significant toxicities. A key feature in the development of joint disease is the overexpression of adhesion molecules, e.g., CD44. Expression of CD44 and its variants in the synovial tissues of patients with DJDs is strongly associated with cartilage damage and appears to be a predicting factor of synovial inflammation in DJDs. Targeting CD44 and its downstream signaling proteins has emerged as a promising therapeutic strategy. PRG4 is a mucinous glycoprotein that binds to the CD44 receptor and is physiologically involved in joint lubrication. PRG4-CD44 is a pivotal regulator of synovial lining cell hemostasis in the joint, where lack of PRG4 expression triggers chronic inflammation and fibrosis, driven by persistent activation of synovial cells. In view of the significance of CD44 in DJD pathogenesis and the potential biological role for PRG4, this review aims to summarize the involvement of PRG4-CD44 signaling in controlling synovitis, synovial hypertrophy, and tissue fibrosis in DJDs.
Collapse
Affiliation(s)
- Marwa M. Qadri
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia;
- Inflammation Pharmacology and Drug Discovery Unit, Medical Research Center (MRC), Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
19
|
Deng W, Ai J, Zhang W, Zhou Z, Li M, Yan L, Zhang L, Huang Z, Wu Z, Ai J, Jiang H. Arginine methylation of HSPA8 by PRMT9 inhibits ferroptosis to accelerate hepatitis B virus-associated hepatocellular carcinoma progression. J Transl Med 2023; 21:625. [PMID: 37715221 PMCID: PMC10503172 DOI: 10.1186/s12967-023-04408-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/31/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND The hepatitis B virus X (HBx) protein is an established cause of hepatitis B virus (HBV)-induced hepatocellular carcinoma (HCC). Whether arginine methylation regulates ferroptosis involved in HBx-induced HCC progression has not been reported. This study aimed to explore whether HBx-regulated protein arginine methyltransferase 9 (PRMT9) mediates the involvement of ferroptosis in the development of HCC. METHODS AND RESULTS HBx inhibited ferroptosis through promoting PRMT9 expression in HCC cells. PRMT9 suppressed ferroptosis to accelerate HCC progression in vivo. PRMT9 targeted HSPA8 and enhanced arginine methylation of HSPA8 at R76 and R100 to regulate ferroptosis in HCC. HSPA8 overexpression altered the transcriptome profile of HepG2 cells, in particular, ferroptosis and immune-related pathways were significantly enriched by differentially expressed genes, including CD44. HSPA8 overexpression up-regulated CD44 expression and knockdown of CD44 significantly reversed the inhibition of ferroptosis caused by PRMT9 overexpression. CONCLUSIONS In conclusion, HBx/PRMT9/HSPA8/CD44 axis is a vital signal pathway regulating ferroptosis in HCC cells. This study provides new opportunities and targets for the treatment of HBV-induced HCC.
Collapse
Affiliation(s)
- Wensheng Deng
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China
| | - Jiaoyu Ai
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Wanlin Zhang
- Department of Clinical Laboratory, Ningbo Yinzhou No. 2 Hospital Ningbo Urology and Nephtology Hospital, Ningbo, 315100, Zhejiang, China
| | - Zhenyu Zhou
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Muqi Li
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China
| | - Likun Yan
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China
| | - Lidong Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China
| | - Zongjing Huang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China
| | - Ziyi Wu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China
| | - Junhua Ai
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China.
| | - Hai Jiang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang City, 330000, Jiangxi, China.
| |
Collapse
|
20
|
Liu S, Kong X, Fang Y, He Z, Wu H, Ji J, Yang X, Ye L, Zhai G. A dual-sensitive nanoparticle-mediated deepening synergistic therapy strategy involving DNA damage and ICD stimuli to treat triple-negative breast cancer. Biomater Sci 2023; 11:6325-6341. [PMID: 37555273 DOI: 10.1039/d3bm00781b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
Triple-negative breast cancer (TNBC) is one of the most aggressive cancers with an immunosuppressive microenvironment, and achieving a satisfactory effect from monotherapies, such as chemotherapy, photodynamic therapy (PDT) or immunotherapy, remains difficult. To solve this puzzle, a deepening synergistic therapy strategy of DNA damage and immunogenic cell death (ICD) stimuli was proposed. We engineered a doxorubicin (DOX) and 4-(hydroxymethyl) phenylboronic acid pinacol ester (PBAP) prodrug polymer, and encapsulated chlorin e6 (Ce6) to obtain the hyaluronidase (HAase) and H2O2 dual-sensitive responsive nanoparticles (Ce6/HDP NPs). The NPs displayed efficient intratumoral accumulation and cellular internalization properties due to the active targeting of the hyaluronic acid (HA). The dual DNA damage of the chemotherapy and ROS production directly caused tumor cell apoptosis. The strong ICD stimuli, which were induced by ROS production and GSH depletion, generated an amplified immunogenicity to activate tumor immunotherapy in vivo. In this manner, the NPs could significantly inhibit primary tumor, abscopal tumor, pulmonary metastasis and recurrent tumor in a subcutaneous 4T1 tumor model, with effective biosafety. This study has provided a promising deepening synergistic therapy strategy against TNBC.
Collapse
Affiliation(s)
- Shangui Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
| | - Xinru Kong
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
| | - Yuelin Fang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
| | - Zhijing He
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
| | - Hang Wu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, P.R. China.
| |
Collapse
|
21
|
Bhattacharjee R, Prabhakar N, Kumar L, Bhattacharjee A, Kar S, Malik S, Kumar D, Ruokolainen J, Negi A, Jha NK, Kesari KK. Crosstalk between long noncoding RNA and microRNA in Cancer. Cell Oncol (Dordr) 2023; 46:885-908. [PMID: 37245177 PMCID: PMC10356678 DOI: 10.1007/s13402-023-00806-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2023] [Indexed: 05/29/2023] Open
Abstract
miRNAs and lncRNAs play a central role in cancer-associated gene regulations. The dysregulated expression of lncRNAs has been reported as a hallmark of cancer progression, acting as an independent prediction marker for an individual cancer patient. The interplay of miRNA and lncRNA decides the variation of tumorigenesis that could be mediated by acting as sponges for endogenous RNAs, regulating miRNA decay, mediating intra-chromosomal interactions, and modulating epigenetic components. This paper focuses on the influence of crosstalk between lncRNA and miRNA on cancer hallmarks such as epithelial-mesenchymal transition, hijacking cell death, metastasis, and invasion. Other cellular roles of crosstalks, such as neovascularization, vascular mimicry, and angiogenesis were also discussed. Additionally, we reviewed crosstalk mechanism with specific host immune responses and targeting interplay (between lncRNA and miRNA) in cancer diagnosis and management.
Collapse
Affiliation(s)
- Rahul Bhattacharjee
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Neeraj Prabhakar
- Centre for Structural System Biology, Department of Physics, University of Hamburg, c/o DESY, Building 15, Notkestr. 852267, Hamburg, Germany
- Pharmacy, Abo Akademi University, Tykistökatu 6A, Turku, Finland
| | - Lamha Kumar
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, India
| | - Arkadyuti Bhattacharjee
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Sulagna Kar
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, Jharkhand, 834001, India
| | - Dhruv Kumar
- School of Health Sciences and Technology (SoHST), UPES University, Dehradun, Uttarakhand, India
| | - Janne Ruokolainen
- Department of Applied Physics, School of Science, Aalto University, Espoo, 00076, Finland
| | - Arvind Negi
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo, 00076, Finland.
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, 201310, UP, India.
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, 144411, India.
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, 248007, India.
| | - Kavindra Kumar Kesari
- Department of Applied Physics, School of Science, Aalto University, Espoo, 00076, Finland.
- Faculty of Biological and Environmental Sciences, University of Helsinki, Biocentre 3, Helsinki, Finland.
| |
Collapse
|
22
|
Poothakulath Krishnan R, Pandiar D, Ramani P, Ramalingam K, Jayaraman S. Utility of CD44/CD24 in the Outcome and Prognosis of Oral Squamous Cell Carcinoma: A Systematic Review. Cureus 2023; 15:e42899. [PMID: 37664387 PMCID: PMC10474850 DOI: 10.7759/cureus.42899] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/03/2023] [Indexed: 09/05/2023] Open
Abstract
Cancer stem cells (CSCs) are characterized by their capacity for self-renewal and differentiation. CD44 and CD24 are two commonly used markers to identify these CSCs. Despite the enormous amount of data available in the literature, their specificity and coexistence remain elusive in oral squamous cell carcinoma (OSCC). In the present review, we aimed to assess the diagnostic utility of the CD44/CD24 combination in tumor development and metastasis in OSCC. Two investigators independently performed a systematic search to identify all the relevant studies from various electronic databases. Out of 694 articles, 9 were found eligible for further evaluation. Details including the number of patients, gender, site, tobacco and alcohol consumption, histological stage, CD24 expression, CD44 expression, CD44/CD24 expression, nodal status, disease-free survival, and overall survival were extracted. CD44+CD24- expression was noted in 35/207 (16.9%) cases, CD44+CD24+ in 53/207 (25.6%) cases, CD44-CD24- in 49/207 subjects (23.67%), and CD44-/CD24+ in 70/207 (33.81%) cases. CD44 or CD24 or their co-expression did not correlate with the disease-free survival rate, and double negatives (CD44-/CD24-) demonstrated a higher overall survival than other immunotypes. CD44/CD24 profile may be used on small incisional biopsies to predict the outcome and treatment planning. This finding may help in developing new therapeutic targets to suppress cancer metastasis and provide a better long-term prognosis for patients diagnosed with OSCC.
Collapse
Affiliation(s)
- Reshma Poothakulath Krishnan
- Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Deepak Pandiar
- Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Pratibha Ramani
- Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Karthikeyan Ramalingam
- Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD) Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
23
|
Prajapati KS, Kumar S. Loss of miR-6844 alters stemness/self-renewal and cancer hallmark(s) markers through CD44-JAK2-STAT3 signaling axis in breast cancer stem-like cells. J Cell Biochem 2023; 124:1186-1202. [PMID: 37436061 DOI: 10.1002/jcb.30441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/27/2023] [Accepted: 07/02/2023] [Indexed: 07/13/2023]
Abstract
MicroRNAs regulate breast stemness and self-renewal properties in breast cancer cells at the molecular level. Recently we reported the clinical relevance and in vitro expression profile of novel miR-6844 in breast cancer and -derived stem-like cells (mammosphere). In the present study, we first time explore the functional role of loss of miR-6844 in breast cancer cells derived mammosphere. Down expression of miR-6844 significantly decreased cell proliferation in MCF-7 and T47D cells derived mammosphere in a time-dependent manner. MiR-6844 down expression reduced the sphere formation in terms of size and number in test cells. Loss of miR-6844 significantly altered stemness and self-renewal markers (Bmi-1, Nanog, c-Myc, Sox2, and CD44) in mammosphere compared to negative control spheres. Moreover, loss of miR-6844 inhibits the JAK2-STAT3 signaling pathway by decreasing p-JAK2 and p-STAT3 levels in breast cancer cells derived mammosphere. Loss of miR-6844 expression significantly decreased CCND1 and CDK4 mRNA/protein levels and arrested breast cancer stem-like cells in G2/M phase. Reduced expression of miR-6844 increased Bax/Bcl-2 ratio, late apoptotic cell population, and Caspase 9 and 3/7 activity in the mammosphere. Low expression of miR-6844 decreased migratory and invasive cells by altering the expression of Snail, E-cad, and Vimentin at mRNA/protein levels. In conclusion, loss of miR-6844 decreases stemness/self-renewal and other cancer hallmark in breast cancer stem-like cells through CD44-JAK2-STAT3 axis. Thus, downregulation of miR-6844 by therapeutic agents might be a novel strategy to target breast cancer stemness and self-renewal.
Collapse
Affiliation(s)
- Kumari Sunita Prajapati
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, Punjab, India
| | - Shashank Kumar
- Molecular Signaling & Drug Discovery Laboratory, Department of Biochemistry, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
24
|
Bashkeran T, Kamaruddin AH, Ngo TX, Suda K, Umakoshi H, Watanabe N, Nadzir MM. Niosomes in cancer treatment: A focus on curcumin encapsulation. Heliyon 2023; 9:e18710. [PMID: 37593605 PMCID: PMC10428065 DOI: 10.1016/j.heliyon.2023.e18710] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/19/2023] Open
Abstract
Curcumin is widely used as a therapeutic drug for cancer treatment. However, its limited absorption and rapid excretion are the major therapeutic limitations to its clinical use. Using niosomes as a curcumin delivery system is a cheap, easy, and less toxic strategy for enhancing the absorption of curcumin by cells and delaying its excretion. Thus, there is a vital need to explore curcumin niosomes to configure the curcumin to suitably serve and aid current pharmacokinetics in treatments for cancer. To date, no comprehensive review has focused on the cytotoxic effects of curcumin niosomes on malignant cells. Thus, this review provides a critical analysis of the curcumin niosomes in cancer treatment, formulations of curcumin niosomes, characterizations of curcumin niosomes, and factors influencing their performance. The findings from this review article can strongly accelerate the understanding of curcumin niosomes and pave a brighter direction towards advances in the pharmaceutical, biotechnology, and medical industries.
Collapse
Affiliation(s)
- Thaaranni Bashkeran
- School of Chemical Engineering, Universiti Sains Malaysia, Engineering Campus, 14300, Nibong Tebal, Pulau Pinang, Malaysia
| | - Azlina Harun Kamaruddin
- School of Chemical Engineering, Universiti Sains Malaysia, Engineering Campus, 14300, Nibong Tebal, Pulau Pinang, Malaysia
| | - Trung Xuan Ngo
- Rohto Pharmaceutical Co., Ltd., Basic Research Division, Research Village Kyoto, 6-5-4 Kunimidai, Kizugawa, Kyoto, 619-0216, Japan
| | - Kazuma Suda
- Rohto Pharmaceutical Co., Ltd., Basic Research Division, Research Village Kyoto, 6-5-4 Kunimidai, Kizugawa, Kyoto, 619-0216, Japan
| | - Hiroshi Umakoshi
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka, 560-8531, Japan
| | - Nozomi Watanabe
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama-cho, Toyonaka, 560-8531, Japan
| | - Masrina Mohd Nadzir
- School of Chemical Engineering, Universiti Sains Malaysia, Engineering Campus, 14300, Nibong Tebal, Pulau Pinang, Malaysia
| |
Collapse
|
25
|
Timofeeva AM, Paramonik AP, Sedykh SS, Nevinsky GA. Milk Exosomes: Next-Generation Agents for Delivery of Anticancer Drugs and Therapeutic Nucleic Acids. Int J Mol Sci 2023; 24:10194. [PMID: 37373342 DOI: 10.3390/ijms241210194] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Exosomes are nanovesicles 40-120 nm in diameter secreted by almost all cell types and providing humoral intercellular interactions. Given the natural origin and high biocompatibility, the potential for loading various anticancer molecules and therapeutic nucleic acids inside, and the surface modification possibility for targeted delivery, exosomes are considered to be a promising means of delivery to cell cultures and experimental animal organisms. Milk is a unique natural source of exosomes available in semi-preparative and preparative quantities. Milk exosomes are highly resistant to the harsh conditions of the gastrointestinal tract. In vitro studies have demonstrated that milk exosomes have an affinity to epithelial cells, are digested by cells by endocytosis mechanism, and can be used for oral delivery. With milk exosome membranes containing hydrophilic and hydrophobic components, exosomes can be loaded with hydrophilic and lipophilic drugs. This review covers a number of scalable protocols for isolating and purifying exosomes from human, cow, and horse milk. Additionally, it considers passive and active methods for drug loading into exosomes, as well as methods for modifying and functionalizing the surface of milk exosomes with specific molecules for more efficient and specific delivery to target cells. In addition, the review considers various approaches to visualize exosomes and determine cellular localization and bio-distribution of loaded drug molecules in tissues. In conclusion, we outline new challenges for studying milk exosomes, a new generation of targeted delivery agents.
Collapse
Affiliation(s)
- Anna M Timofeeva
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Anastasia P Paramonik
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Sergey S Sedykh
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Georgy A Nevinsky
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
26
|
Ahmad SMS, Nazar H, Rahman MM, Rusyniak RS, Ouhtit A. ITGB1BP1, a Novel Transcriptional Target of CD44-Downstream Signaling Promoting Cancer Cell Invasion. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:373-380. [PMID: 37252376 PMCID: PMC10225144 DOI: 10.2147/bctt.s404565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023]
Abstract
Breast cancer (BC) is the most common malignancy worldwide and has a poor prognosis, because it begins in the breast and disseminates to lymph nodes and distant organs. While invading, BC cells acquire aggressive characteristics from the tumor microenvironment through several mechanisms. Thus, understanding the mechanisms underlying the process of BC cell invasion can pave the way towards the development of targeted therapeutics focused on metastasis. We have previously reported that the activation of CD44 receptor with its major ligand hyaluronan (HA) promotes BC metastasis to the liver in vivo. Next, a gene expression profiling microarray analysis was conducted to identify and validate CD44-downstream transcriptional targets mediating its pro-metastatic function from RNA samples collected from Tet CD44-induced versus control MCF7-B5 cells. We have already validated a number of novel CD44-target genes and published their underlying signaling pathways in promoting BC cell invasion. From the same microarray analysis, Integrin subunit beta 1 binding protein 1 (ITGB1BP1) was also identified as a potential CD44-target gene that was upregulated (2-fold) upon HA activation of CD44. This report will review the lines of evidence collected from the literature to support our hypothesis, and further discuss the possible mechanisms linking HA activation of CD44 to its novel potential transcriptional target ITGB1BP1.
Collapse
Affiliation(s)
- Salma M S Ahmad
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Hanan Nazar
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Md Mizanur Rahman
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Radoslaw Stefan Rusyniak
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Allal Ouhtit
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| |
Collapse
|
27
|
Ding YH, Wang XL, Li SR, Li QX, Xu TA, Zhang YT. The crosslink between the hyaluronic acid and drugs treated by reactive oxygen species produced in plasma based on the molecular dynamics simulation. Int J Biol Macromol 2023; 242:124944. [PMID: 37210061 DOI: 10.1016/j.ijbiomac.2023.124944] [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: 03/23/2023] [Revised: 04/29/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Hyaluronic Acid (HA)-based pre-drugs can enable targeted drug delivery to cancer cells with CD44-high expressing, thus, it is essential to design an efficient, target specific drug delivery system based on HA. Plasma, as a simple and clean tool, has been widely used in the modification and crosslinking of biological materials in recent years. In this paper, we used the Reactive Molecular Dynamic (RMD) to explore the reaction between reactive oxygen species (ROS) in plasma and HA with drugs (PTX, SN-38, and DOX), in order to examine possible drug-coupled systems. The simulation results indicated the acetylamino groups in HA could be oxidized to unsaturated acyl groups, which offers the possibility of crosslinking. Three drugs also exposed the unsaturated atoms under the impact of ROS, which can cross-link directly to HA through CO and CN bonds, forming a drug coupling system with better release. This study revealed the exposure of active sites on HA and drugs by ROS impact in plasma, allowing us to study the crosslinking mechanism between HA and drugs at molecular level deeply, and also provided a new light for establishment of HA-based targeted drug delivery system.
Collapse
Affiliation(s)
- Yun-Han Ding
- School of Electrical Engineeringe, Shandong University, Jinan, Shandong Province 250061, China
| | - Xiao-Long Wang
- School of Electrical Engineeringe, Shandong University, Jinan, Shandong Province 250061, China.
| | - Shan-Rui Li
- School of Electrical Engineeringe, Shandong University, Jinan, Shandong Province 250061, China
| | - Quan-Xin Li
- The Second Hospital of Shandong University, Shandong University, Jinan, Shandong 250033, China
| | - Tian-Ao Xu
- School of Electrical Engineeringe, Shandong University, Jinan, Shandong Province 250061, China
| | - Yuan-Tao Zhang
- School of Electrical Engineeringe, Shandong University, Jinan, Shandong Province 250061, China
| |
Collapse
|
28
|
Conte M, De Feo MS, Sidrak MMA, Corica F, Gorica J, Filippi L, Schillaci O, De Vincentis G, Frantellizzi V. Radiolabeled Dendrimer Coated Nanoparticles for Radionuclide Imaging and Therapy: A Systematic Review. Pharmaceutics 2023; 15:pharmaceutics15030867. [PMID: 36986728 PMCID: PMC10051715 DOI: 10.3390/pharmaceutics15030867] [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: 01/30/2023] [Revised: 02/23/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Dendrimers are nanoscale-size polymers with a globular structure. They are composed of an internal core and branching dendrons with surface active groups which can be functionalized for medical applications. Different complexes have been developed for imaging and therapeutic purposes. This systematic review aims to summarize the development of newer dendrimers for oncological applications in nuclear medicine. METHODS An online literature search was conducted on Pubmed, Scopus, Medline, Cochrane Library, and Web Of Science databases selecting published studies from January 1999 to December 2022. The accepted studies considered the synthesis of dendrimer complexes for oncological nuclear medicine imaging and therapy. RESULTS 111 articles were identified; 69 articles were excluded because they did not satisfy the selection criteria. Thus, nine duplicate records were removed. The remaining 33 articles were included and selected for quality assessment. CONCLUSION Nanomedicine has led researchers to create novel nanocarriers with high affinity for the target. Dendrimers represent feasible imaging probes and therapeutic agents since, through the functionalization of external chemical groups and thanks to the possibility to carry pharmaceuticals, it can be possible to exploit different therapeutic strategies and develop a useful weapon for oncological treatments.
Collapse
Affiliation(s)
- Miriam Conte
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00161 Rome, Italy
| | - Maria Silvia De Feo
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00161 Rome, Italy
| | - Marko Magdi Abdou Sidrak
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00161 Rome, Italy
| | - Ferdinando Corica
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00161 Rome, Italy
| | - Joana Gorica
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00161 Rome, Italy
| | - Luca Filippi
- Department of Nuclear Medicine, Santa Maria Goretti Hospital, 04100 Latina, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University Tor Vergata, 00133 Rome, Italy
| | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00161 Rome, Italy
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomo-Pathology, Sapienza, University of Rome, 00161 Rome, Italy
| |
Collapse
|
29
|
Gheybi E, Asoodeh A, Amani J. In silico designing and expression of novel recombinant construct containing the variable part of CD44 extracellular domain for prediagnostic breast cancer. Cancer Rep (Hoboken) 2023; 6:e1745. [PMID: 36289579 PMCID: PMC10026285 DOI: 10.1002/cnr2.1745] [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: 07/15/2022] [Revised: 09/22/2022] [Accepted: 10/13/2022] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND CD44, as a tumor-associated marker, can be used to detect stem cells in breast cancer. While CD44 is expressed in normal epithelial cells, carcinoma cells overexpress CD44. AIMS In the current study, we designed a recombinant protein that included the variable component of the CD44 (CD44v) extracellular domain to apply in clinical diagnosis of breast cancer. METHODS A total of 100 CD44v amino-acid residues were determined, and the structure was examined using bioinformatics tools. The construct was inserted into the PET28a vector and transformed in E. coli BL21(DE3). A nearly 12 kDa fusion protein was obtained by Ni-NTA affinity metal chromatography. Recombinant CD44v was examined by Western blotting, ELISA, and immunohistochemistry (IHC) assays. RESULTS The findings revealed that the structure of rCD44v was stable, and its antigenic domain was exposed. The recombinant CD44v was confirmed by western blotting, and the presence of antibodies against recombinant CD44v protein in the patient's serum was detected by the ELISA. Our data demonstrated a link between CD44v serum levels and the prevalence of breast cancer. CONCLUSION Assessments of antiCD44v antibodies with rCD44v could be a useful tool for identifying breast cancer in its early stages, which can lead to better outcomes.
Collapse
Affiliation(s)
- Elaheh Gheybi
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Asoodeh
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Chakraborty A, Perez M, Carroll JD, Antonopoulos A, Dell A, Ortega L, Mohammed NBB, Wells M, Staudinger C, Griswold A, Chandler KB, Marrero C, Jimenez R, Tani Y, Wilmott JS, Thompson JF, Wang W, Sackstein R, Scolyer RA, Murphy GF, Haslam SM, Dimitroff CJ. Hypoxia Controls the Glycome Signature and Galectin-8-Ligand Axis to Promote Protumorigenic Properties of Metastatic Melanoma. J Invest Dermatol 2023; 143:456-469.e8. [PMID: 36174713 PMCID: PMC10123958 DOI: 10.1016/j.jid.2022.07.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 06/29/2022] [Accepted: 07/14/2022] [Indexed: 01/14/2023]
Abstract
The prognosis for patients with metastatic melanoma (MM) involving distant organs is grim, and treatment resistance is potentiated by tumor-initiating cells (TICs) that thrive under hypoxia. MM cells, including TICs, express a unique glycome featuring i-linear poly-N-acetyllactosamines through the loss of I-branching enzyme, β1,6 N-acetylglucosaminyltransferase 2. Whether hypoxia instructs MM TIC development by modulating the glycome signature remains unknown. In this study, we explored hypoxia-dependent alterations in MM glycome‒associated genes and found that β1,6 N-acetylglucosaminyltransferase 2 was downregulated and a galectin (Gal)-8-ligand axis, involving both extracellular and cell-intrinsic Gal-8, was induced. Low β1,6 N-acetylglucosaminyltransferase 2 levels correlated with poor patient outcomes, and patient serum samples were elevated for Gal-8. Depressed β1,6 N-acetylglucosaminyltransferase 2 in MM cells upregulated TIC marker, NGFR/CD271, whereas loss of MM cell‒intrinsic Gal-8 markedly lowered NGFR and reduced TIC activity in vivo. Extracellular Gal-8 bound preferentially to i-linear poly-N-acetyllactosamines on N-glycans of the TIC marker and prometastatic molecule CD44, among other receptors, and activated prosurvival factor protein kinase B. This study reveals the importance of hypoxia governing the MM glycome by enforcing i-linear poly-N-acetyllactosamine and Gal-8 expression. This mechanistic investigation also uncovers glycome-dependent regulation of pro-MM factor, NGFR, implicating i-linear poly-N-acetyllactosamine and Gal-8 as biomarkers and therapeutic targets of MM.
Collapse
Affiliation(s)
- Asmi Chakraborty
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Mariana Perez
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Jordan D Carroll
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | | | - Anne Dell
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Liettel Ortega
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Norhan B B Mohammed
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA; Department of Medical Biochemistry, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Michael Wells
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Caleb Staudinger
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Anthony Griswold
- John P. Hussman Institute for Human Genomics (HIHG), Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Kevin B Chandler
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Cristina Marrero
- Miami Cancer Institute, Baptist Health-South Florida, Miami, Florida, USA
| | - Ramon Jimenez
- Miami Cancer Institute, Baptist Health-South Florida, Miami, Florida, USA
| | - Yoshihiko Tani
- Japanese Red Cross Kinki Block Blood Center, Osaka, Japan
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Department of Melanoma and Surgical Oncology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Wei Wang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachussetts, USA
| | - Robert Sackstein
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, Australia; Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - George F Murphy
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stuart M Haslam
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Charles J Dimitroff
- Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA.
| |
Collapse
|
31
|
Gheybi E, Asoodeh A, Amani J. Preparation of chitosan nanoparticle containing recombinant CD44v antigen and evaluation of its immunization capacity against breast cancer in BALB/c mice. BMC Cancer 2023; 23:134. [PMID: 36759786 PMCID: PMC9912563 DOI: 10.1186/s12885-023-10614-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
OBJECTIVE(S) Breast tumors show heterogeneity containing cancer stem cells as a small subpopulation of a tumor mass. CD44 as a cancer stem cells antigen is abnormally expressed by carcinomas of epithelial origin. Also, overexpression of CD44 variable isoforms (CD44v) is associated with malignancy in breast cancer. In the present research, our objective was to evaluate the immunogenicity of prepared nanoparticles containing a novel recombinant CD44v (rCD44v) protein in the mouse model. MATERIALS AND METHODS CD44 gene was expressed in E. coli BL21 DE3 using the pET28a-CD44 vector. The expressed rCD44v protein was purified, encapsulated into the chitosan nanoparticles, and administered to BALB/c mice. ELISA was used to evaluate the immunoglobulin levels of immunized animals. For challenge experiment, 2 × 106 4T1-CD44 tumor cells were injected subcutaneously in mice, and tumor size, necrosis, and metastases were measured. Finally, cell proliferation assay, cytokines assay, and neutralization assay of the mouse anti-rCD44v on the human breast cancer cell line were examined. RESULTS The measured size of chitosan-rCD44v nanoparticles was 146.5 nm. Recombinant CD44v encapsulated by chitosan nanoparticles increases immunological responses via the adjuvant nature of chitosan nanoparticles. In the immunized mice, IgG and IgA titers were significantly increased. Tumor growth in injection and nano-injection test groups compared with the mice control groups displayed a significant reduction (P < 0.05). A high amount of splenocytes secreting IFNγ and IL-17 was seen in immunized mice with rCD44v (P < 0.05). Furthermore, a smaller size of lung metastases compared to the control mice groups was detected. CONCLUSION The encapsulated rCD44v within the chitosan nanoparticles induced a significant immune response in mice and can establish significant protection against breast cancer. Therefore, it can be considered a vaccine candidate for breast cancer therapeutic modalities.
Collapse
Affiliation(s)
- Elaheh Gheybi
- grid.411301.60000 0001 0666 1211Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran ,grid.411301.60000 0001 0666 1211Cellular and Molecular Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Asoodeh
- Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran. .,Cellular and Molecular Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Jafar Amani
- grid.411521.20000 0000 9975 294XApplied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Yao H, Lyu F, Ma J, Sun F, Tang G, Wu J, Zhou Z. PIMREG is a prognostic biomarker involved in immune microenvironment of clear cell renal cell carcinoma and associated with the transition from G1 phase to S phase. Front Oncol 2023; 13:1035321. [PMID: 36776322 PMCID: PMC9909346 DOI: 10.3389/fonc.2023.1035321] [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: 09/02/2022] [Accepted: 01/12/2023] [Indexed: 01/28/2023] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is one of the most common tumors in the world and affects human health seriously. PIMREG is a mitotic regulator which is essential to the metaphase-to-anaphase transition in cell cycle. Although PIMREG plays a crucial role in the malignant progression of tumors, there are few reports on its role in ccRCC. Methods The transcriptional expression profile and clinical data of PIMREG were downloaded from TCGA database and verified by qRT-PCR. Kaplan-Meier plotter was used to analyze the effect of PIMREG on overall survival (OS), disease specific survival (DSS) and progression-free interval (PFI) of patients with ccRCC. Univariable and multivariable Cox regression analysis were used to determine the independent prognostic factors of ccRCC. The effects of PIMREG on cell migration and invasion were detected by wound healing assay and transwell invasion assay, and CCK-8 assay, colony formation assay and cell cycle assay were used to detect the effect of PIMREG on cell proliferation. In addition, the changes in cell cycle related proteins were detected by western blot. Results PIMREG was highly expressed in human ccRCC and was positively correlated with pathologic stage, TNM stage and histologic grade. In addition, patients with high expression of PIMREG had a poor prognosis. Univariable and multivariable Cox regression analysis identified that PIMREG was an independent prognostic factor of ccRCC. Additionally, PIMREG was also closely related to immune cell infiltration. Experiments in vitro identified that the knockdown of PIMREG could significantly inhibit the proliferation, migration and invasion abilities of ccRCC. The expression of cyclin D1, CDK4 and CDK6 was also significantly reduced after PIMREG knockdown. Conclusions PIMREG plays a vital role in the development of ccRCC and may become a potential therapeutic target in the future.
Collapse
Affiliation(s)
- Huibao Yao
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Feifei Lyu
- Department of Traditional Chinese Medicine, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Jian Ma
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Fengze Sun
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Gonglin Tang
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Jitao Wu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China,*Correspondence: Zhongbao Zhou, ; Jitao Wu,
| | - Zhongbao Zhou
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China,Department of Urology, Beijing TianTan Hospital, Capital Medical University, Beijing, China,*Correspondence: Zhongbao Zhou, ; Jitao Wu,
| |
Collapse
|
33
|
Brennan J, Lu ML, Kang Y. A New Model of Esophageal Cancers by Using a Detergent-Free Decellularized Matrix in a Perfusion Bioreactor. Bioengineering (Basel) 2023; 10:96. [PMID: 36671668 PMCID: PMC9854977 DOI: 10.3390/bioengineering10010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/06/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
The lack of physiologically relevant human esophageal cancer models has as a result that many esophageal cancer studies are encountering major bottleneck challenges in achieving breakthrough progress. To address the issue, here we engineered a 3D esophageal tumor tissue model using a biomimetic decellularized esophageal matrix in a customized bioreactor. To obtain a biomimetic esophageal matrix, we developed a detergent-free, rapid decellularization method to decellularize porcine esophagus. We characterized the decellularized esophageal matrix (DEM) and utilized the DEM for the growth of esophageal cancer cell KYSE30 in well plates and the bioreactor. We then analyzed the expression of cancer-related markers of KYSE30 cells and compared them with formalin-fixed, paraffin-embedded (FFPE) esophageal squamous cell carcinoma (ESCC) tissue biospecimens. Our results show that the detergent-free decellularization method preserved the esophageal matrix components and effectively removed cell nucleus. KYSE30 cancer cells proliferated well on and inside the DEM. KYSE30 cells cultured on the DEM in the dynamic bioreactor show different cancer marker expressions than those in the static well plate, and also share some similarities to the FFPE-ESCC biospecimens. These findings built a foundation with potential for further study of esophageal cancer behavior in a biomimetic microenvironment using this new esophageal cancer model.
Collapse
Affiliation(s)
- Jordan Brennan
- Department of Ocean and Mechanical Engineering, College of Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Michael L. Lu
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
- Faculty of Integrative Biology PhD Program, Department of Biological Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Yunqing Kang
- Department of Ocean and Mechanical Engineering, College of Engineering and Computer Science, Florida Atlantic University, Boca Raton, FL 33431, USA
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
- Faculty of Integrative Biology PhD Program, Department of Biological Science, Florida Atlantic University, Boca Raton, FL 33431, USA
| |
Collapse
|
34
|
Wöhner B, Li W, Hey S, Drobny A, Werny L, Becker-Pauly C, Lucius R, Zunke F, Linder S, Arnold P. Proteolysis of CD44 at the cell surface controls a downstream protease network. Front Mol Biosci 2023; 10:1026810. [PMID: 36876041 PMCID: PMC9981664 DOI: 10.3389/fmolb.2023.1026810] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 01/31/2023] [Indexed: 02/19/2023] Open
Abstract
The cell surface receptor cluster of differentiation 44 (CD44) is the main hyaluronan receptor of the human body. At the cell surface, it can be proteolytically processed by different proteases and was shown to interact with different matrix metalloproteinases. Upon proteolytic processing of CD44 and generation of a C-terminal fragment (CTF), an intracellular domain (ICD) is released after intramembranous cleavage by the γ-secretase complex. This intracellular domain then translocates to the nucleus and induces transcriptional activation of target genes. In the past CD44 was identified as a risk gene for different tumor entities and a switch in CD44 isoform expression towards isoform CD44s associates with epithelial to mesenchymal transition (EMT) and cancer cell invasion. Here, we introduce meprin β as a new sheddase of CD44 and use a CRISPR/Cas9 approach to deplete CD44 and its sheddases ADAM10 and MMP14 in HeLa cells. We here identify a regulatory loop at the transcriptional level between ADAM10, CD44, MMP14 and MMP2. We show that this interplay is not only present in our cell model, but also across different human tissues as deduced from GTEx (Gene Tissue Expression) data. Furthermore, we identify a close relation between CD44 and MMP14 that is also reflected in functional assays for cell proliferation, spheroid formation, migration and adhesion.
Collapse
Affiliation(s)
- Birte Wöhner
- Anatomical Institute, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Wenjia Li
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sven Hey
- Institute for Medical Microbiology, Virology, and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | - Alice Drobny
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ludwig Werny
- Biochemical Institute, Christian-Albrechts-University Kiel, Kiel, Germany
| | | | - Ralph Lucius
- Anatomical Institute, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Stefan Linder
- Institute for Medical Microbiology, Virology, and Hygiene, University Medical Center Eppendorf, Hamburg, Germany
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
35
|
Shi J, Yang Y, He W, Moses M, Gu YH, Li N, Di W. Correlation between CD44 and membrane fluidity-a study on biopsies of high-grade serous ovarian tumor. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1928-1930. [PMID: 36789687 PMCID: PMC10157600 DOI: 10.3724/abbs.2022190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Jun Shi
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yang Yang
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200032, China
| | - Wei He
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Munika Moses
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yi-Hua Gu
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200032, China
| | - Ningli Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wen Di
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
36
|
Michalczyk M, Humeniuk E, Adamczuk G, Korga-Plewko A. Hyaluronic Acid as a Modern Approach in Anticancer Therapy-Review. Int J Mol Sci 2022; 24:ijms24010103. [PMID: 36613567 PMCID: PMC9820514 DOI: 10.3390/ijms24010103] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/11/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Hyaluronic acid (HA) is a linear polysaccharide and crucial component of the extracellular matrix (ECM), maintaining tissue hydration and tension. Moreover, HA contributes to embryonic development, healing, inflammation, and cancerogenesis. This review summarizes new research on the metabolism and interactions of HA with its binding proteins, known as hyaladherins (CD44, RHAMM), revealing the molecular basis for its distinct biological function in the development of cancer. The presence of HA on the surface of tumor cells is a sign of an adverse prognosis. The involvement of HA in malignancy has been extensively investigated using cancer-free naked mole rats as a model. The HA metabolic components are examined for their potential impact on promoting or inhibiting tumor formation, proliferation, invasion, and metastatic spread. High molecular weight HA is associated with homeostasis and protective action due to its ability to preserve tissue integrity. In contrast, low molecular weight HA indicates a pathological condition in the tissue and plays a role in pro-oncogenic activity. A systematic approach might uncover processes related to cancer growth, establish novel prognostic indicators, and identify potential targets for treatment action.
Collapse
|
37
|
Raigorodskaya MP, Novosad VO, Tonevitskaya SA, Maltseva DV. Expression of CD44 Isoforms in Human Colorectal Cancer Cell Lines. APPL BIOCHEM MICRO+ 2022. [DOI: 10.1134/s0003683822090071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
38
|
Chen KW, Hsu PH, Huang HL, Liu HL, Lin YT, Hsu CY, Lin JH, Lin YH. Targeting nanoparticle-conjugated microbubbles combined with ultrasound-mediated microbubble destruction for enhanced tumor therapy. Pharmacol Res 2022; 186:106532. [DOI: 10.1016/j.phrs.2022.106532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/20/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022]
|
39
|
Ahmad SMS, Al-Mansoob M, Ouhtit A. SIRT1, a novel transcriptional downstream target of CD44, linking its deacetylase activity to tumor cell invasion/metastasis. Front Oncol 2022; 12:1038121. [PMID: 36505828 PMCID: PMC9727296 DOI: 10.3389/fonc.2022.1038121] [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: 09/06/2022] [Accepted: 10/11/2022] [Indexed: 11/24/2022] Open
Abstract
Our tetracycline-off-inducible CD44 expression system previously established in mouse model, revealed that activation of CD44 with its major ligand hyaluronan (HA) promoted breast cancer (BC) metastasis to the liver. To identify the mechanisms that underpin CD44-promoted BC cell invasion, microarray gene expression profiling using RNA samples from (Tet)-Off-regulated expression system of CD44s in MCF7 cells, revealed a set of upregulated genes including, nuclear sirtuin-1 (SIRT1 also known as NAD-dependent deacetylase), an enzyme that requires NAD+ as a cofactor to deacetylate several histones and transcription factors. It stimulates various oncogenic pathways promoting tumorigenesis. This data suggests that SIRT1 is a potential novel transcriptional target of CD44-downstream signaling that promote BC cell invasion/metastasis. This review will discuss the evidence supporting this hypothesis as well as the mechanisms linking SIRT1 to cell proliferation and invasion.
Collapse
|
40
|
Duan Y, Xu L, Song W, Gao H, Sun L, Chen F, Ma F. Label-free electrogenerated chemiluminescence biosensor for quantization of CD44 on basis of its heterodimerization with matrix metalloproteinase-14. Microchem J 2022. [DOI: 10.1016/j.microc.2022.107872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
41
|
Zhu K, Lang Z, Zhan Y, Tao Q, Yu Z, Chen L, Fan C, Jin Y, Yu K, Zhu B, Gao Y, Wang C, Jiang S, Shi Y. A novel 10-gene ferroptosis-related prognostic signature in acute myeloid leukemia. Front Oncol 2022; 12:1023040. [PMID: 36338716 PMCID: PMC9630338 DOI: 10.3389/fonc.2022.1023040] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is one of the most common hematopoietic malignancies and exhibits a high rate of relapse and unfavorable outcomes. Ferroptosis, a relatively recently described type of cell death, has been reported to be involved in cancer development. However, the prognostic value of ferroptosis-related genes (FRGs) in AML remains unclear. In this study, we found 54 differentially expressed ferroptosis-related genes (DEFRGs) between AML and normal marrow tissues. 18 of 54 DEFRGs were correlated with overall survival (OS) (P<0.05). Using the least absolute shrinkage and selection operator (LASSO) Cox regression analysis, we selected 10 DEFRGs that were associated with OS to build a prognostic signature. Data from AML patients from the International Cancer Genome Consortium (ICGC) cohort as well as the First Affiliated Hospital of Wenzhou Medical University (FAHWMU) cohort were used for validation. Notably, the prognostic survival analyses of this signature passed with a significant margin, and the riskscore was identified as an independent prognostic marker using Cox regression analyses. Then we used a machine learning method (SHAP) to judge the importance of each feature in this 10-gene signature. Riskscore was shown to have the highest correlation with this 10-gene signature compared with each gene in this signature. Further studies showed that AML was significantly associated with immune cell infiltration. In addition, drug-sensitive analysis showed that 8 drugs may be beneficial for treatment of AML. Finally, the expressions of 10 genes in this signature were verified by real-time quantitative polymerase chain reaction. In conclusion, our study establishes a novel 10-gene prognostic risk signature based on ferroptosis-related genes for AML patients and FRGs may be novel therapeutic targets for AML.
Collapse
Affiliation(s)
- Kai Zhu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhichao Lang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yating Zhan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiqi Tao
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhijie Yu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lili Chen
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Congcong Fan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Jin
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kang Yu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bihan Zhu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuxiang Gao
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chengchi Wang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Songfu Jiang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Clinical Research Center For Hematological disorders, Wenzhou, China
- *Correspondence: Yifen Shi, ; Songfu Jiang,
| | - Yifen Shi
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Zhejiang Provincial Clinical Research Center For Hematological disorders, Wenzhou, China
- *Correspondence: Yifen Shi, ; Songfu Jiang,
| |
Collapse
|
42
|
Babst N, Isbell LK, Rommel F, Tura A, Ranjbar M, Grisanti S, Tschuch C, Schueler J, Doostkam S, Reinacher PC, Duyster J, Kakkassery V, von Bubnoff N. CXCR4, CXCR5 and CD44 May Be Involved in Homing of Lymphoma Cells into the Eye in a Patient Derived Xenograft Homing Mouse Model for Primary Vitreoretinal Lymphoma. Int J Mol Sci 2022; 23:11757. [PMID: 36233057 PMCID: PMC9569795 DOI: 10.3390/ijms231911757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/25/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Primary vitreoretinal lymphoma (PVRL), a rare malignancy of the eye, is strongly related to primary central nervous system lymphoma (PCNSL). We hypothesized that lymphoma cells disseminate to the CNS and eye tissue via distinct homing receptors. The objective of this study was to test expression of CXCR4, CXCR5, CXCR7 and CD44 homing receptors on CD20 positive B-lymphoma cells on enucleated eyes using a PCNSL xenograft mouse model. Methods: We used indirect immunofluorescence double staining for CD20/CXCR4, CD20/CXCR5, CD20/CXCR7 and CD20/CD44 on enucleated eyes of a PCNSL xenograft mouse model with PVRL phenotype (PCNSL group) in comparison to a secondary CNS lymphoma xenograft mouse model (SCNSL group). Lymphoma infiltration was evaluated with an immunoreactive score (IRS). Results: 11/13 paired eyes of the PCNSL but none of the SCNSL group were infiltrated by CD20-positive cells. Particularly the choroid and to a lesser extent the retina of the PCNSL group were infiltrated by CD20+/CXCR4+, CD20+/CXCR5+, few CD20+/CD44+ but no CD20+/CXCR7+ cells. Expression of CXCR4 (p = 0.0205), CXCR5 (p = 0.0004) and CD44 (p < 0.0001) was significantly increased in the PCNSL compared to the SCNSL group. Conclusions: CD20+ PCNSL lymphoma cells infiltrating the eye co-express distinct homing receptors such as CXCR4 and CXCR5 in a PVRL homing mouse model. These receptors may be involved in PVRL homing into the eye.
Collapse
Affiliation(s)
- Neele Babst
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Lisa K. Isbell
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, 79106 Freiburg, Germany
| | - Felix Rommel
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Aysegul Tura
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Mahdy Ranjbar
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Salvatore Grisanti
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Cordula Tschuch
- Charles River Discovery Research Services GmbH, 79108 Freiburg, Germany
| | - Julia Schueler
- Charles River Discovery Research Services GmbH, 79108 Freiburg, Germany
| | - Soroush Doostkam
- Institute for Neuropathology, Medical Center—University of Freiburg, Faculty of Medicine, 79106 Freiburg, Germany
| | - Peter C. Reinacher
- Department of Stereotactic and Functional Neurosurgery, Medical Center—University of Freiburg, Faculty of Medicine, 79106 Freiburg, Germany
- Fraunhofer Institute for Laser Technology (ILT), 52074 Aachen, Germany
| | - Justus Duyster
- Department of Medicine I, Medical Center—University of Freiburg, Faculty of Medicine, 79106 Freiburg, Germany
- German Cancer Consortium (DKTK), Freiburg, Germany and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Vinodh Kakkassery
- Department of Ophthalmology, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, Medical Center, University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| |
Collapse
|
43
|
Wu W, Zhou C, Wang C, Zhou F. incidental finding of combined non-Hodgkin's lymphoma after radical nephrectomy for renal cell carcinoma: A case report and literature review. Asian J Surg 2022; 46:1737-1738. [PMID: 36270943 DOI: 10.1016/j.asjsur.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/03/2022] [Indexed: 11/26/2022] Open
|
44
|
Lv H, Wang T, Zhai S, Hou Z, Chen S. Dynamic transcriptome changes during osteogenic differentiation of bone marrow-derived mesenchymal stem cells isolated from chicken. Front Cell Dev Biol 2022; 10:940248. [PMID: 36120570 PMCID: PMC9478182 DOI: 10.3389/fcell.2022.940248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
Osteoblasts are indispensable for skeletal growth and maintenance. Bone marrow-derived mesenchymal stem cells (BMSCs) are useful in studying osteogenesis. In this study, BMSCs isolated from White Leghorns were differentiated into osteoblasts in vitro. Cells induced for -1, 0, 1, 11, and 22 d were used for transcriptomic analyses using the HISAT2-Stringtie-DESeq2 pipeline. Weighted correlation network analysis was processed to investigate significant modules, including differentially expressed genes (DEGs), correlated with osteogenic differentiation. Gene ontology and pathway enrichment analyses of DEGs were performed to elucidate the mechanisms of osteoblast differentiation. A total of 534, 1,144, 1,077, and 337 DEGs were identified between cells induced for -1 and 0, 0 and 1, 1 and 11, and 11 and 22 d, respectively (|log2FC| > 1.0, FDR <0.05). DEGs were mainly enriched in pathways related to cell proliferation in the early stage of osteogenic differentiation and pathways, such as the TGF-β signaling pathway, in the middle and late stages of osteogenic differentiation. A protein–protein interaction network of the 87 DEGs in the MEturquoise module within top 5-%-degree value was built utilizing the STRING database. This study is the first to elucidate the transcriptomic changes in the osteogenic differentiation of BMSCs isolated from White Leghorns at different times. Our results provide insight into the dynamic transcriptome changes during BMSC differentiation into osteoblasts in chicken.
Collapse
|
45
|
Chen Q, Zou J, He Y, Pan Y, Yang G, Zhao H, Huang Y, Zhao Y, Wang A, Chen W, Lu Y. A narrative review of circulating tumor cells clusters: A key morphology of cancer cells in circulation promote hematogenous metastasis. Front Oncol 2022; 12:944487. [PMID: 36059616 PMCID: PMC9434215 DOI: 10.3389/fonc.2022.944487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/18/2022] [Indexed: 11/28/2022] Open
Abstract
Circulating tumor cells (CTCs) that survive in the blood are playing an important role in the metastasis process of tumor. In addition, they have become a tool for tumor diagnosis, prognosis and recurrence monitoring. CTCs can exist in the blood as individual cells or as clumps of aggregated cells. In recent years, more and more studies have shown that clustered CTCs have stronger metastasis ability compared to single CTCs. With the deepening of studies, scholars have found that cancer cells can combine not only with each other, but also with non-tumor cells present in the blood, such as neutrophils, platelets, etc. At the same time, it was confirmed that non-tumor cells bound to CTCs maintain the survival and proliferation of cancer cells through a variety of ways, thus promoting the occurrence and development of tumor. In this review, we collected information on tumorigenesis induced by CTC clusters to make a summary and a discussion about them. Although CTC clusters have recently been considered as a key role in the transition process, many characteristics of them remain to be deeply explored. A detailed understanding of their vulnerability can prospectively pave the way for new inhibitors for metastasis.
Collapse
Affiliation(s)
- Qiong Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jueyao Zou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yong He
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanhong Pan
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pharmacy, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Gejun Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Han Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing, China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing, China
| | - Wenxing Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing, China
| |
Collapse
|
46
|
Ivanova EL, Costa B, Eisemann T, Lohr S, Boskovic P, Eichwald V, Meckler J, Jugold M, Orian-Rousseau V, Peterziel H, Angel P. CD44 expressed by myeloid cells promotes glioma invasion. Front Oncol 2022; 12:969787. [PMID: 35992852 PMCID: PMC9386454 DOI: 10.3389/fonc.2022.969787] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/11/2022] [Indexed: 12/07/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and malignant brain tumors in adulthood with a median survival of only 15 months. This poor prognosis is related to GBM’s ability to extensively infiltrate the surrounding brain parenchyma resulting in diffuse spread of neoplastic cells in the brain, responsible for high rate of recurrence. CD44 (Cluster of Differentiation 44) is a transmembrane protein, overexpressed in multiple cancer types, including gliomas, and implicated in cell motility, proliferation and angiogenesis. Multiple studies have investigated the role of CD44 in GBM cells and have highlighted a link between tumor malignancy and CD44 expression. However up to date, little is known of the role of CD44 on cells from the tumor microenvironment (TME). Here, we have investigated a potential role of CD44 in the TME in regards to GBM invasiveness. Using an ex-vivo organotypic brain slice invasion assay, we show that absence of CD44 from the TME impairs the ability of glioma cells to invade the surrounding brain parenchyma. By deleting CD44 in the astrocytic, endothelial and myeloid compartments, we show that it is specifically CD44 expression in myeloid cells that is responsible for the observed phenotype. Combining in vivo studies in cell-specific knock-out mice and in vitro analyses on primary microglia we demonstrate that myeloid CD44 is implicated in Toll Like Receptor 2 signaling and is a major regulator of Matrix metalloproteinase 9 expression.
Collapse
Affiliation(s)
- Ekaterina L. Ivanova
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Barbara Costa
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tanja Eisemann
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabrina Lohr
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pavle Boskovic
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Viktoria Eichwald
- Core Facility Small Animal Imaging Center, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jasmin Meckler
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manfred Jugold
- Core Facility Small Animal Imaging Center, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Veronique Orian-Rousseau
- Karlsruhe Institute of Technology (KIT), Institute of Biological and Chemical Systems – Functional Molecular Systems (IBCS-FMS), Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, Germany
| | - Heike Peterziel
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Angel
- Division of Signal Transduction and Growth Control, DKFZ/ZMBH Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
- *Correspondence: Peter Angel,
| |
Collapse
|
47
|
CD44 Contributes to the Regulation of MDR1 Protein and Doxorubicin Chemoresistance in Osteosarcoma. Int J Mol Sci 2022; 23:ijms23158616. [PMID: 35955749 PMCID: PMC9368984 DOI: 10.3390/ijms23158616] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma is the most common type of pediatric bone tumor. Despite great advances in chemotherapy during the past decades, the survival rates of osteosarcoma patients remain unsatisfactory. Drug resistance is one of the main reasons, leading to treatment failure and poor prognosis. Previous reports correlated expression of cluster of differentiation 44 (CD44) with drug resistance and poor survival of osteosarcoma patients, however the underlying mechanisms are poorly defined. Here, we investigated the role of CD44 in the regulation of drug chemoresistance, using osteosarcoma cells isolated from mice carrying a mutation of the tumor suppressor neurofibromatosis type 2 (Nf2) gene. CD44 expression was knocked-down in the cells using CRISPR/Cas9 approach. Subsequently, CD44 isoforms and mutants were re-introduced to investigate CD44-dependent processes. Sensitivity to doxorubicin was analyzed in the osteosarcoma cells with modified CD44 expression by immunoblot, colony formation- and WST-1 assay. To dissect the molecular alterations induced by deletion of Cd44, RNA sequencing was performed on Cd44-positive and Cd44-negative primary osteosarcoma tissues isolated from Nf2-mutant mice. Subsequently, expression of candidate genes was evaluated by quantitative reverse transcription PCR (qRT-PCR). Our results indicate that CD44 increases the resistance of osteosarcoma cells to doxorubicin by up-regulating the levels of multidrug resistance (MDR) 1 protein expression, and suggest the role of proteolytically released CD44 intracellular domain, and hyaluronan interactions in this process. Moreover, high throughput sequencing analysis identified differential regulation of several apoptosis-related genes in Cd44-positive and -negative primary osteosarcomas, including p53 apoptosis effector related to PMP-22 (Perp). Deletion of Cd44 in osteosarcoma cells led to doxorubicin-dependent p53 activation and a profound increase in Perp mRNA expression. Overall, our results suggest that CD44 might be an important regulator of drug resistance and suggest that targeting CD44 can sensitize osteosarcoma to standard chemotherapy.
Collapse
|
48
|
Tanaka T, Sano K, Munemura M, Hagimori M, Moriyama R, Yamamoto A, Ozaki KI, Munekane M, Yamasaki T, Mukai T. A radiolabeled nanoparticle probe coated with hyaluronic acid via electrostatic interaction to diagnose CD44-positive tumors. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
49
|
Ultra-wide, attomolar-level limit detection of CD44 biomarker with a silanized optical fiber biosensor. Biosens Bioelectron 2022; 208:114217. [DOI: 10.1016/j.bios.2022.114217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/02/2022] [Accepted: 03/22/2022] [Indexed: 12/15/2022]
|
50
|
Hu W, Fang T, Chen X. Identification of Differentially Expressed Genes and miRNAs for Ulcerative Colitis Using Bioinformatics Analysis. Front Genet 2022; 13:914384. [PMID: 35719390 PMCID: PMC9201719 DOI: 10.3389/fgene.2022.914384] [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: 04/06/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: Ulcerative colitis (UC) is a chronic inflammatory disease of the intestine whose cause and underlying mechanisms are not fully understood. The aim of this study was to use bioinformatics analysis to identify differentially expressed genes (DEGs) with diagnostic and therapeutic potential in UC.Materials and methods: Three UC datasets (GSE179285, GSE75214, GSE48958) were downloaded from the Gene Expression Omnibus (GEO) database. DEGs between normal and UC tissues were identified using the GEO2R online tool. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of the DEGs were performed using Metascape. Protein-protein interaction network (PPI) analysis and visualization using STRING and Cytoscape. Finally, the miRNA gene regulatory network was constructed by Cytoscape to predict potential microRNAs (miRNAs) associated with DEGs.Results: A total of 446 DEGs were identified, consisting of 309 upregulated genes and 137 downregulated genes. The enriched functions and pathways of the DEGs include extracellular matrix, regulation of cell adhesion, inflammatory response, response to cytokine, monocarboxylic acid metabolic process, response to toxic substance. The analysis of KEGG pathway indicates that the DEGs were significantly enriched in Complement and coagulation cascades, Amoebiasis, TNF signaling pathway, bile secretion, and Mineral absorption. Combining the results of the PPI network and CytoHubba, 9 hub genes including CXCL8, ICAM1, CXCR4, CD44, IL1B, MMP9, SPP1, TIMP1, and HIF1A were selected. Based on the DEG-miRNAs network construction, 7 miRNAs including miR-335-5p, mir-204-5p, miR-93-5p, miR106a-5p, miR-21-5p, miR-146a-5p, and miR-155-5p were identified as potential critical miRNAs.Conclusion: In summary, we identified DEGs that may be involved in the progression or occurrence of UC. A total of 446 DEGs,9 hub genes and 7 miRNAs were identified, which may be considered as biomarkers of UC. Further studies, however, are needed to elucidate the biological functions of these genes in UC.
Collapse
Affiliation(s)
- Weitao Hu
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Taiyong Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xiaoqing Chen
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- *Correspondence: Xiaoqing Chen,
| |
Collapse
|