151
|
Jahanbani Y, Shafiee S, Davaran S, Roshangar L, Ahmadian E, Eftekhari A, Dolati S, Yousefi M. Stem cells technology as a platform for generating reproductive system organoids and treatment of infertility-related diseases. Cell Biol Int 2022; 46:512-522. [PMID: 34918417 DOI: 10.1002/cbin.11747] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/14/2021] [Indexed: 02/06/2023]
Abstract
In recent years, stem cells have known as a helpful biological tool for the accurate diagnosis, treatment and recognition of diseases. Using stem cells as biomarkers have presented high potential in the early detection of many diseases. Another advancement in stem cell technology includes stem cell derived organoids model that could be a promising platform for diagnosis and modeling different diseases. Furthermore, therapeutic capabilities of stem cell therapy have increased hope in the face of different disability managements. All of these technologies are also widely used in reproductive related diseases especially in today's world that many couples encounter infertility problems. However, with the aid of numerous improvements in the treatment of infertility, over 80% of couples who dreamed of having children could now have children. Due to the fact that infertility has many negative effects on personal and social lives of young couples, many researchers have focused on the treatment of male and female reproductive system abnormalities with different types of stem cells, including embryonic stem cells, bone marrow mesenchymal stem cells (MSCs), and umbilical cord-derived MSCs. Also, design and formation of reproductive system organoids provide a fascinating window into disease modeling, drug screening, personalized therapy, and regeneration medicine. Utilizing these techniques to study, model and treat the infertility-related diseases has drawn attention of many scientists. This review explains different applications of stem cells in generating reproductive system organoids and stem cell-based therapies for male and female infertility related diseases treatment.
Collapse
Affiliation(s)
- Yalda Jahanbani
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Shafiee
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soodabeh Davaran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Eftekhari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Russian Institute for Advanced Study, Moscow State Pedagogical University, Moscow, Russian Federation
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
152
|
Liang H, Zhang F, Hong Y, Wu Y, Xie H, Zhang C, Wang Z, Lu Z, Yang H. Synergistic Silencing of Skp2 by siRNA Self-Assembled Nanoparticles as a Therapeutic Strategy for Advanced Prostate Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106046. [PMID: 35182014 DOI: 10.1002/smll.202106046] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/21/2022] [Indexed: 06/14/2023]
Abstract
Advanced prostate cancer, harboring multiple mutations of tumor suppressor genes, is refractory to conventional therapies. Knockout of the Skp2 gene blocks pRb/p53 doubly deficient prostate cancer in mice, which inspired the authors to develop an approach for delivering siRNA that would efficiently silence Skp2 (siSkp2) in vivo. Here, a facile strategy is reported to directly assemble siSkp2 with the natural compound quercetin (Que) into supramolecular nanoparticles (NPs). This carrier-free siSkp2 delivery system could effectively protect siSkp2 from degradation in serum and enhance its cellular internalization. Furthermore, the siSkp2/Que NPs exhibit synergistic effects in Skp2 silencing, because they can degrade the mRNA and protein of Skp2 simultaneously. Indeed, siSkp2/Que NPs remarkably diminish the Skp2 abundance and further inhibit the proliferation and migration of TMU cells (RB1/TP53/KRAS triple mutations) in vitro. The in vivo results further show that i.v. administration of siSkp2/Que NPs efficiently accumulates in tumor sites and strongly inhibits the growth of TMU tumors in nude mice. Importantly, the siSkp2/Que NPs do not induce any abnormality in the treated mice, which suggests satisfactory biocompatibility. Collectively, this study describes a tractable siRNA self-assembled strategy for Skp2 silencing, which might be a promising nanodrug to cure multitherapy-resistant advanced prostate cancer.
Collapse
Affiliation(s)
- Hong Liang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, P. R. China
- Fujian Key Laboratory of Functional Marine Sensing Materials, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, 350108, P. R. China
| | - Fangming Zhang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Yannv Hong
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Yue Wu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Huanzhang Xie
- Fujian Key Laboratory of Functional Marine Sensing Materials, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, 350108, P. R. China
| | - Chen Zhang
- Fujian Key Laboratory of Functional Marine Sensing Materials, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, 350108, P. R. China
| | - Zonghua Wang
- Fujian Key Laboratory of Functional Marine Sensing Materials, Fuzhou Institute of Oceanography, Minjiang University, Fuzhou, 350108, P. R. China
| | - Zhonglei Lu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, P. R. China
| |
Collapse
|
153
|
Cao J, Chan WC, Chow MSS. Use of conditional reprogramming cell, patient derived xenograft and organoid for drug screening for individualized prostate cancer therapy: Current and future perspectives (Review). Int J Oncol 2022; 60:52. [PMID: 35322860 DOI: 10.3892/ijo.2022.5342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 01/14/2022] [Indexed: 11/06/2022] Open
Abstract
Prostate cancer mortality is ranked second among all cancer mortalities in men worldwide. There is a great need for a method of efficient drug screening for precision therapy, especially for patients with existing drug‑resistant prostate cancer. Based on the concept of bacterial cell culture and drug sensitivity testing, the traditional approach of cancer drug screening is inadequate. The current and more innovative use of cancer cell culture and in vivo tumor models in drug screening for potential individualization of anti‑cancer therapy is reviewed and discussed in the present review. An ideal screening model would have the ability to identify drug activity for the targeted cells resembling what would have occurred in the in vivo environment. Based on this principle, three available cell culture/tumor screening models for prostate cancer are reviewed and considered. The culture conditions, advantages and disadvantages for each model together with ideas to best utilize these models are discussed. The first screening model uses conditional reprogramed cells derived from patient cancer cells. Although these cells are convenient to grow and use, they are likely to have different markers and characteristics from original tumor cells and thus not likely to be informative. The second model employs patient derived xenograft (PDX) which resembles an in vivo approach, but its main disadvantages are that it cannot be easily genetically modified and it is not suitable for high‑throughput drug screening. Finally, high‑throughput screening is more feasible with tumor organoids grown from patient cancer cells. The last system still needs a large number of tumor cells. It lacks in situ blood vessels, immune cells and the extracellular matrix. Based on these current models, future establishment of an organoid data bank would allow the selection of a specific organoid resembling that of an individual's prostate cancer and used for screening of suitable anticancer drugs. This can be further confirmed using the PDX model. Thus, this combined organoid‑PDX approach is expected to be able to provide the drug sensitivity testing approach for individualization of prostate cancer therapy in the near future.
Collapse
Affiliation(s)
- Jessica Cao
- College of Osteopathic Medicine of The Pacific, Western University of Health Sciences, Pomona, CA 91766‑1854, USA
| | - Wing C Chan
- City of Hope Comprehensive Cancer Center, City of Hope Medical Center, Duarte, CA 91010‑3012, USA
| | - Moses S S Chow
- College of Pharmacy, Western University of Health Sciences, Pomona, CA 91766‑1854, USA
| |
Collapse
|
154
|
Lee S, Mendoza TR, Burner DN, Muldong MT, Wu CCN, Arreola-Villanueva C, Zuniga A, Greenburg O, Zhu WY, Murtadha J, Koutouan E, Pineda N, Pham H, Kang SG, Kim HT, Pineda G, Lennon KM, Cacalano NA, Jamieson CHM, Kane CJ, Kulidjian AA, Gaasterland T, Jamieson CAM. Novel Dormancy Mechanism of Castration Resistance in Bone Metastatic Prostate Cancer Organoids. Int J Mol Sci 2022; 23:ijms23063203. [PMID: 35328625 PMCID: PMC8952299 DOI: 10.3390/ijms23063203] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/07/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Advanced prostate cancer (PCa) patients with bone metastases are treated with androgen pathway directed therapy (APDT). However, this treatment invariably fails and the cancer becomes castration resistant. To elucidate resistance mechanisms and to provide a more predictive pre-clinical research platform reflecting tumor heterogeneity, we established organoids from a patient-derived xenograft (PDX) model of bone metastatic prostate cancer, PCSD1. APDT-resistant PDX-derived organoids (PDOs) emerged when cultured without androgen or with the anti-androgen, enzalutamide. Transcriptomics revealed up-regulation of neurogenic and steroidogenic genes and down-regulation of DNA repair, cell cycle, circadian pathways and the severe acute respiratory syndrome (SARS)-CoV-2 host viral entry factors, ACE2 and TMPRSS2. Time course analysis of the cell cycle in live cells revealed that enzalutamide induced a gradual transition into a reversible dormant state as shown here for the first time at the single cell level in the context of multi-cellular, 3D living organoids using the Fucci2BL fluorescent live cell cycle tracker system. We show here a new mechanism of castration resistance in which enzalutamide induced dormancy and novel basal-luminal-like cells in bone metastatic prostate cancer organoids. These PDX organoids can be used to develop therapies targeting dormant APDT-resistant cells and host factors required for SARS-CoV-2 viral entry.
Collapse
MESH Headings
- Androgens/pharmacology
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensin-Converting Enzyme 2/metabolism
- Animals
- Benzamides/pharmacology
- Bone Neoplasms/genetics
- Bone Neoplasms/metabolism
- Bone Neoplasms/secondary
- COVID-19/genetics
- COVID-19/metabolism
- COVID-19/virology
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Profiling/methods
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Male
- Mice
- Nitriles/pharmacology
- Organoids/metabolism
- Phenylthiohydantoin/pharmacology
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- SARS-CoV-2/metabolism
- SARS-CoV-2/physiology
- Serine Endopeptidases/genetics
- Serine Endopeptidases/metabolism
- Transplantation, Heterologous
- Virus Internalization
Collapse
Affiliation(s)
- Sanghee Lee
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
- Rady Children’s Hospital, San Diego, CA 92123, USA
| | - Theresa R. Mendoza
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Danielle N. Burner
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Michelle T. Muldong
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Christina C. N. Wu
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (G.P.); (K.M.L.)
| | - Catalina Arreola-Villanueva
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Abril Zuniga
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Olga Greenburg
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - William Y. Zhu
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Jamillah Murtadha
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Evodie Koutouan
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Naomi Pineda
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Hao Pham
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | - Sung-Gu Kang
- Department of Urology, Korea University College of Medicine, Seongbuk-Gu, Seoul 02841, Korea;
| | - Hyun Tae Kim
- Department of Urology, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
| | - Gabriel Pineda
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (G.P.); (K.M.L.)
| | - Kathleen M. Lennon
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (G.P.); (K.M.L.)
| | - Nicholas A. Cacalano
- Department of Radiation Oncology, University of California, Los Angeles, CA 90095, USA;
| | - Catriona H. M. Jamieson
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
- Department of Urology, Korea University College of Medicine, Seongbuk-Gu, Seoul 02841, Korea;
| | - Christopher J. Kane
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
| | | | - Terry Gaasterland
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA 92093, USA;
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Christina A. M. Jamieson
- Department of Urology, University of California San Diego, La Jolla, CA 92093, USA; (S.L.); (T.R.M.); (D.N.B.); (M.T.M.); (C.A.-V.); (A.Z.); (O.G.); (W.Y.Z.); (J.M.); (E.K.); (N.P.); (H.P.); (C.J.K.)
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA; (C.C.N.W.); (C.H.M.J.)
- Correspondence: ; Tel.: +1-858-534-2921
| |
Collapse
|
155
|
Yao L, Zao XL, Pan XF, Zhang HG, Wang FJ, Qiao PF. Application of tumoroids derived from advanced colorectal cancer patients to predict individual response to chemotherapy. J Chemother 2022; 35:104-116. [PMID: 35285783 DOI: 10.1080/1120009x.2022.2045827] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Therapeutic approaches of advanced colorectal cancer are more complex, here we present a living biobank of patient-derived tumoroids from advanced colorectal cancer patients and show examples of how these tumoroids can be used to to simulate cancer behavior ex vivo and provide more evidence for tumoroids could be utilized as a predictive platform during chemotherapy treatment to identify the chemotherapy response. Morphological, histological and genomic characterization analysis of colorectal cancer tumoroids was conducted. Further, we treated colorectal cancer tumoroids with different drugs to detect cellular activities to evaluate drug sensitivity using CellTiter-Glo 3 D cell viability assay. Then the drug sensitivity of tumoroids was compared with clinical outcomes. Our results implied that tumoroids recapitulated the histological features of the original tumours and genotypic profiling of tumoroids showed a high-level of similarity to the matched primary tumours. Dose-response curves, area under the curve and tumour inhibitory rate of each therapeutic profiling calculations in tumoroids demonstrated a great diversity and we gained 88.24% match ratio between the sensitivity data of tumoroids with their paired patients' clinical outcomes. tumour inhibitory rate of each treatment parameters in tumoroids performed positive correlation with progression-free survival while area under the curve of each treatment parameters performed negative correlation with progression-free survival of the corresponding patients. In summary, We presented a living biobank of tumoroids from advanced colorectal cancer patients and show tumoroids got great potential for predicting clinical responses to chemotherapy treatment of advanced colorectal cancer.
Collapse
Affiliation(s)
- Lei Yao
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao-Long Zao
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiao-Fei Pan
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao-Gang Zhang
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fu-Jing Wang
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Peng-Fei Qiao
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
156
|
Luo L, Ma Y, Zheng Y, Su J, Huang G. Application Progress of Organoids in Colorectal Cancer. Front Cell Dev Biol 2022; 10:815067. [PMID: 35273961 PMCID: PMC8902504 DOI: 10.3389/fcell.2022.815067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/31/2022] [Indexed: 12/24/2022] Open
Abstract
Currently, colorectal cancer is still the third leading cause of cancer-related mortality, and the incidence is rising. It is a long time since the researchers used cancer cell lines and animals as the study subject. However, these models possess various limitations to reflect the cancer progression in the human body. Organoids have more clinical significance than cell lines, and they also bridge the gap between animal models and humans. Patient-derived organoids are three-dimensional cultures that simulate the tumor characteristics in vivo and recapitulate tumor cell heterogeneity. Therefore, the emergence of colorectal cancer organoids provides an unprecedented opportunity for colorectal cancer research. It retains the molecular and cellular composition of the original tumor and has a high degree of homology and complexity with patient tissues. Patient-derived colorectal cancer organoids, as personalized tumor organoids, can more accurately simulate colorectal cancer patients’ occurrence, development, metastasis, and predict drug response in colorectal cancer patients. Colorectal cancer organoids show great potential for application, especially preclinical drug screening and prediction of patient response to selected treatment options. Here, we reviewed the application of colorectal cancer organoids in disease model construction, basic biological research, organoid biobank construction, drug screening and personalized medicine, drug development, drug toxicity and safety, and regenerative medicine. In addition, we also displayed the current limitations and challenges of organoids and discussed the future development direction of organoids in combination with other technologies. Finally, we summarized and analyzed the current clinical trial research of organoids, especially the clinical trials of colorectal cancer organoids. We hoped to lay a solid foundation for organoids used in colorectal cancer research.
Collapse
Affiliation(s)
- Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.,Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Yucui Ma
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Yilin Zheng
- Clinical Research Center, Shantou Central Hospital, Shantou, China
| | - Jiating Su
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
| | - Guoxin Huang
- Clinical Research Center, Shantou Central Hospital, Shantou, China
| |
Collapse
|
157
|
Foo MA, You M, Chan SL, Sethi G, Bonney GK, Yong WP, Chow EKH, Fong ELS, Wang L, Goh BC. Clinical translation of patient-derived tumour organoids- bottlenecks and strategies. Biomark Res 2022; 10:10. [PMID: 35272694 PMCID: PMC8908618 DOI: 10.1186/s40364-022-00356-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
Multiple three-dimensional (3D) tumour organoid models assisted by multi-omics and Artificial Intelligence (AI) have contributed greatly to preclinical drug development and precision medicine. The intrinsic ability to maintain genetic and phenotypic heterogeneity of tumours allows for the reconciliation of shortcomings in traditional cancer models. While their utility in preclinical studies have been well established, little progress has been made in translational research and clinical trials. In this review, we identify the major bottlenecks preventing patient-derived tumour organoids (PDTOs) from being used in clinical setting. Unsuitable methods of tissue acquisition, disparities in establishment rates and a lengthy timeline are the limiting factors for use of PDTOs in clinical application. Potential strategies to overcome this include liquid biopsies via circulating tumour cells (CTCs), an automated organoid platform and optical metabolic imaging (OMI). These proposed solutions accelerate and optimize the workflow of a clinical organoid drug screening. As such, PDTOs have the potential for potential applications in clinical oncology to improve patient outcomes. If remarkable progress is made, cancer patients can finally benefit from this revolutionary technology.
Collapse
Affiliation(s)
- Malia Alexandra Foo
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Mingliang You
- Hangzhou Cancer Institute, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, 31002, China.,Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 31002, China
| | - Shing Leng Chan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Surgery, National University Hospital, Singapore, Singapore
| | - Gautam Sethi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Glenn K Bonney
- Department of Surgery, National University Hospital, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei-Peng Yong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore, Singapore
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Eliza Li Shan Fong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore. .,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Boon-Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore. .,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore, Singapore.
| |
Collapse
|
158
|
Cambuli F, Foletto V, Alaimo A, De Felice D, Gandolfi F, Palumbieri MD, Zaffagni M, Genovesi S, Lorenzoni M, Celotti M, Bertossio E, Mazzero G, Bertossi A, Bisio A, Berardinelli F, Antoccia A, Gaspari M, Barbareschi M, Fiorentino M, Shen MM, Loda M, Romanel A, Lunardi A. Intra-epithelial non-canonical Activin A signaling safeguards prostate progenitor quiescence. EMBO Rep 2022; 23:e54049. [PMID: 35253958 PMCID: PMC9066067 DOI: 10.15252/embr.202154049] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 01/21/2023] Open
Abstract
The healthy prostate is a relatively quiescent tissue. Yet, prostate epithelium overgrowth is a common condition during aging, associated with urinary dysfunction and tumorigenesis. For over thirty years, TGF-β ligands have been known to induce cytostasis in a variety of epithelia, but the intracellular pathway mediating this signal in the prostate, and its relevance for quiescence, have remained elusive. Here, using mouse prostate organoids to model epithelial progenitors, we find that intra-epithelial non-canonical Activin A signaling inhibits cell proliferation in a Smad-independent manner. Mechanistically, Activin A triggers Tak1 and p38 ΜAPK activity, leading to p16 and p21 nuclear import. Spontaneous evasion from this quiescent state occurs upon prolonged culture, due to reduced Activin A secretion, a condition associated with DNA replication stress and aneuploidy. Organoids capable to escape quiescence in vitro are also able to implant with increased frequency into immunocompetent mice. This study demonstrates that non-canonical Activin A signaling safeguards epithelial quiescence in the healthy prostate, with potential implications for the understanding of cancer initiation, and the development of therapies targeting quiescent tumor progenitors.
Collapse
Affiliation(s)
- Francesco Cambuli
- The Armenise‐Harvard Laboratory of Cancer Biology & GeneticsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly,Department of Medicine, Genetics and DevelopmentUrologySystems BiologyHerbert Irving Comprehensive Cancer CenterColumbia University Irving Medical CenterNew YorkNYUSA,Present address:
Molecular Pharmacology ProgramSloan Kettering InstituteMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Veronica Foletto
- The Armenise‐Harvard Laboratory of Cancer Biology & GeneticsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Alessandro Alaimo
- The Armenise‐Harvard Laboratory of Cancer Biology & GeneticsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Dario De Felice
- The Armenise‐Harvard Laboratory of Cancer Biology & GeneticsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Francesco Gandolfi
- Laboratory of Bioinformatics and Computational GenomicsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Maria Dilia Palumbieri
- The Armenise‐Harvard Laboratory of Cancer Biology & GeneticsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Michela Zaffagni
- The Armenise‐Harvard Laboratory of Cancer Biology & GeneticsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Sacha Genovesi
- The Armenise‐Harvard Laboratory of Cancer Biology & GeneticsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Marco Lorenzoni
- The Armenise‐Harvard Laboratory of Cancer Biology & GeneticsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Martina Celotti
- The Armenise‐Harvard Laboratory of Cancer Biology & GeneticsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Emiliana Bertossio
- The Armenise‐Harvard Laboratory of Cancer Biology & GeneticsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | | | - Arianna Bertossi
- The Armenise‐Harvard Laboratory of Cancer Biology & GeneticsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Alessandra Bisio
- The Armenise‐Harvard Laboratory of Cancer Biology & GeneticsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Francesco Berardinelli
- Department of ScienceUniversity of Roma TreRomaItaly,Laboratory of Neurodevelopment, Neurogenetics and Molecular Neurobiology UnitIRCCS Santa Lucia FoundationRomaItaly
| | | | - Marco Gaspari
- Department of Experimental and Clinical MedicineUniversity of CatanzaroCatanzaroItaly
| | | | - Michelangelo Fiorentino
- Department of Experimental, Diagnostic and Specialty MedicineUniversity of BolognaBolognaItaly
| | - Michael M Shen
- Department of Medicine, Genetics and DevelopmentUrologySystems BiologyHerbert Irving Comprehensive Cancer CenterColumbia University Irving Medical CenterNew YorkNYUSA
| | - Massimo Loda
- Department of Pathology and Laboratory MedicineWeill Medical College of Cornell UniversityNew YorkNYUSA
| | - Alessandro Romanel
- Laboratory of Bioinformatics and Computational GenomicsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Andrea Lunardi
- The Armenise‐Harvard Laboratory of Cancer Biology & GeneticsDepartment of Cellular, Computational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| |
Collapse
|
159
|
Lorenzoni M, De Felice D, Beccaceci G, Di Donato G, Foletto V, Genovesi S, Bertossi A, Cambuli F, Lorenzin F, Savino A, Avalle L, Cimadamore A, Montironi R, Weber V, Carbone FG, Barbareschi M, Demichelis F, Romanel A, Poli V, Del Sal G, Julio MKD, Gaspari M, Alaimo A, Lunardi A. ETS-related gene (ERG) undermines genome stability in mouse prostate progenitors via Gsk3β dependent Nkx3.1 degradation. Cancer Lett 2022; 534:215612. [PMID: 35259458 PMCID: PMC8968219 DOI: 10.1016/j.canlet.2022.215612] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 12/13/2022]
Abstract
21q22.2–3 deletion is the most common copy number alteration in prostate cancer (PCa). The genomic rearrangement results in the androgen-dependent de novo expression of ETS-related gene (ERG) in prostate cancer cells, a condition promoting tumor progression to advanced stages of the disease. Interestingly, ERG expression characterizes 5–30% of tumor precursor lesions – High Grade Prostatic Intraepithelial Neoplasia (HGPIN) - where its role remains unclear. Here, by combining organoids technology with Click-chemistry coupled Mass Spectrometry, we demonstrate a prominent role of ERG in remodeling the protein secretome of prostate progenitors. Functionally, by lowering autocrine Wnt-4 signaling, ERG represses canonical Wnt pathway in prostate progenitors, and, in turn, promotes the accumulation of DNA double strand breaks via Gsk3β-dependent degradation of the tumor suppressor Nkx3.1. On the other hand, by shaping extracellular paracrine signals, ERG strengthens the pro-oxidative transcriptional signature of inflammatory macrophages, which we demonstrate to infiltrate pre-malignant ERG positive prostate lesions. These findings highlight previously unrecognized functions of ERG in undermining adult prostate progenitor niche through cell autonomous and non-autonomous mechanisms. Overall, by supporting the survival and proliferation of prostate progenitors in the absence of growth stimuli and promoting the accumulation of DNA damage through destabilization of Nkx3.1, ERG could orchestrate the prelude to neoplastic transformation. Expression of ERGM40 in mouse prostate organoids promotes their survival and growth in the absence of Egf. ERGM40 alters the extracellular signaling network of mouse prostate organoids. Canonical Wnt pathway is substantially reduced in ERG + prostate organoids due to decreased autocrine signaling of Wnt4. Gsk3b promotes Nkx3.1 proteolysis and, in turn, accumulation of double strand breaks in ERG + prostate organoids. Paracrine signaling of ERG + prostate organoids modulates Arginase 1 expression in M1-polarized macrophages.
Collapse
Affiliation(s)
- Marco Lorenzoni
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Dario De Felice
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Giulia Beccaceci
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Giorgia Di Donato
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Veronica Foletto
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Sacha Genovesi
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Arianna Bertossi
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Francesco Cambuli
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Francesca Lorenzin
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Aurora Savino
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Lidia Avalle
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Alessia Cimadamore
- Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, Ancona, Italy
| | - Rodolfo Montironi
- Molecular Medicine and Cell Therapy Foundation, Polytechnic University of the Marche Region, Via Tronto, 10, Ancona, Italy
| | - Veronica Weber
- Unit of Surgical Pathology, Santa Chiara Hospital, Trento, Italy
| | | | | | - Francesca Demichelis
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Alessandro Romanel
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Valeria Poli
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Giannino Del Sal
- University of Trieste Department Life Sciences, ICGEB-Area Science Park Trieste, IFOM, Milan, Italy
| | - Marianna Kruithof-de Julio
- Urology Research Laboratory, Department for BioMedical Research DBMR, University of Bern, Bern, Switzerland; Translational Organoid Resource CORE, Department for BioMedical Research, University of Bern, Bern, Switzerland; Bern Center for Precision Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland; Department of Urology, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Marco Gaspari
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, Catanzaro, Italy.
| | - Alessandro Alaimo
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy.
| | - Andrea Lunardi
- Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy.
| |
Collapse
|
160
|
Koushyar S, Meniel VS, Phesse TJ, Pearson HB. Exploring the Wnt Pathway as a Therapeutic Target for Prostate Cancer. Biomolecules 2022; 12:309. [PMID: 35204808 PMCID: PMC8869457 DOI: 10.3390/biom12020309] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/09/2022] [Accepted: 02/12/2022] [Indexed: 12/24/2022] Open
Abstract
Aberrant activation of the Wnt pathway is emerging as a frequent event during prostate cancer that can facilitate tumor formation, progression, and therapeutic resistance. Recent discoveries indicate that targeting the Wnt pathway to treat prostate cancer may be efficacious. However, the functional consequence of activating the Wnt pathway during the different stages of prostate cancer progression remains unclear. Preclinical work investigating the efficacy of targeting Wnt signaling for the treatment of prostate cancer, both in primary and metastatic lesions, and improving our molecular understanding of treatment responses is crucial to identifying effective treatment strategies and biomarkers that help guide treatment decisions and improve patient care. In this review, we outline the type of genetic alterations that lead to activated Wnt signaling in prostate cancer, highlight the range of laboratory models used to study the role of Wnt genetic drivers in prostate cancer, and discuss new mechanistic insights into how the Wnt cascade facilitates prostate cancer growth, metastasis, and drug resistance.
Collapse
Affiliation(s)
- Sarah Koushyar
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (S.K.); (V.S.M.)
- School of Life Sciences, Pharmacy and Chemistry, Faculty of Science, Engineering and Computing, Kingston University, Penrhyn Road, Kingston Upon Thames KT1 2EE, UK
| | - Valerie S. Meniel
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (S.K.); (V.S.M.)
| | - Toby J. Phesse
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (S.K.); (V.S.M.)
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne 3000, Australia
| | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (S.K.); (V.S.M.)
| |
Collapse
|
161
|
Schwarz FM, Schniewind I, Besso MJ, Lange S, Linge A, Patil SG, Löck S, Klusa D, Dietrich A, Voss-Böhme A, Nowrouzi A, Krause M, Dubrovska A, Kurth I, Peitzsch C. Plasticity within aldehyde dehydrogenase-positive cells determines prostate cancer radiosensitivity. Mol Cancer Res 2022; 20:794-809. [PMID: 35135863 DOI: 10.1158/1541-7786.mcr-21-0806] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 01/08/2022] [Accepted: 02/02/2022] [Indexed: 11/16/2022]
Abstract
Tumor heterogeneity and cellular plasticity are key determinants of tumor progression, metastatic spread and therapy response driven by the cancer stem cell (CSC) population. Within the present study, we analyzed irradiation-induced plasticity within the aldehyde dehydrogenase (ALDH)-positive population in prostate cancer (PCa). The radiosensitivity of xenograft tumors derived from ALDH+ and ALDH- cells was determined with local tumor control analyses and demonstrated different dose-response profiles, time to relapse and focal adhesion signaling. The transcriptional heterogeneity was analyzed in pools of ten DU145 and PC3 cells with multiplex gene expression analyses and illustrated a higher degree of heterogeneity within the ALDH+ population that even increases upon irradiation in comparison to ALDH- cells. Phenotypic conversion and clonal competition were analyzed with fluorescence protein-labeled cells to distinguish cellular origins in competitive 3D cultures and xenograft tumors. We found that the ALDH+ population outcompetes ALDH- cells and drives tumor growth, in particular upon irradiation. The observed dynamics of the cellular state compositions between ALDH+ and ALDH- cells in vivo before and after tumor irradiation was reproduced by a probabilistic Markov compartment model that incorporates cellular plasticity, clonal competition and phenotype-specific radiosensitivities. Transcriptional analyses indicate that the cellular conversion from ALDH- into ALDH+ cells within xenograft tumors under therapeutic pressure was partially mediated through induction of the transcriptional repressor SNAI2. In summary, irradiation-induced cellular conversion events are present in xenograft tumors derived from PCa cells and may be responsible for radiotherapy failure.
Collapse
Affiliation(s)
- Franziska M Schwarz
- Institute of Radiooncology - OncoRay, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- OncoRay - National Center for Radiation Research in Oncology, National Center for Radiation Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Iñaki Schniewind
- OncoRay - National Center for Radiation Research in Oncology, National Center for Radiation Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Department of Neurology, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Maria J Besso
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steffen Lange
- Faculty of Informatics/Mathematics, University of Applied Science, Dresden, Germany
| | - Annett Linge
- Institute of Radiooncology - OncoRay, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- OncoRay - National Center for Radiation Research in Oncology, National Center for Radiation Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - Shivaprasad G Patil
- Institute of Radiooncology - OncoRay, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- OncoRay - National Center for Radiation Research in Oncology, National Center for Radiation Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Steffen Löck
- OncoRay - National Center for Radiation Research in Oncology, National Center for Radiation Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daria Klusa
- Institute of Radiooncology - OncoRay, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- OncoRay - National Center for Radiation Research in Oncology, National Center for Radiation Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - Antje Dietrich
- OncoRay - National Center for Radiation Research in Oncology, National Center for Radiation Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anja Voss-Böhme
- Faculty of Informatics/Mathematics, University of Applied Science, Dresden, Germany
| | - Ali Nowrouzi
- Clinical Cooperation Unit Translational Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, Heidelberg University Hospital (UKHD), National Center for Tumor Diseases (NCT) Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
- Heidelberg Ion-Beam Therapy Center (HIT), Heidelberg, Germany
| | - Mechthild Krause
- Institute of Radiooncology - OncoRay, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- OncoRay - National Center for Radiation Research in Oncology, National Center for Radiation Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - Anna Dubrovska
- Institute of Radiooncology - OncoRay, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- OncoRay - National Center for Radiation Research in Oncology, National Center for Radiation Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ina Kurth
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University, Heidelberg, Germany
| | - Claudia Peitzsch
- Institute of Radiooncology - OncoRay, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- OncoRay - National Center for Radiation Research in Oncology, National Center for Radiation Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| |
Collapse
|
162
|
Rawlings TM, Makwana K, Tryfonos M, Lucas ES. Organoids to model the endometrium: implantation and beyond. REPRODUCTION AND FERTILITY 2022; 2:R85-R101. [PMID: 35118399 PMCID: PMC8801025 DOI: 10.1530/raf-21-0023] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/05/2021] [Indexed: 12/18/2022] Open
Abstract
Despite advances in assisted reproductive techniques in the 4 decades since the first human birth after in vitro fertilisation, 1–2% of couples experience recurrent implantation failure, and some will never achieve a successful pregnancy even in the absence of a confirmed dysfunction. Furthermore, 1–2% of couples who do conceive, either naturally or with assistance, will experience recurrent early loss of karyotypically normal pregnancies. In both cases, embryo-endometrial interaction is a clear candidate for exploration. The impossibility of studying implantation processes within the human body has necessitated the use of animal models and cell culture approaches. Recent advances in 3-dimensional modelling techniques, namely the advent of organoids, present an exciting opportunity to elucidate the unanswerable within human reproduction. In this review, we will explore the ontogeny of implantation modelling and propose a roadmap to application and discovery.
Collapse
Affiliation(s)
- Thomas M Rawlings
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Komal Makwana
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Maria Tryfonos
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Emma S Lucas
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK.,Centre for Early Life, Warwick Medical School, University of Warwick, Coventry, UK
| |
Collapse
|
163
|
Ali A, Du Feu A, Oliveira P, Choudhury A, Bristow RG, Baena E. Prostate zones and cancer: lost in transition? Nat Rev Urol 2022; 19:101-115. [PMID: 34667303 DOI: 10.1038/s41585-021-00524-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2021] [Indexed: 12/16/2022]
Abstract
Localized prostate cancer shows great clinical, genetic and environmental heterogeneity; however, prostate cancer treatment is currently guided solely by clinical staging, serum PSA levels and histology. Increasingly, the roles of differential genomics, multifocality and spatial distribution in tumorigenesis are being considered to further personalize treatment. The human prostate is divided into three zones based on its histological features: the peripheral zone (PZ), the transition zone (TZ) and the central zone (CZ). Each zone has variable prostate cancer incidence, prognosis and outcomes, with TZ prostate tumours having better clinical outcomes than PZ and CZ tumours. Molecular and cell biological studies can improve understanding of the unique molecular, genomic and zonal cell type features that underlie the differences in tumour progression and aggression between the zones. The unique biology of each zonal tumour type could help to guide individualized treatment and patient risk stratification.
Collapse
Affiliation(s)
- Amin Ali
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK.,The Christie NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Alexander Du Feu
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Pedro Oliveira
- The Christie NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Ananya Choudhury
- The Christie NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK.,The University of Manchester, Manchester Cancer Research Centre, Manchester, UK.,Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Robert G Bristow
- The Christie NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK.,The University of Manchester, Manchester Cancer Research Centre, Manchester, UK.,Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK
| | - Esther Baena
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK. .,Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, UK.
| |
Collapse
|
164
|
da Silva Lima D, da Silva Gomes L, de Sousa Figueredo E, E Silva YIF, Silva EM, de Souza Bovi T, Taboga SR, Marques MR, Biancardi MF, Dos Santos FCA. Subacute exposure to aluminum chloride causes prolonged morphological insults in the ventral male prostate and in the female prostate of adult gerbils. ENVIRONMENTAL TOXICOLOGY 2022; 37:299-309. [PMID: 34726835 DOI: 10.1002/tox.23398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/07/2021] [Accepted: 10/22/2021] [Indexed: 06/13/2023]
Abstract
Aluminum (Al) is a widespread metal in the environment, and is found in fresh or processed foods, household utensils, packaging, and medicines. In addition to its high toxicity, Al can also have estrogenic agonistic effects on target organs. Considering that the Al effects on the prostate are little known, the aim of this study was to evaluate the impact of aluminum chloride (AlCl3 ) subacute exposure on the morphophysiology of the male ventral prostate and the female prostate of adult gerbils. Furthermore, the glandular restoration capacity in face of the Al insults was evaluated in gerbils that were submitted to 30 days of recovery. Male and female gerbils were orally exposed to AlCl3 (10 mg/kg) for 30 consecutive days. The animals were euthanized 1 day (Al1D) or 30 days (Al30D) after the end of treatment. Prostates were dissected out and processed for structural, ultrastructural and immunohistochemical analyses. Male ventral prostates and female prostates of the Al1D group showed increased cell proliferation, glandular hyperplasia, increased secretory activity and greater androgen receptor immunoreactivity. In males, Al withdrawal (Al30D) allowed a partial recovery of the prostate, as the glandular secretory activity, and frequency of androgen receptor positive cells were similar to the control group. In females, the recuperation interval (Al30D) was not enough to restore the prostatic morphology, since the gland remained hyperplastic, proliferative, and with greater androgen and estrogen receptor immunoreactivity. These data alert to the importance of avoiding Al exposure, since this metal can have a harmful and prolonged action on the prostate.
Collapse
Affiliation(s)
- Danilo da Silva Lima
- Department of Histology, Embryology and Cell Biology, Laboratory of Microscopy Applied to Reproduction, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Liana da Silva Gomes
- Department of Histology, Embryology and Cell Biology, Laboratory of Microscopy Applied to Reproduction, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Esther de Sousa Figueredo
- Department of Histology, Embryology and Cell Biology, Laboratory of Microscopy Applied to Reproduction, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Yasmin Inocêncio Fernandes E Silva
- Department of Histology, Embryology and Cell Biology, Laboratory of Microscopy Applied to Reproduction, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Edvaldo Mendes Silva
- Department of Histology, Embryology and Cell Biology, Laboratory of Microscopy Applied to Reproduction, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Thais de Souza Bovi
- Department of Histology, Embryology and Cell Biology, Laboratory of Microscopy Applied to Reproduction, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Sebastião Roberto Taboga
- Department of Biology, Laboratory of Microscopy and Microanalysis, State University of São Paulo - UNESP, São Paulo, Brazil
| | - Mara Rúbia Marques
- Department of Histology, Embryology and Cell Biology, Laboratory of Microscopy Applied to Reproduction, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Manoel Francisco Biancardi
- Department of Histology, Embryology and Cell Biology, Laboratory of Microscopy Applied to Reproduction, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Fernanda Cristina Alcantara Dos Santos
- Department of Histology, Embryology and Cell Biology, Laboratory of Microscopy Applied to Reproduction, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| |
Collapse
|
165
|
Crowley L, Shen MM. Heterogeneity and complexity of the prostate epithelium: New findings from single-cell RNA sequencing studies. Cancer Lett 2022; 525:108-114. [PMID: 34728312 PMCID: PMC8629925 DOI: 10.1016/j.canlet.2021.10.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/07/2021] [Accepted: 10/25/2021] [Indexed: 01/30/2023]
Abstract
The recent advent of single-cell RNA-sequencing technology has provided new fundamental insights into the heterogeneity of the prostate epithelium. Several independent studies have described extensive heterogeneity of the luminal epithelial compartment, including a major division between a novel population of luminal cells located in the proximal region of the prostate ducts versus luminal cells located more distally. Proximal luminal cells as well as novel periurethral cells display increased progenitor potential in organoid culture and tissue reconstitution assays, but not in lineage-tracing analyses during prostate homeostasis, suggesting context-dependent plasticity of these populations. Here we describe and synthesize recent findings regarding the epithelial cell populations in the mouse prostate, draw comparisons to the human prostate, and address the relevance of these findings to prostate diseases and cancer.
Collapse
Affiliation(s)
| | - Michael M. Shen
- Author for correspondence at: phone: (212) 851-4723; fax: (212) 851-4572;
| |
Collapse
|
166
|
Prostate luminal progenitor cells: from mouse to human, from health to disease. Nat Rev Urol 2022; 19:201-218. [DOI: 10.1038/s41585-021-00561-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 12/11/2022]
|
167
|
Qian C, Li D, Chen Y. ETS factors in prostate cancer. Cancer Lett 2022; 530:181-189. [PMID: 35033589 PMCID: PMC8832285 DOI: 10.1016/j.canlet.2022.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/01/2022] [Accepted: 01/10/2022] [Indexed: 12/21/2022]
Abstract
The ETS family of proteins consists of 28 transcription factors, many of which play critical roles in both normal tissue development and homeostasis and have been implicated in development and progression of a variety of cancers. In prostate cancer, gene fusion and overexpression of ETS factors ERG, FLI1, ETV1, ETV4 and ETV5 have been found in half of prostate cancer patients in Caucasian men and define the largest genetic subtype of prostate cancer. This review summarizes the data on the discovery, modeling, molecular taxonomy, lineage plasticity and therapeutic targeting of ETS family members in prostate cancer.
Collapse
Affiliation(s)
- Cheng Qian
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Urology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Dan Li
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, 10065, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
168
|
Song H, Weinstein HNW, Allegakoen P, Wadsworth MH, Xie J, Yang H, Castro EA, Lu KL, Stohr BA, Feng FY, Carroll PR, Wang B, Cooperberg MR, Shalek AK, Huang FW. Single-cell analysis of human primary prostate cancer reveals the heterogeneity of tumor-associated epithelial cell states. Nat Commun 2022; 13:141. [PMID: 35013146 PMCID: PMC8748675 DOI: 10.1038/s41467-021-27322-4] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 10/29/2021] [Indexed: 12/31/2022] Open
Abstract
Prostate cancer is the second most common malignancy in men worldwide and consists of a mixture of tumor and non-tumor cell types. To characterize the prostate cancer tumor microenvironment, we perform single-cell RNA-sequencing on prostate biopsies, prostatectomy specimens, and patient-derived organoids from localized prostate cancer patients. We uncover heterogeneous cellular states in prostate epithelial cells marked by high androgen signaling states that are enriched in prostate cancer and identify a population of tumor-associated club cells that may be associated with prostate carcinogenesis. ERG-negative tumor cells, compared to ERG-positive cells, demonstrate shared heterogeneity with surrounding luminal epithelial cells and appear to give rise to common tumor microenvironment responses. Finally, we show that prostate epithelial organoids harbor tumor-associated epithelial cell states and are enriched with distinct cell types and states from their parent tissues. Our results provide diagnostically relevant insights and advance our understanding of the cellular states associated with prostate carcinogenesis.
Collapse
Affiliation(s)
- Hanbing Song
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Hannah N. W. Weinstein
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Paul Allegakoen
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Marc H. Wadsworth
- grid.116068.80000 0001 2341 2786The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.66859.340000 0004 0546 1623Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142 USA
| | - Jamie Xie
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Heiko Yang
- grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Department of Urology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Ethan A. Castro
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Kevin L. Lu
- grid.266102.10000 0001 2297 6811Department of Pathology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Bradley A. Stohr
- grid.266102.10000 0001 2297 6811Department of Pathology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Felix Y. Feng
- grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Department of Urology, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Departments of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Peter R. Carroll
- grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Department of Urology, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Bruce Wang
- grid.266102.10000 0001 2297 6811Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA 94143 USA
| | - Matthew R. Cooperberg
- grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Department of Urology, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.410372.30000 0004 0419 2775Division of Hematology and Oncology, Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121 USA
| | - Alex K. Shalek
- grid.116068.80000 0001 2341 2786The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Institute for Medical Engineering and Science (IMES), Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.66859.340000 0004 0546 1623Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142 USA
| | - Franklin W. Huang
- grid.266102.10000 0001 2297 6811Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.266102.10000 0001 2297 6811Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143 USA ,grid.410372.30000 0004 0419 2775Division of Hematology and Oncology, Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121 USA
| |
Collapse
|
169
|
Wang J, Feng X, Li Z, Chen Y, Huang W. Patient-derived organoids as a model for tumor research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:259-326. [PMID: 35595351 DOI: 10.1016/bs.pmbts.2022.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cancer represents a leading cause of death, despite the rapid progress of cancer research, leading to urgent need for accurate preclinical model to further study of tumor mechanism and accelerate translational applications. Cancer cell lines cannot fully recapitulate tumors of different patients due to the lack of tumor complexity and specification, while the high technical difficulty, long time, and substantial cost of patient-derived xenograft model makes it unable to be used extensively for all types of tumors and large-scale drug screening. Patient-derived organoids can be established rapidly with a high success rate from many tumors, and precisely replicate the key histopathological, genetic, and phenotypic features, as well as therapeutic response of patient tumor. Therefore, they are extensively used in cancer basic research, biobanking, disease modeling and precision medicine. The combinations of cancer organoids with other advanced technologies, such as 3D bio-printing, organ-on-a-chip, and CRISPR-Cas9, contributes to the more complete replication of complex tumor microenvironment and tumorigenesis. In this review, we discuss the various methods of the establishment and the application of patient-derived organoids in diverse tumors as well as the limitations and future prospects of these models. Further advances of tumor organoids are expected to bridge the huge gap between bench and bedside and provide the unprecedented opportunities to advance cancer research.
Collapse
Affiliation(s)
- Jia Wang
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Xiaoying Feng
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Zhichao Li
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China; International Cancer Center of Shenzhen University, Shenzhen, China
| | - Yongsong Chen
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Weiren Huang
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China; Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China; International Cancer Center of Shenzhen University, Shenzhen, China; Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
170
|
Liang J, Li X, Dong Y, Zhao B. Modeling Human Organ Development and Diseases With Fetal Tissue-Derived Organoids. Cell Transplant 2022; 31:9636897221124481. [PMID: 36121224 PMCID: PMC9490458 DOI: 10.1177/09636897221124481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Recent advances in human organoid technology have greatly facilitated the study of organ development and pathology. In most cases, these organoids are derived from either pluripotent stem cells or adult stem cells for the modeling of developmental events and tissue homeostasis. However, due to the lack of human fetal tissue references and research model, it is still challenging to capture early developmental changes and underlying mechanisms in human embryonic development. The establishment of fetal tissue–derived organoids in rigorous time points is necessary. Here we provide an overview of the strategies and applications of fetal tissue–derived organoids, mainly focusing on fetal organ development research, developmental defect disease modeling, and organ–organ interaction study. Discussion of the importance of fetal tissue research also highlights the prospects and challenges in this field.
Collapse
Affiliation(s)
- Jianqing Liang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinyang Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yateng Dong
- bioGenous Biotechnology, Inc., Hangzhou, China
| | - Bing Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
171
|
Flores-Téllez TDNJ, Baena E. Experimental challenges to modeling prostate cancer heterogeneity. Cancer Lett 2022; 524:194-205. [PMID: 34688843 DOI: 10.1016/j.canlet.2021.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/23/2021] [Accepted: 10/09/2021] [Indexed: 12/24/2022]
Abstract
Tumor heterogeneity plays a key role in prostate cancer prognosis, therapy selection, relapse, and acquisition of treatment resistance. Prostate cancer presents a heterogeneous diversity at inter- and intra-tumor and inter-patient levels which are influenced by multiple intrinsic and/or extrinsic factors. Recent studies have started to characterize the complexity of prostate tumors and these different tiers of heterogeneity. In this review, we discuss the most common factors that contribute to tumoral diversity. Moreover, we focus on the description of the in vitro and in vivo approaches, as well as high-throughput technologies, that help to model intra-tumoral diversity. Further understanding tumor heterogeneities and the challenges they present will guide enhanced patient risk stratification, aid the design of more precise therapies, and ultimately help beat this chameleon-like disease.
Collapse
Affiliation(s)
- Teresita Del N J Flores-Téllez
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield, SK10 4TG, UK
| | - Esther Baena
- Prostate Oncobiology Group, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Alderley Edge, Macclesfield, SK10 4TG, UK; Belfast-Manchester Movember Centre of Excellence, Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, SK10 4TG, UK.
| |
Collapse
|
172
|
Cheaito K, Bahmad HF, Hadadeh O, Msheik H, Monzer A, Ballout F, Dagher C, Telvizian T, Saheb N, Tawil A, El-Sabban M, El-Hajj A, Mukherji D, Al-Sayegh M, Abou-Kheir W. Establishment and characterization of prostate organoids from treatment-naïve patients with prostate cancer. Oncol Lett 2022; 23:6. [PMID: 34820005 PMCID: PMC8607232 DOI: 10.3892/ol.2021.13124] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
Three-dimensional (3D) organoid culture systems are emerging as potential reliable tools to investigate basic developmental processes of human disease, especially cancer. The present study used established and modified culture conditions to report successful generation and characterization of patient-derived organoids from fresh primary tissue specimens of patients with treatment-naïve prostate cancer (PCa). Fresh tissue specimens were collected, digested enzymatically and the resulting cell suspensions were plated in a 3D environment using Matrigel as an extracellular matrix. Previously established 12-factor medium for organoid culturing was modified to create a minimal 5-factor medium. Organoids and corresponding tissue specimens were characterized using transcriptomic analysis, immunofluorescent analysis, and immunohistochemistry. Furthermore, patient-derived organoids were used to assess the drug response. Treatment-naïve patient-derived PCa organoids were obtained from fresh radical prostatectomy specimens. These PCa organoids mimicked the heterogeneity of corresponding parental tumor tissue. Histopathological analysis demonstrated similar tissue architecture and cellular morphology, as well as consistent immunohistochemical marker expression. Also, the results confirmed the potential of organoids as an in vitro model to assess potential personalized treatment responses as there was a differential drug response between different patient samples. In conclusion, the present study investigated patient-derived organoids from a cohort of treatment-naïve patients. Derived organoids mimicked the histological features and prostate lineage profiles of their corresponding parental tissue and may present a potential model to predict patient-specific treatment response in a pre-clinical setting.
Collapse
Affiliation(s)
- Katia Cheaito
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Hisham F. Bahmad
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
- Arkadi M. Rywlin M.D. Department of Pathology and Laboratory Medicine, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| | - Ola Hadadeh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Hiba Msheik
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Alissar Monzer
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Farah Ballout
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Christelle Dagher
- Department of Internal Medicine, Division of Hematology/Oncology, Faculty of Medicine, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon
| | - Talar Telvizian
- Department of Internal Medicine, Division of Hematology/Oncology, Faculty of Medicine, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon
| | - Nour Saheb
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon
| | - Ayman Tawil
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Albert El-Hajj
- Department of Surgery, Division of Urology, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon
| | - Deborah Mukherji
- Department of Internal Medicine, Division of Hematology/Oncology, Faculty of Medicine, American University of Beirut Medical Center, Beirut 1107-2020, Lebanon
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107-2020, Lebanon
| |
Collapse
|
173
|
Huang Y, Liu X, Feng Y, Nie X, Liu Q, Du X, Wu Y, Liu T, Zhu X. Rotenone, an environmental toxin, causes abnormal methylation of the mouse brain organoid's genome and ferroptosis. Int J Med Sci 2022; 19:1184-1197. [PMID: 35919817 PMCID: PMC9339416 DOI: 10.7150/ijms.74569] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/24/2022] [Indexed: 11/25/2022] Open
Abstract
More and more reports have pointed out that rotenone, as an insecticide, has high neurotoxicity and reproductive toxicity to livestock and mammals. As a highly physiological correlation system of internal organs, quasi-organs have great potential in the fields of drug toxicity and efficacy test, toxicology research, developmental biology and so on. In this study, brain organs (mBOs) derived from mouse neural stem cells were used to investigate the effects of rotenone on the physiological activity and epigenetic modification of mBOs. At the same time, Rotenone could significantly stimulate the increase of the concentration of LPO, lactic acid and hydroxyl radical in mBOs, and inhibit the expression of neuronal marker Tuj1, CHAT, PAX6 and so on. Further analysis showed that Rotenonem could induce mitochondrial damage in mBOs. The results of qPCR and Western blot showed that Rotenone could up-regulate the expressions of ferroptosis promoting protein p53, Cox2 and so on, while inhibit the expressions of negative regulatory protein of ferroptosis GPX4, FTH1, SLC7A11. In addition, the results of RRBS-Seq sequencing showed that the methylation modification at DMR level in Rotenone-treated mBOs group was significantly higher than that in Ctrl group. The results of KEGG analysis showed that compared with Ctrl, the genes with hypermethylation of promoter and Genebody in Rotenone-treated mBOs were mainly located in the Neuro active ligand-receptor interaction signal transduction pathway. In summary, rotenone can significantly lead to abnormal methylation of mouse brain organs, and lead to the loss of normal physiological function of neurons by inducing ferroptosis.
Collapse
Affiliation(s)
- Yongyi Huang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Xin Liu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Ya Feng
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xiaoli Nie
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, China
| | - Qiang Liu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Xiling Du
- School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Yuncheng Wu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, China
| | - Xiaoying Zhu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
174
|
Patient-derived functional organoids as a personalized approach for drug screening against hepatobiliary cancers. Adv Cancer Res 2022; 156:319-341. [DOI: 10.1016/bs.acr.2022.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
175
|
Li Y, Chan JWY, Lau RWH, Cheung WWY, Wong AM, Wong AM, Wong N, Ng CSH. Organoids in Lung Cancer Management. Front Surg 2021; 8:753801. [PMID: 34957199 PMCID: PMC8698743 DOI: 10.3389/fsurg.2021.753801] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is a complex milieu of genomically altered cancer cells, a diverse collection of differentiated cells and nonneoplastic stroma. Lung cancer organoids is a three-dimensional structure grown from patient cancer tissue that could mimic in vivo complex behavior and cellular architecture of the cancer. Furthermore, the genomic alterations of the primary lung tumor is captured ex vivo. Lung cancer organoids have become an important preclinical model for oncology studies in recent years. It could be used to model the development of lung cancer, investigate the process of tumorigenesis, and also study the signaling pathways. The organoids could also be a platform to perform drug screening and biomarker validation of lung cancer, providing a promising prediction of patient-specific drug response. In this review, we described how lung cancer organoids have opened new avenues for translating basic cancer research into clinical therapy and discussed the latest and future developments in organoid technology, which could be further applied in lung cancer organoids research.
Collapse
Affiliation(s)
- Yushi Li
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Joyce W Y Chan
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Rainbow W H Lau
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Winnie W Y Cheung
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Alissa Michelle Wong
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Aikha M Wong
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Nathalie Wong
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Calvin Sze Hang Ng
- Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
176
|
Piro G, Agostini A, Larghi A, Quero G, Carbone C, Esposito A, Rizzatti G, Attili F, Alfieri S, Costamagna G, Tortora G. Pancreatic Cancer Patient-Derived Organoid Platforms: A Clinical Tool to Study Cell- and Non-Cell-Autonomous Mechanisms of Treatment Response. Front Med (Lausanne) 2021; 8:793144. [PMID: 35004765 PMCID: PMC8733292 DOI: 10.3389/fmed.2021.793144] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/03/2021] [Indexed: 12/13/2022] Open
Abstract
For many years, cell lines and animal models have been essential to improve our understanding of the basis of cell metabolism, signaling, and genetics. They also provided an essential boost to cancer drug discovery. Nevertheless, these model systems failed to reproduce the tumor heterogeneity and the complex biological interactions between cancer cells and human hosts, making a high priority search for alternative methods that are able to export results from model systems to humans, which has become a major bottleneck in the drug development. The emergent human in vitro 3D cell culture technologies have attracted widespread attention because they seem to have the potential to overcome these limitations. Organoids are unique 3D culture models with the ability to self-organize in contained structures. Their versatility has offered an exceptional window of opportunity to approach human cancers. Pancreatic cancers (PCs) patient-derived-organoids (PDOs) preserve histological, genomic, and molecular features of neoplasms they originate from and therefore retain their heterogeneity. Patient-derived organoids can be established with a high success rate from minimal tissue core specimens acquired with endoscopic-ultrasound-guided techniques and assembled into platforms, representing tens to hundreds of cancers each conserving specific features, expanding the types of patient samples that can be propagated and analyzed in the laboratory. Because of their nature, PDO platforms are multipurpose systems that can be easily adapted in co-culture settings to perform a wide spectrum of studies, ranging from drug discovery to immune response evaluation to tumor-stroma interaction. This possibility to increase the complexity of organoids creating a hybrid culture with non-epithelial cells increases the interest in organoid-based platforms giving a pragmatic way to deeply study biological interactions in vitro. In this view, implementing organoid models in co-clinical trials to compare drug responses may represent the next step toward even more personalized medicine. In the present review, we discuss how PDO platforms are shaping modern-day oncology aiding to unravel the most complex aspects of PC.
Collapse
Affiliation(s)
- Geny Piro
- Department of Medical and Surgical Sciences, Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Antonio Agostini
- Department of Medical and Surgical Sciences, Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Alberto Larghi
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- CERTT, Center for Endoscopic Research Therapeutics and Training, Catholic University, Rome, Italy
| | - Giuseppe Quero
- Department of Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Carmine Carbone
- Department of Medical and Surgical Sciences, Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Annachiara Esposito
- Department of Medical and Surgical Sciences, Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Gianenrico Rizzatti
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- CERTT, Center for Endoscopic Research Therapeutics and Training, Catholic University, Rome, Italy
| | - Fabia Attili
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- CERTT, Center for Endoscopic Research Therapeutics and Training, Catholic University, Rome, Italy
| | - Sergio Alfieri
- Department of Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Guido Costamagna
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- CERTT, Center for Endoscopic Research Therapeutics and Training, Catholic University, Rome, Italy
| | - Giampaolo Tortora
- Department of Medical and Surgical Sciences, Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Department of Translational Medicine, Medical Oncology, Catholic University of the Sacred Heart, Rome, Italy
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- *Correspondence: Giampaolo Tortora
| |
Collapse
|
177
|
Interruption of Klf5 acetylation in basal progenitor cells promotes luminal commitment by activating Notch signaling. J Genet Genomics 2021; 49:579-582. [PMID: 34952235 DOI: 10.1016/j.jgg.2021.11.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 11/24/2022]
|
178
|
Fu X, He Q, Tao Y, Wang M, Wang W, Wang Y, Yu QC, Zhang F, Zhang X, Chen YG, Gao D, Hu P, Hui L, Wang X, Zeng YA. Recent advances in tissue stem cells. SCIENCE CHINA. LIFE SCIENCES 2021; 64:1998-2029. [PMID: 34865207 DOI: 10.1007/s11427-021-2007-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022]
Abstract
Stem cells are undifferentiated cells capable of self-renewal and differentiation, giving rise to specialized functional cells. Stem cells are of pivotal importance for organ and tissue development, homeostasis, and injury and disease repair. Tissue-specific stem cells are a rare population residing in specific tissues and present powerful potential for regeneration when required. They are usually named based on the resident tissue, such as hematopoietic stem cells and germline stem cells. This review discusses the recent advances in stem cells of various tissues, including neural stem cells, muscle stem cells, liver progenitors, pancreatic islet stem/progenitor cells, intestinal stem cells, and prostate stem cells, and the future perspectives for tissue stem cell research.
Collapse
Affiliation(s)
- Xin Fu
- Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200233, China
| | - Qiang He
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Tao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mengdi Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Bioland Laboratory (Guangzhou), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Bioland Laboratory (Guangzhou), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yalong Wang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qing Cissy Yu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Fang Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiaoyu Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Max-Planck Center for Tissue Stem Cell Research and Regenerative Medicine, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510530, China.
| | - Dong Gao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Ping Hu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200233, China.
- Max-Planck Center for Tissue Stem Cell Research and Regenerative Medicine, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, 510530, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Bio-Research Innovation Center, Shanghai Institute of Biochemistry and Cell Biology, Suzhou, 215121, China.
| | - Lijian Hui
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Bio-Research Innovation Center, Shanghai Institute of Biochemistry and Cell Biology, Suzhou, 215121, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Xiaoqun Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Bioland Laboratory (Guangzhou), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Advanced Innovation Center for Human Brain Protection, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| | - Yi Arial Zeng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Bio-Research Innovation Center, Shanghai Institute of Biochemistry and Cell Biology, Suzhou, 215121, China.
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, 310024, China.
| |
Collapse
|
179
|
Mukhopadhyay C, Yang C, Xu L, Liu D, Wang Y, Huang D, Deonarine LD, Cyrta J, Davicioni E, Sboner A, Robinson BD, Chinnaiyan AM, Rubin MA, Barbieri CE, Zhou P. G3BP1 inhibits Cul3 SPOP to amplify AR signaling and promote prostate cancer. Nat Commun 2021; 12:6662. [PMID: 34795264 PMCID: PMC8602290 DOI: 10.1038/s41467-021-27024-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 10/27/2021] [Indexed: 01/03/2023] Open
Abstract
SPOP, an E3 ubiquitin ligase, acts as a prostate-specific tumor suppressor with several key substrates mediating oncogenic function. However, the mechanisms underlying SPOP regulation are largely unknown. Here, we have identified G3BP1 as an interactor of SPOP and functions as a competitive inhibitor of Cul3SPOP, suggesting a distinctive mode of Cul3SPOP inactivation in prostate cancer (PCa). Transcriptomic analysis and functional studies reveal a G3BP1-SPOP ubiquitin signaling axis that promotes PCa progression through activating AR signaling. Moreover, AR directly upregulates G3BP1 transcription to further amplify G3BP1-SPOP signaling in a feed-forward manner. Our study supports a fundamental role of G3BP1 in disabling the tumor suppressive Cul3SPOP, thus defining a PCa cohort independent of SPOP mutation. Therefore, there are significantly more PCa that are defective for SPOP ubiquitin ligase than previously appreciated, and these G3BP1high PCa are more susceptible to AR-targeted therapy.
Collapse
Affiliation(s)
- Chandrani Mukhopadhyay
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Chenyi Yang
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Limei Xu
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Deli Liu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Department of Urology, Weill Cornell Medicine, New York, NY, 10065, USA
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Yu Wang
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | - Dennis Huang
- Department of Urology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Lesa Dayal Deonarine
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Joanna Cyrta
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
| | | | - Andrea Sboner
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medical College, New York, NY, 10065, USA
- Englander Institute for Precision Medicine of Weill Cornell Medicine and New York-Presbyterian Hospital, New York, NY, 10065, USA
| | - Brian D Robinson
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
- Englander Institute for Precision Medicine of Weill Cornell Medicine and New York-Presbyterian Hospital, New York, NY, 10065, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Mark A Rubin
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA
- Department of Urology, Weill Cornell Medicine, New York, NY, 10065, USA
- Englander Institute for Precision Medicine of Weill Cornell Medicine and New York-Presbyterian Hospital, New York, NY, 10065, USA
- Department for Biomedical Research, University of Bern, 3008, Bern, Switzerland
| | - Christopher E Barbieri
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Department of Urology, Weill Cornell Medicine, New York, NY, 10065, USA
- Englander Institute for Precision Medicine of Weill Cornell Medicine and New York-Presbyterian Hospital, New York, NY, 10065, USA
| | - Pengbo Zhou
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, 10065, USA.
| |
Collapse
|
180
|
Hu LF, Yang X, Lan HR, Fang XL, Chen XY, Jin KT. Preclinical tumor organoid models in personalized cancer therapy: Not everyone fits the mold. Exp Cell Res 2021; 408:112858. [PMID: 34600901 DOI: 10.1016/j.yexcr.2021.112858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/10/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022]
Abstract
In contrast to conventional cancer treatment, in personalized cancer medicine each patient receives a specific treatment. The response to therapy, clinical outcomes, and tumor behavior such as metastases, tumor progression, carcinogenesis can be significantly affected by the heterogeneous tumor microenvironment (TME) and interpersonal differences. Therefore, using native tumor microenvironment mimicking models is necessary to improving personalized cancer therapy. Both in vitro 2D cell culture and in vivo animal models poorly recapitulate the heterogeneous tumor (immune) microenvironments of native tumors. The development of 3D culture models, native tumor microenvironment mimicking models, made it possible to evaluate the chemoresistance of tumor tissue and the functionality of drugs in the presence of cell-extracellular matrix and cell-cell interactions in a 3D construction. Various personalized tumor models have been designed to preserving the native tumor microenvironment, including patient-derived tumor xenografts and organoid culture strategies. In this review, we will discuss the patient-derived organoids as a native tumor microenvironment mimicking model in personalized cancer therapy. In addition, we will also review the potential and the limitations of organoid culture systems for predicting patient outcomes and preclinical drug screening. Finally, we will discuss immunotherapy drug screening in tumor organoids by using microfluidic technology.
Collapse
Affiliation(s)
- Li-Feng Hu
- Department of Colorectal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, Zhejiang Province, 312000, China
| | - Xue Yang
- Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang Province, 310014, China; Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang Province, 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang Province, 310014, China
| | - Huan-Rong Lan
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province, 321000, China
| | - Xing-Liang Fang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Shaoxing University College of Medicine (Shaoxing Municipal Hospital), Shaoxing, Zhejiang Province, 312000, China
| | - Xiao-Yi Chen
- Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang Province, 310014, China; Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang Province, 310014, China; Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang Province, 310014, China.
| | - Ke-Tao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province, 321000, China.
| |
Collapse
|
181
|
Thompson RE, Meyers MA, Pukazhenthi BS, Hollinshead FK. Evaluation of growth, viability, and structural integrity of equine endometrial organoids following cryopreservation. Cryobiology 2021; 104:56-62. [PMID: 34788682 DOI: 10.1016/j.cryobiol.2021.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/03/2021] [Accepted: 11/11/2021] [Indexed: 12/18/2022]
Abstract
Reproductive diseases in mares are a significant cause of subfertility and profound economic loss in the equine industry. Utilizing a 3D in vitro cell culture system that recapitulates the in vivo physiology will reduce time, cost, and welfare concerns associated with in vivo reproductive research in mares. If this 3D model is combined with effective cryopreservation, reproductive research on mares can occur year-round, which is not currently possible in this seasonal species. Endometrial organoids, 3D in vitro cell clusters that exhibit in vivo uterine physiology, have been established in mice, women, and mares. Here we report the first comprehensive assessment of cryopreservation of endometrial organoids in the domestic mare. Organoid growth rate was not affected by the type of freezing media. However, growth rate varied among non-cryopreserved controls, organoids cryopreserved at passage 0 (P0), and organoids cryopreserved at passage 3 (P3). Additionally, there was no difference in organoid viability among freezing media or freezing timepoint (passages). Furthermore, fresh and frozen-thawed organoids displayed positive immunohistochemical staining for ZO-1, which is a marker for intercellular tight junctions, and for periodic acid-Schiff staining as marker for organoid function through mucin production. Results demonstrate that equine endometrial organoids can be cryopreserved with 10% dimethyl sulfoxide with minimal detrimental effects while maintaining intercellular tight junctions (ZO-1) and secretory function. Availability of cryopreserved endometrial organoids may permit expanded research on uterine pathologies that negatively affect mare fertility and improve efficiency, reduce cost, and minimize animal welfare concerns associated with in vivo research in the domestic mare.
Collapse
Affiliation(s)
- Riley E Thompson
- Colorado State University, Department of Clinical Sciences, 1601 Campus Delivery, Fort Collins, CO, 80523, USA.
| | - Melinda A Meyers
- Colorado State University, Department of Clinical Sciences, 1601 Campus Delivery, Fort Collins, CO, 80523, USA
| | - Budhan S Pukazhenthi
- Smithsonian Conservation Biology Institute, Center for Species Survival, 1500 Remount Road, Front Royal, VA, 22630, USA
| | - Fiona K Hollinshead
- Colorado State University, Department of Clinical Sciences, 1601 Campus Delivery, Fort Collins, CO, 80523, USA
| |
Collapse
|
182
|
Ermini L, Francis JC, Rosa GS, Rose AJ, Ning J, Greaves M, Swain A. Evolutionary selection of alleles in the melanophilin gene that impacts on prostate organ function and cancer risk. EVOLUTION MEDICINE AND PUBLIC HEALTH 2021; 9:311-321. [PMID: 34754452 PMCID: PMC8573191 DOI: 10.1093/emph/eoab026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 09/03/2021] [Indexed: 11/21/2022]
Abstract
Background and objectives Several hundred inherited genetic variants or SNPs that alter the risk of cancer have been identified through genome-wide association studies. In populations of European ancestry, these variants are mostly present at relatively high frequencies. To gain insight into evolutionary origins, we screened a series of genes and SNPs linked to breast or prostate cancer for signatures of historical positive selection. Methodology We took advantage of the availability of the 1000 genome data and we performed genomic scans for positive selection in five different Caucasian populations as well as one African reference population. We then used prostate organoid cultures to provide a possible functional explanation for the interplay between the action of evolutionary forces and the disease risk association. Results Variants in only one gene showed genomic signatures of positive, evolutionary selection within Caucasian populations melanophilin (MLPH). Functional depletion of MLPH in prostate organoids, by CRISPR/Cas9 mutation, impacted lineage commitment of progenitor cells promoting luminal versus basal cell differentiation and on resistance to androgen deprivation. Conclusions and implications The MLPH variants influencing prostate cancer risk may have been historically selected for their adaptive benefit on skin pigmentation but MLPH is highly expressed in the prostate and the derivative, positively selected, alleles decrease the risk of prostate cancer. Our study suggests a potential functional mechanism via which MLPH and its genetic variants could influence risk of prostate cancer, as a serendipitous consequence of prior evolutionary benefits to another tissue. Lay Summary We screened a limited series of genomic variants associated with breast and prostate cancer risk for signatures of historical positive selection. Variants within the melanophilin (MLPH) gene fell into this category. Depletion of MLPH in prostate organoid cultures, suggested a potential functional mechanism for impacting on cancer risk, as a serendipitous consequence of prior evolutionary benefits to another tissue.
Collapse
Affiliation(s)
- Luca Ermini
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Jeffrey C Francis
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Gabriel S Rosa
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Alexandra J Rose
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Jian Ning
- Division of Cancer Biology, The Institute of Cancer Research, London, UK.,Tumour Profiling Unit, The Institute of Cancer Research, London, UK
| | - Mel Greaves
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Amanda Swain
- Division of Cancer Biology, The Institute of Cancer Research, London, UK.,Tumour Profiling Unit, The Institute of Cancer Research, London, UK
| |
Collapse
|
183
|
Yi SA, Zhang Y, Rathnam C, Pongkulapa T, Lee KB. Bioengineering Approaches for the Advanced Organoid Research. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007949. [PMID: 34561899 PMCID: PMC8682947 DOI: 10.1002/adma.202007949] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 06/09/2021] [Indexed: 05/09/2023]
Abstract
Recent advances in 3D cell culture technology have enabled scientists to generate stem cell derived organoids that recapitulate the structural and functional characteristics of native organs. Current organoid technologies have been striding toward identifying the essential factors for controlling the processes involved in organoid development, including physical cues and biochemical signaling. There is a growing demand for engineering dynamic niches characterized by conditions that resemble in vivo organogenesis to generate reproducible and reliable organoids for various applications. Innovative biomaterial-based and advanced engineering-based approaches have been incorporated into conventional organoid culture methods to facilitate the development of organoid research. The recent advances in organoid engineering, including extracellular matrices and genetic modulation, are comprehensively summarized to pinpoint the parameters critical for organ-specific patterning. Moreover, perspective trends in developing tunable organoids in response to exogenous and endogenous cues are discussed for next-generation developmental studies, disease modeling, and therapeutics.
Collapse
Affiliation(s)
- Sang Ah Yi
- Epigenome Dynamics Control Research Center, School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, 16419, Republic of Korea
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Yixiao Zhang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Christopher Rathnam
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Thanapat Pongkulapa
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Road, Piscataway, NJ, 08854, USA
| |
Collapse
|
184
|
Abstract
Organoids have complex three-dimensional structures that exhibit functionalities and feature architectures similar to those of in vivo organs and are developed from adult stem cells, embryonic stem cells, and pluripotent stem cells through a self-organization process. Organoids derived from adult epithelial stem cells are the most mature and extensive. In recent years, using organoid culture techniques, researchers have established various adult human tissue-derived epithelial organoids, including intestinal, colon, lung, liver, stomach, breast, and oral mucosal organoids, all of which exhibit strong research and application prospects. Studies have shown that epithelial organoids are mainly applied in drug discovery, personalized drug response testing, disease mechanism research, and regenerative medicine. In this review, we mainly discuss current organoid culture systems and potential applications of this technique with human epithelial tissue.
Collapse
Affiliation(s)
- Fengjiao Li
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Peng Zhang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China.,Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Hunan Normal University), Ministry of Education, College of Chemistry & Chemical Engineering, Changsha, Hunan 410081, China
| | - Saizhi Wu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Lianwen Yuan
- Department of Geriatric Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| |
Collapse
|
185
|
Ter Steege EJ, Bakker ERM. The role of R-spondin proteins in cancer biology. Oncogene 2021; 40:6469-6478. [PMID: 34663878 PMCID: PMC8616751 DOI: 10.1038/s41388-021-02059-y] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/23/2021] [Accepted: 10/01/2021] [Indexed: 02/07/2023]
Abstract
R-spondin (RSPO) proteins constitute a family of four secreted glycoproteins (RSPO1-4) that have appeared as multipotent signaling ligands. The best-known molecular function of RSPOs lie within their capacity to agonize the Wnt/β-catenin signaling pathway. As RSPOs act upon cognate receptors LGR4/5/6 that are typically expressed by stem cells and progenitor cells, RSPO proteins importantly potentiate Wnt/β-catenin signaling especially within these proliferative stem cell compartments. Since multiple organs express LGR4/5/6 receptors and RSPO ligands within their stem cell niches, RSPOs can exert an influential role in stem cell regulation throughout the body. Inherently, over the last decade a multitude of reports implicated the deregulation of RSPOs in cancer development. First, RSPO2 and RSPO3 gene fusions with concomitant enhanced expression have been identified in colon cancer patients, and proposed as an alternative driver of Wnt/β-catenin hyperactivation that earmarks cancer in the colorectal tract. Moreover, the causal oncogenic capacity of RSPO3 overactivation has been demonstrated in the mouse intestine. As a paradigm organ in this field, most of current knowledge about RSPOs in cancer is derived from studies in the intestinal tract. However, RSPO gene fusions as well as enhanced RSPO expression have been reported in multiple additional cancer types, affecting different organs that involve divergent stem cell hierarchies. Importantly, the emerging oncogenic role of RSPO and its potential clinical utility as a therapeutic target have been recognized and investigated in preclinical and clinical settings. This review provides a survey of current knowledge on the role of RSPOs in cancer biology, addressing the different organs implicated, and of efforts made to explore intervention opportunities in cancer cases with RSPO overrepresentation, including the potential utilization of RSPO as novel therapeutic target itself.
Collapse
Affiliation(s)
- Eline J Ter Steege
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Elvira R M Bakker
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
186
|
Ebrahimi N, Nasr Esfahani A, Samizade S, Mansouri A, Ghanaatian M, Adelian S, Shadman Manesh V, Hamblin MR. The potential application of organoids in breast cancer research and treatment. Hum Genet 2021; 141:193-208. [PMID: 34713317 DOI: 10.1007/s00439-021-02390-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 10/16/2021] [Indexed: 12/23/2022]
Abstract
Tumor heterogeneity is a major challenge for breast cancer researchers who have struggled to find effective treatments despite recent advances in oncology. Although the use of 2D cell culture methods in breast cancer research has been effective, it cannot model the heterogeneity of breast cancer as found within the body. The development of 3D culture of tumor cells and breast cancer organoids has provided a new approach in breast cancer research, allowing the identification of biomarkers, study of the interaction of tumor cells with the microenvironment, and for drug screening and discovery. In addition, the possibility of gene editing in organoids, especially using the CRISPR/Cas9 system, is convenient, and has allowed a more detailed study of tumor behavior in models closer to the physiological condition. The present review covers the application of organoids in breast cancer research. The recent use of gene-editing systems to provide insights into therapeutic approaches for breast cancer, is highlighted. The study of organoids and the possibility of gene manipulation may be a step towards the personalized treatment of breast cancer, which has so far remained unattainable due to the high heterogeneity of breast cancer.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Division of Genetics, Department of Cell, Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Islamic Republic of Iran
| | - Alireza Nasr Esfahani
- Department of Cellular and Molecular Biology, School of Biological Sciences, Islamic Azad University of Falavarjan, Falavarjan, Iran
| | - Setare Samizade
- Department of Cellular and Molecular Biology, School of Biological Sciences, Islamic Azad University of Falavarjan, Falavarjan, Iran
| | - Atena Mansouri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Masoud Ghanaatian
- Department of Microbiology, Islamic Azad University of Jahrom, Jahrom, Fars, Iran
| | - Samaneh Adelian
- Department of Genetics, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Vida Shadman Manesh
- Medical Engineering Tissue Engineering, Department of Medical Technologies, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Michael R Hamblin
- Faculty of Health Science, Laser Research Centre, University of Johannesburg, Doornfontein, Johannesburg, 2028, South Africa.
| |
Collapse
|
187
|
Indana D, Agarwal P, Bhutani N, Chaudhuri O. Viscoelasticity and Adhesion Signaling in Biomaterials Control Human Pluripotent Stem Cell Morphogenesis in 3D Culture. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2101966. [PMID: 34499389 DOI: 10.1002/adma.202101966] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/27/2021] [Indexed: 06/13/2023]
Abstract
Organoids are lumen-containing multicellular structures that recapitulate key features of the organs, and are increasingly used in models of disease, drug testing, and regenerative medicine. Recent work has used 3D culture models to form organoids from human induced pluripotent stem cells (hiPSCs) in reconstituted basement membrane (rBM) matrices. However, rBM matrices offer little control over the microenvironment. More generally, the role of matrix viscoelasticity in directing lumen formation remains unknown. Here, viscoelastic alginate hydrogels with independently tunable stress relaxation (viscoelasticity), stiffness, and arginine-glycine-aspartate (RGD) ligand density are used to study hiPSC morphogenesis in 3D culture. A phase diagram that shows how these properties control hiPSC morphogenesis is reported. Higher RGD density and fast stress relaxation promote hiPSC viability, proliferation, apicobasal polarization, and lumen formation, while slow stress relaxation at low RGD densities triggers hiPSC apoptosis. Notably, hiPSCs maintain pluripotency in alginate hydrogels for much longer times than is reported in rBM matrices. Lumen formation is regulated by actomyosin contractility and is accompanied by translocation of Yes-associated protein (YAP) from the nucleus to the cytoplasm. The results reveal matrix viscoelasticity as a potent factor regulating stem cell morphogenesis and provide new insights into how engineered biomaterials may be leveraged to build organoids.
Collapse
Affiliation(s)
- Dhiraj Indana
- Department of Mechanical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Pranay Agarwal
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Nidhi Bhutani
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
188
|
Youk J, Kwon HW, Kim R, Ju YS. Dissecting single-cell genomes through the clonal organoid technique. Exp Mol Med 2021; 53:1503-1511. [PMID: 34663940 PMCID: PMC8569207 DOI: 10.1038/s12276-021-00680-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 07/13/2021] [Indexed: 12/11/2022] Open
Abstract
The revolution in genome sequencing technologies has enabled the comprehensive detection of genomic variations in human cells, including inherited germline polymorphisms, de novo mutations, and postzygotic mutations. When these technologies are combined with techniques for isolating and expanding single-cell DNA, the landscape of somatic mosaicism in an individual body can be systematically revealed at a single-cell resolution. Here, we summarize three strategies (whole-genome amplification, microdissection of clonal patches in the tissue, and in vitro clonal expansion of single cells) that are currently applied for single-cell mutational analyses. Among these approaches, in vitro clonal expansion, particularly via adult stem cell-derived organoid culture technologies, yields the most sensitive and precise catalog of somatic mutations in single cells. Moreover, because it produces living mutant cells, downstream validation experiments and multiomics profiling are possible. Through the synergistic combination of organoid culture and genome sequencing, researchers can track genome changes at a single-cell resolution, which will lead to new discoveries that were previously impossible.
Collapse
Affiliation(s)
- Jeonghwan Youk
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
- GENOME INSIGHT Inc, Daejeon, 34051, Republic of Korea
| | - Hyun Woo Kwon
- Department of Nuclear Medicine, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Ryul Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Young Seok Ju
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
- GENOME INSIGHT Inc, Daejeon, 34051, Republic of Korea.
| |
Collapse
|
189
|
Olson AW, Le V, Wang J, Hiroto A, Kim WK, Lee DH, Aldahl J, Wu X, Kim M, Cunha GR, You S, Sun Z. Stromal androgen and hedgehog signaling regulates stem cell niches in pubertal prostate development. Development 2021; 148:271928. [PMID: 34427305 DOI: 10.1242/dev.199738] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/17/2021] [Indexed: 12/13/2022]
Abstract
Stromal androgen-receptor (AR) action is essential for prostate development, morphogenesis and regeneration. However, mechanisms underlying how stromal AR maintains the cell niche in support of pubertal prostatic epithelial growth are unknown. Here, using advanced mouse genetic tools, we demonstrate that selective deletion of stromal AR expression in prepubescent Shh-responsive Gli1-expressing cells significantly impedes pubertal prostate epithelial growth and development. Single-cell transcriptomic analyses showed that AR loss in these prepubescent Gli1-expressing cells dysregulates androgen signaling-initiated stromal-epithelial paracrine interactions, leading to growth retardation of pubertal prostate epithelia and significant development defects. Specifically, AR loss elevates Shh-signaling activation in both prostatic stromal and adjacent epithelial cells, directly inhibiting prostatic epithelial growth. Single-cell trajectory analyses further identified aberrant differentiation fates of prostatic epithelial cells directly altered by stromal AR deletion. In vivo recombination of AR-deficient stromal Gli1-lineage cells with wild-type prostatic epithelial cells failed to develop normal prostatic epithelia. These data demonstrate previously unidentified mechanisms underlying how stromal AR-signaling facilitates Shh-mediated cell niches in pubertal prostatic epithelial growth and development.
Collapse
Affiliation(s)
- Adam W Olson
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010-3000, USA
| | - Vien Le
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010-3000, USA
| | - Jinhui Wang
- Integrative Genomics Core, City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA 91010-3000, USA
| | - Alex Hiroto
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010-3000, USA
| | - Won Kyung Kim
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010-3000, USA
| | - Dong-Hoon Lee
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010-3000, USA
| | - Joseph Aldahl
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010-3000, USA
| | - Xiwei Wu
- Integrative Genomics Core, City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA 91010-3000, USA
| | - Minhyung Kim
- Division of Cancer Biology and Therapeutics, Departments of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gerald R Cunha
- Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Sungyong You
- Division of Cancer Biology and Therapeutics, Departments of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Zijie Sun
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA 91010-3000, USA
| |
Collapse
|
190
|
Menche C, Farin HF. Strategies for genetic manipulation of adult stem cell-derived organoids. Exp Mol Med 2021; 53:1483-1494. [PMID: 34663937 PMCID: PMC8569115 DOI: 10.1038/s12276-021-00609-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/21/2021] [Accepted: 03/05/2021] [Indexed: 01/08/2023] Open
Abstract
Organoid technology allows the expansion of primary epithelial cells from normal and diseased tissues, providing a unique model for human (patho)biology. In a three-dimensional environment, adult stem cells self-organize and differentiate to gain tissue-specific features. Accessibility to genetic manipulation enables the investigation of the molecular mechanisms underlying cell fate regulation, cell differentiation and cell interactions. In recent years, powerful methodologies using lentiviral transgenesis, CRISPR/Cas9 gene editing, and single-cell readouts have been developed to study gene function and carry out genetic screens in organoids. However, the multicellularity and dynamic nature of stem cell-derived organoids also present challenges for genetic experimentation. In this review, we focus on adult gastrointestinal organoids and summarize the state-of-the-art protocols for successful transgenesis. We provide an outlook on emerging genetic techniques that could further increase the applicability of organoids and enhance the potential of organoid-based techniques to deepen our understanding of gene function in tissue biology.
Collapse
Affiliation(s)
- Constantin Menche
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Henner F Farin
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany.
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
191
|
Pamarthy S, Sabaawy HE. Patient derived organoids in prostate cancer: improving therapeutic efficacy in precision medicine. Mol Cancer 2021; 20:125. [PMID: 34587953 PMCID: PMC8480086 DOI: 10.1186/s12943-021-01426-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/11/2021] [Indexed: 12/30/2022] Open
Abstract
With advances in the discovery of the clinical and molecular landscapes of prostate cancer (PCa), implementation of precision medicine-guided therapeutic testing in the clinic has become a priority. Patient derived organoids (PDOs) are three-dimensional (3D) tissue cultures that promise to enable the validation of preclinical drug testing in precision medicine and coclinical trials by modeling PCa for predicting therapeutic responses with a reliable efficacy. We evaluate the advances in 3D culture and PDO use to model clonal heterogeneity and screen for effective targeted therapies, with a focus on the technological advances in generating PDOs. Recent innovations include the utilization of PDOs both in original research and/or correlative studies in clinical trials to examine drug effects within the PCa tumor microenvironment (TME). There has also been a significant improvement with the utilization of various extracellular matrices and single cell assays for the generation and long-term propagation of PDOs. Single cell derived PDOs could faithfully recapitulate the original tumor and reflect the heterogeneity features. While most PDO use for precision medicine understandably involved tissues derived from metastatic patients, we envision that the generation of PDOs from localized PCa along with the incorporation of cells of the TME in tissue models would fulfill the great potential of PDOs in predicting drug clinical benefits. We conclude that single cell derived PDOs reiterate the molecular features of the original tumor and represent a reliable pre-clinical PCa model to understand individual tumors and design tailored targeted therapies.
Collapse
Affiliation(s)
- Sahithi Pamarthy
- Rutgers Cancer Institute of New Jersey, Rutgers University, 195 Little Albany St, Rm 4557, New Brunswick, NJ, 08901, USA
| | - Hatem E Sabaawy
- Rutgers Cancer Institute of New Jersey, Rutgers University, 195 Little Albany St, Rm 4557, New Brunswick, NJ, 08901, USA.
- Clinical Investigations and Precision Therapeutics Program, Devision of Medical Oncology, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA.
- Departments of Pathology and Laboratory Medicine, RBHS-Robert Wood Johnson Medical School, New Brunswick, USA.
- Departments of Medicine, RBHS-Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
192
|
Saleh J, Mercier B, Xi W. Bioengineering methods for organoid systems. Biol Cell 2021; 113:475-491. [PMID: 34580889 DOI: 10.1111/boc.202000119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 08/05/2021] [Accepted: 09/16/2021] [Indexed: 12/23/2022]
Abstract
Organoids have been widely used in fundamental, biomimetic, and therapeutic studies. These multicellular systems form via cell-autonomous self-organization where a cohort of stem cells undergoes in vivo-like proliferation, differentiation, and morphogenesis. They also recapitulate a series of physiological cell organization, complexity and functions that are untouchable by conventional bio-model systems using immortal cell lines. However, the development of organoids is often not easily controlled and their shape and size are yet fully physiological. Recent research has demonstrated that multiple bioengineering tools could be harnessed to control important internal and external cues that dictate stem cell behavior and stem-cell based organoid development. In this review, we introduce the current development of organoid systems and their potentials, as well as their limitations that impede their further utility in research and clinical fields. In comparison to conventional autonomous organoid system, we then review bioengineering approaches that offer improved control over organoid growth and development. We focus on the genetic editing tools that allow the program of build-in responses and phenotypes for organoid systems with enhanced physiological relevance. We also highlight the advances in bioengineering methods to modify cellular external milieus to generate desirable cell composition, 3D micro-architectures, and complex microfluidic systems. We conclude that the emerging biomimetic methods that employ multidisciplinary approaches could prevail in the future development of organoid systems.
Collapse
Affiliation(s)
- Jad Saleh
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| | - Barbara Mercier
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| | - Wang Xi
- Université de Paris, CNRS, Institut Jacques Monod, Paris, France
| |
Collapse
|
193
|
Heninger E, Kosoff D, Rodems TS, Sethakorn N, Singh A, Gungurthi H, Carlson KN, Yang B, Gilsdorf C, Pasch CA, Deming DA, Ellis L, Beebe DJ, Jarrard DF, Lang JM. Live cell molecular analysis of primary prostate cancer organoids identifies persistent androgen receptor signaling. Med Oncol 2021; 38:135. [PMID: 34581895 PMCID: PMC8478748 DOI: 10.1007/s12032-021-01582-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 09/12/2021] [Indexed: 11/29/2022]
Abstract
Prostate Cancer (PC) is a disease with remarkable tumor heterogeneity that often manifests in significant intra-patient variability with regards to clinical outcomes and treatment response. Commonly available PC cell lines do not accurately reflect the complexity of this disease and there is critical need for development of new models to recapitulate the intricate hierarchy of tumor pathogenesis. In current study, we established ex vivo primary patient-derived cancer organoid (PDCO) cultures from prostatectomy specimens of patients with locally advanced PC. We then performed a comprehensive multi-parameter characterization of the cellular composition utilizing a novel approach for live-cell staining and direct imaging in the integrated microfluidic Stacks device. Using orthogonal flow cytometry analysis, we demonstrate that primary PDCOs maintain distinct subsets of epithelial cells throughout culture and that these cells conserve expression of androgen receptor (AR)-related elements. Furthermore, to confirm the tumor-origin of the PDCOs we have analyzed the expression of PC-associated epigenetic biomarkers including promoter methylation of the GSTP1, RASSF1 and APC and RARb genes by employing a novel microfluidic rare-event screening protocol. These results demonstrate that this ex vivo PDCO model recapitulates the complexity of the epithelial tumor microenvironment of multifocal PC using orthogonal analyses. Furthermore, we propose to leverage the Stacks microfluidic device as a high-throughput, translational platform to interrogate phenotypic and molecular endpoints with the capacity to incorporate a complex tumor microenvironment.
Collapse
Affiliation(s)
- Erika Heninger
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - David Kosoff
- Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Tamara S Rodems
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Nan Sethakorn
- Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Anupama Singh
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Harshitha Gungurthi
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Kristin N Carlson
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Bing Yang
- Department of Urology, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Cole Gilsdorf
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Cheri A Pasch
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA
| | - Dustin A Deming
- Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Leigh Ellis
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, 1111 Highland Ave., Madison, WI, 53705, USA
| | - David F Jarrard
- Department of Urology, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Joshua M Lang
- University of Wisconsin Carbone Cancer Center, 1111 Highland Ave., Madison, USA. .,Department of Medicine, University of Wisconsin, Madison, 1111 Highland Ave., Madison, WI, 53705, USA. .,Wisconsin Institutes for Medical Research, Rm 7151, 1111 Highland Ave., Madison, WI, 53705, USA.
| |
Collapse
|
194
|
Zhou L, Zhang C, Zhang Y, Shi C. Application of Organoid Models in Prostate Cancer Research. Front Oncol 2021; 11:736431. [PMID: 34646778 PMCID: PMC8504437 DOI: 10.3389/fonc.2021.736431] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/09/2021] [Indexed: 11/24/2022] Open
Abstract
Complex heterogeneity is an important characteristic in the development of prostate cancer (PCa), which further leads to the failure of known therapeutic options. PCa research has been hampered by the current in vitro model systems that cannot fully reflect the biological characteristics and clinical diversity of PCa. The tumor organoid model in three-dimensional culture retains the heterogeneity of primary tumor tissues in vitro well and enables high-throughput screening and genome editing. Therefore, the establishment of a PCa organoid model that recapitulates the diverse heterogeneity observed in clinical settings is of great significance for the study of PCa. In this review, we summarize the culture conditions, establishments, and limitations of PCa organoids and further review their application for the study of pathogenesis, drug screening, mechanism of drug resistance, and individualized treatment for PCa. Additionally, we look forward to other potential developmental directions of PCa organoids, such as the interaction between prostate cancer tumor cells and their microenvironment, clinical individualized treatments, heterogeneous transformation model, tumor immunotherapy, and organoid models combined with liquid biopsy. Through this, we provide more effective preclinical experimental schemes using the PCa organoid model.
Collapse
Affiliation(s)
- Ligui Zhou
- Animal Experiment Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi’an, China
| | - Caiqin Zhang
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi’an, China
| | - Yongbin Zhang
- Animal Experiment Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changhong Shi
- Division of Cancer Biology, Laboratory Animal Center, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
195
|
Veninga V, Voest EE. Tumor organoids: Opportunities and challenges to guide precision medicine. Cancer Cell 2021; 39:1190-1201. [PMID: 34416168 DOI: 10.1016/j.ccell.2021.07.020] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 01/20/2023]
Abstract
Tumor organoids have been proposed as a model system for precision medicine. The ability of tumor organoids to retain characteristics of the original tumor makes them unique for cancer research on an individual patient level. Hence, the idea to use tumor organoids for clinical decision making and optimize patient outcome is tempting. In vitro responses of tumor organoids to a wide array of drugs have been positively correlated to patient responses. However, substantial challenges remain and prospective studies with large cohorts are highly needed before implementation in clinical cancer care can be considered. Because of their personalized characteristics and the immediate link with patient data, tumor organoids also have great potential in preclinical research. Here, we provide a critical overview of both clinical and preclinical advances using tumor organoids.
Collapse
Affiliation(s)
- Vivien Veninga
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam 1066CX, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Emile E Voest
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam 1066CX, The Netherlands; Oncode Institute, Utrecht, The Netherlands.
| |
Collapse
|
196
|
Marx U, Accastelli E, David R, Erfurth H, Koenig L, Lauster R, Ramme AP, Reinke P, Volk HD, Winter A, Dehne EM. An Individual Patient's "Body" on Chips-How Organismoid Theory Can Translate Into Your Personal Precision Therapy Approach. Front Med (Lausanne) 2021; 8:728866. [PMID: 34589503 PMCID: PMC8473633 DOI: 10.3389/fmed.2021.728866] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/16/2021] [Indexed: 11/24/2022] Open
Abstract
The first concepts for reproducing human systemic organismal biology in vitro were developed over 12 years ago. Such concepts, then called human- or body-on-a-chip, claimed that microphysiological systems would become the relevant technology platform emulating the physiology and morphology of human organisms at the smallest biologically acceptable scale in vitro and, therefore, would enable the selection of personalized therapies for any patient at unprecedented precision. Meanwhile, the first human organoids-stem cell-derived complex three-dimensional organ models that expand and self-organize in vitro-have proven that in vitro self-assembly of minute premature human organ-like structures is feasible, once the respective stimuli of ontogenesis are provided to human stem cells. Such premature organoids can precisely reflect a number of distinct physiological and pathophysiological features of their respective counterparts in the human body. We now develop the human-on-a-chip concepts of the past into an organismoid theory. We describe the current concept and principles to create a series of organismoids-minute, mindless and emotion-free physiological in vitro equivalents of an individual's mature human body-by an artificially short process of morphogenetic self-assembly mimicking an individual's ontogenesis from egg cell to sexually mature organism. Subsequently, we provide the concept and principles to maintain such an individual's set of organismoids at a self-sustained functional healthy homeostasis over very long time frames in vitro. Principles how to perturb a subset of healthy organismoids by means of the natural or artificial induction of diseases are enrolled to emulate an individual's disease process. Finally, we discuss using such series of healthy and perturbed organismoids in predictively selecting, scheduling and dosing an individual patient's personalized therapy or medicine precisely. The potential impact of the organismoid theory on our healthcare system generally and the rapid adoption of disruptive personalized T-cell therapies particularly is highlighted.
Collapse
Affiliation(s)
- Uwe Marx
- Department of Medical Biotechnology, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
- TissUse GmbH, Berlin, Germany
| | | | - Rhiannon David
- Functional and Mechanistic Safety, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom
| | | | | | - Roland Lauster
- Department of Medical Biotechnology, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | | | - Petra Reinke
- Berlin Center for Advanced Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
- BIH-Center for Regenerative Therapies, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hans-Dieter Volk
- BIH-Center for Regenerative Therapies, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | |
Collapse
|
197
|
Bipotent Progenitors Do Not Require Androgen Receptor for Luminal Specification during Prostate Organogenesis. Stem Cell Reports 2021; 15:1026-1036. [PMID: 33176121 PMCID: PMC7664048 DOI: 10.1016/j.stemcr.2020.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 12/31/2022] Open
Abstract
Androgen receptor (AR) plays a fundamental role in most aspects of adult prostate homeostasis, and anti-androgen therapy represents the cornerstone of prostate cancer treatment. However, early prostate organogenesis takes place during pre-pubertal stages when androgen levels are low, raising the possibility that AR function is more limited during prostate development. Here, we use inducible AR deletion and lineage tracing in genetically engineered mice to show that basal and luminal epithelial progenitors do not require cell-autonomous AR activity during prostate development. We also demonstrate the existence of a transient bipotent luminal progenitor that can generate luminal and basal progeny, yet is also independent of AR function. Furthermore, molecular analyses of AR-deleted luminal cells isolated from developing prostates indicate their similarity to wild-type cells. Our findings suggest that low androgen levels correlate with luminal plasticity in prostate development and may have implications for understanding how AR inhibition promotes lineage plasticity in prostate cancer. Prostate epithelial cell types are specified when epithelial AR expression is low Bipotent basal progenitors can generate luminal cells without cell-autonomous AR Transient bipotent luminal progenitors can generate luminal and basal cells AR-deleted luminal cells during development are molecularly similar to wild-type
Collapse
|
198
|
Grbesa I, Augello MA, Liu D, McNally DR, Gaffney CD, Huang D, Lin K, Ivenitsky D, Goueli R, Robinson BD, Khani F, Deonarine LD, Blattner M, Elemento O, Davicioni E, Sboner A, Barbieri CE. Reshaping of the androgen-driven chromatin landscape in normal prostate cells by early cancer drivers and effect on therapeutic sensitivity. Cell Rep 2021; 36:109625. [PMID: 34496233 PMCID: PMC8477443 DOI: 10.1016/j.celrep.2021.109625] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/06/2021] [Accepted: 08/05/2021] [Indexed: 12/21/2022] Open
Abstract
The normal androgen receptor (AR) cistrome and transcriptional program are fundamentally altered in prostate cancer (PCa). Here, we profile the chromatin landscape and AR-directed transcriptional program in normal prostate cells and show the impact of SPOP mutations, an early event in prostate tumorigenesis. In genetically normal mouse prostate organoids, SPOP mutation results in accessibility and AR binding patterns similar to that of human PCa. Consistent with dependence on AR signaling, castration of SPOP mutant mouse models results in the loss of neoplastic phenotypes, and human SPOP mutant PCa shows a favorable response to AR-targeted therapies. Together, these data validate mouse prostate organoids as a robust model for studying epigenomic and transcriptional alterations in normal prostate, provide valuable datasets for further studies, and show that a single genomic alteration may be sufficient to reprogram the chromatin of normal prostate cells toward oncogenic phenotypes, with potential therapeutic implications for AR-targeting therapies.
Collapse
Affiliation(s)
- Ivana Grbesa
- Department of Urology, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Michael A Augello
- Department of Urology, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Deli Liu
- Department of Urology, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Dylan R McNally
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA; Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA; Department of Medicine and Weill Cornell Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | | | - Dennis Huang
- Department of Urology, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Kevin Lin
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Daria Ivenitsky
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ramy Goueli
- Department of Urology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Brian D Robinson
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA; Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Francesca Khani
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA; Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lesa D Deonarine
- Department of Urology, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mirjam Blattner
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Olivier Elemento
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA; Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA; Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Andrea Sboner
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA; Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA; Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Christopher E Barbieri
- Department of Urology, Weill Cornell Medicine, New York, NY 10065, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
199
|
He J, Zhang X, Xia X, Han M, Li F, Li C, Li Y, Gao D. Organoid technology for tissue engineering. J Mol Cell Biol 2021; 12:569-579. [PMID: 32249317 PMCID: PMC7683016 DOI: 10.1093/jmcb/mjaa012] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 01/11/2020] [Accepted: 02/04/2020] [Indexed: 12/18/2022] Open
Abstract
For centuries, attempts have been continuously made to artificially reconstitute counterparts of in vivo organs from their tissues or cells. Only in the recent decade has organoid technology as a whole technological field systematically emerged and been shown to play important roles in tissue engineering. Based on their self-organizing capacities, stem cells of versatile organs, both harvested and induced, can form 3D structures that are structurally and functionally similar to their in vivo counterparts. These organoid models provide a powerful platform for elucidating the development mechanisms, modeling diseases, and screening drug candidates. In this review, we will summarize the advances of this technology for generating various organoids of tissues from the three germ layers and discuss their drawbacks and prospects for tissue engineering.
Collapse
Affiliation(s)
- Juan He
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaoyu Zhang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinyi Xia
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Ming Han
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Fei Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Chunfeng Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yunguang Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Dong Gao
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
200
|
Zhang MM, Yang KL, Cui YC, Zhou YS, Zhang HR, Wang Q, Ye YJ, Wang S, Jiang KW. Current Trends and Research Topics Regarding Intestinal Organoids: An Overview Based on Bibliometrics. Front Cell Dev Biol 2021; 9:609452. [PMID: 34414174 PMCID: PMC8369504 DOI: 10.3389/fcell.2021.609452] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 06/08/2021] [Indexed: 01/10/2023] Open
Abstract
Currently, research on intestinal diseases is mainly based on animal models and cell lines in monolayers. However, these models have drawbacks that limit scientific advances in this field. Three-dimensional (3D) culture systems named organoids are emerging as a reliable research tool for recapitulating the human intestinal epithelium and represent a unique platform for patient-specific drug testing. Intestinal organoids (IOs) are crypt–villus structures that can be derived from adult intestinal stem cells (ISCs), embryonic stem cells (ESCs), or induced pluripotent stem cells (iPSCs) and have the potential to serve as a platform for individualized medicine and research. However, this emerging field has not been bibliometric summarized to date. Here, we performed a bibliometric analysis of the Web of Science Core Collection (WoSCC) database to evaluate 5,379 publications concerning the use of organoids; the studies were divided into four clusters associated with the current situation and future directions for the application of IOs. Based on the results of our bibliometric analysis of IO applications, we systematically summarized the latest advances and analyzed the limitations and prospects.
Collapse
Affiliation(s)
- Meng-Meng Zhang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China
| | - Ke-Lu Yang
- Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou, China
| | - Yan-Cheng Cui
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Yu-Shi Zhou
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Hao-Ran Zhang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Quan Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China
| | - Ying-Jiang Ye
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China
| | - Shan Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China
| | - Ke-Wei Jiang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China
| |
Collapse
|