1
|
Xin M, Li Q, Wang D, Wang Z. Organoids for Cancer Research: Advances and Challenges. Adv Biol (Weinh) 2024; 8:e2400056. [PMID: 38977414 DOI: 10.1002/adbi.202400056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/04/2024] [Indexed: 07/10/2024]
Abstract
As 3D culture technology advances, new avenues have opened for the development of physiological human cancer models. These preclinical models provide efficient ways to translate basic cancer research into clinical tumor therapies. Recently, cancer organoids have emerged as a model to dissect the more complex tumor microenvironment. Incorporating cancer organoids into preclinical programs have the potential to increase the success rate of oncology drug development and recapitulate the most efficacious treatment regimens for cancer patients. In this review, four main types of cancer organoids are introduced, their applications, advantages, limitations, and prospects are discussed, as well as the recent application of single-cell RNA-sequencing (scRNA-seq) in exploring cancer organoids to advance this field.
Collapse
Affiliation(s)
- Miaomaio Xin
- Assisted Reproductive Center, Women's & Children's Hospital of Northwest, Xi'an, Shanxi Province, 710000, China
- University of South Bohemia in Ceske Budejovice, Vodnany, 38925, Czech Republic
| | - Qian Li
- Changsha Medical University, Changsha, Hunan Province, 410000, China
| | - Dongyang Wang
- Assisted Reproductive Center, Women's & Children's Hospital of Northwest, Xi'an, Shanxi Province, 710000, China
| | - Zheng Wang
- Medical Center of Hematology, the Second Affiliated Hospital, Army Medical University, Chongqing, Sichuan Province, 404100, China
| |
Collapse
|
2
|
Li DM, Li GS, Li JD, Chen F, Huang H, Huang WY, Huang ZG, Dang YW, Tang YL, Tang ZQ, Tang WJ, Chen G, Lu HP. Clinical significance and prospective mechanism of increased CDKN2A expression in small cell lung cancer. Clin Transl Oncol 2024; 26:1519-1531. [PMID: 38206516 PMCID: PMC11108933 DOI: 10.1007/s12094-023-03376-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Although it has been shown that cyclin dependent kinase inhibitor 2A (CDKN2A) plays a significant role in a number of malignancies, its clinicopathological value and function in small cell lung cancer (SCLC) is unclear and warrants additional research. METHODS The clinical significance of CDKN2A expression in SCLC was examined by multiple methods, including comprehensive integration of mRNA level by high throughput data, Kaplan-Meier survival analysis for prognostic value, and validation of its protein expression using in-house immunohistochemistry. RESULTS The expression of CDKN2A mRNA in 357 cases of SCLC was evidently higher than that in the control group (n = 525) combing the data from 20 research centers worldwide. The standardized mean difference (SMD) was 3.07, and the area under the curve (AUC) of summary receiver operating characteristic curve (sROC) was 0.97 for the overexpression of CDKN2A. ACC, COAD, KICH, KIRC, PCPG, PRAD, UCEC, UVM patients with higher CDKN2A expression had considerably worse overall survival rates than those with lower CDKN2A expression with the hazard ratio (HR) > 1. CONCLUSION CDKN2A upregulation extensively enhances the carcinogenesis and progression of SCLC.
Collapse
Affiliation(s)
- Dong-Ming Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Guo-Sheng Li
- Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Jian-Di Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Feng Chen
- Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Hong Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Wan-Ying Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Zhi-Guang Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Yi-Wu Dang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Yu-Lu Tang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Zhong-Qing Tang
- Department of Pathology, Wuzhou Gongren Hospital, The Seventh Affiliated Hospital of Guangxi Medical University, No.1, Nansanxiang Gaodi Road, Guangxi Zhuang Autonomous Region, Wuzhou, 543000, People's Republic of China
| | - Wen-Jia Tang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China
| | - Hui-Ping Lu
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region, Nanning, People's Republic of China.
| |
Collapse
|
3
|
Zhong Y, Zeng W, Chen Y, Zhu X. The effect of lipid metabolism on cuproptosis-inducing cancer therapy. Biomed Pharmacother 2024; 172:116247. [PMID: 38330710 DOI: 10.1016/j.biopha.2024.116247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024] Open
Abstract
Cuproptosis provides a new therapeutic strategy for cancer treatment and is thought to have broad clinical application prospects. Nevertheless, some oncological clinical trials have yet to demonstrate favorable outcomes, highlighting the need for further research into the molecular mechanisms underlying cuproptosis in tumors. Cuproptosis primarily hinges on the intracellular accumulation of copper, with lipid metabolism exerting a profound influence on its course. The interaction between copper metabolism and lipid metabolism is closely related to cuproptosis. Copper imbalance can affect mitochondrial respiration and lipid metabolism changes, while lipid accumulation can promote copper uptake and absorption, and inhibit cuproptosis induced by copper. Anomalies in lipid metabolism can disrupt copper homeostasis within cells, potentially triggering cuproptosis. The interaction between cuproptosis and lipid metabolism regulates the occurrence, development, metastasis, chemotherapy drug resistance, and tumor immunity of cancer. Cuproptosis is a promising new target for cancer treatment. However, the influence of lipid metabolism and other factors should be taken into consideration. This review provides a brief overview of the characteristics of the interaction between cuproptosis and lipid metabolism in cancer and analyses potential strategies of applying cuproptosis for cancer treatment.
Collapse
Affiliation(s)
- Yue Zhong
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Wei Zeng
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yongbo Chen
- Rehabilitation College of Gannan Medical University, Ganzhou 341000, China
| | - Xiuzhi Zhu
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
4
|
Song T, Kong B, Liu R, Luo Y, Wang Y, Zhao Y. Bioengineering Approaches for the Pancreatic Tumor Organoids Research and Application. Adv Healthc Mater 2024; 13:e2300984. [PMID: 37694339 DOI: 10.1002/adhm.202300984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/29/2023] [Indexed: 09/12/2023]
Abstract
Pancreatic cancer is a highly lethal form of digestive malignancy that poses significant health risks to individuals worldwide. Chemotherapy-based comprehensive treatment is the primary therapeutic approach for midlife and late-life patients. Nevertheless, the heterogeneity of the tumor and individual genetic backgrounds result in substantial variations in drug sensitivity among patients, rendering a single treatment regimen unsuitable for all patients. Conventional pancreatic cancer tumor organoid models are capable of emulating the biological traits of pancreatic cancer and are utilized in drug development and screening. However, these tumor organoids can still not mimic the tumor microenvironment (TME) in vivo, and the poor controllability in the preparation process hinders translation from essential drug screening to clinical pharmacological therapy. In recent years, many engineering methods with remarkable results have been used to develop pancreatic cancer organoid models, including bio-hydrogel, co-culture, microfluidic, and gene editing. Here, this work summarizes and analyzes the recent developments in engineering pancreatic tumor organoid models. In addition, the future direction of improving engineered pancreatic cancer organoids is discussed for their application prospects in clinical treatment.
Collapse
Affiliation(s)
- Taiyu Song
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Bin Kong
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Rui Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
5
|
Ei ZZ, Racha S, Yokoya M, Hotta D, Zou H, Chanvorachote P. Simplified Synthesis of Renieramycin T Derivatives to Target Cancer Stem Cells via β-Catenin Proteasomal Degradation in Human Lung Cancer. Mar Drugs 2023; 21:627. [PMID: 38132948 PMCID: PMC10744608 DOI: 10.3390/md21120627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Cancer stem cells (CSCs) found within cancer tissue play a pivotal role in its resistance to therapy and its potential to metastasize, contributing to elevated mortality rates among patients. Significant strides in understanding the molecular foundations of CSCs have led to preclinical investigations and clinical trials focused on CSC regulator β-catenin signaling targeted interventions in malignancies. As part of the ongoing advancements in marine-organism-derived compound development, it was observed that among the six analogs of Renieramycin T (RT), a potential lead alkaloid from the blue sponge Xestospongia sp., the compound DH_32, displayed the most robust anti-cancer activity in lung cancer A549, H23, and H292 cells. In various lung cancer cell lines, DH_32 exhibited the highest efficacy, with IC50 values of 4.06 ± 0.24 μM, 2.07 ± 0.11 μM, and 1.46 ± 0.06 μM in A549, H23, and H292 cells, respectively. In contrast, parental RT compounds had IC50 values of 5.76 ± 0.23 μM, 2.93 ± 0.07 μM, and 1.52 ± 0.05 μM in the same order. Furthermore, at a dosage of 25 nM, DH_32 showed a stronger ability to inhibit colony formation compared to the lead compound, RT. DH_32 was capable of inducing apoptosis in lung cancer cells, as demonstrated by increased PARP cleavage and reduced levels of the proapoptotic protein Bcl2. Our discovery confirms that DH_32 treatment of lung cancer cells led to a reduced level of CD133, which is associated with the suppression of stem-cell-related transcription factors like OCT4. Moreover, DH_32 significantly suppressed the ability of tumor spheroids to form compared to the original RT compound. Additionally, DH_32 inhibited CSCs by promoting the degradation of β-catenin through ubiquitin-proteasomal pathways. In computational molecular docking, a high-affinity interaction was observed between DH_32 (grid score = -35.559 kcal/mol) and β-catenin, indicating a stronger binding interaction compared to the reference compound R9Q (grid score = -29.044 kcal/mol). In summary, DH_32, a newly developed derivative of the right-half analog of RT, effectively inhibited the initiation of lung cancer spheroids and the self-renewal of lung cancer cells through the upstream process of β-catenin ubiquitin-proteasomal degradation.
Collapse
Affiliation(s)
- Zin Zin Ei
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (Z.Z.E.); (S.R.)
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Satapat Racha
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (Z.Z.E.); (S.R.)
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Interdisciplinary Program in Pharmacology, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand
| | - Masashi Yokoya
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo 204-8588, Japan; (M.Y.); (D.H.)
| | - Daiki Hotta
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, 2-522-1, Noshio, Kiyose, Tokyo 204-8588, Japan; (M.Y.); (D.H.)
| | - Hongbin Zou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China;
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (Z.Z.E.); (S.R.)
- Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
6
|
He R, Weng Z, Liu Y, Li B, Wang W, Meng W, Li B, Li L. Application of Induced Pluripotent Stem Cells in Malignant Solid Tumors. Stem Cell Rev Rep 2023; 19:2557-2575. [PMID: 37755647 PMCID: PMC10661832 DOI: 10.1007/s12015-023-10633-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 09/28/2023]
Abstract
In the past decade, induced pluripotent stem cells (iPSCs) technology has significantly progressed in studying malignant solid tumors. This technically feasible reprogramming techniques can reawaken sequestered dormant regions that regulate the fate of differentiated cells. Despite the evolving therapeutic modalities for malignant solid tumors, treatment outcomes have not been satisfactory. Recently, scientists attempted to apply induced pluripotent stem cell technology to cancer research, from modeling to treatment. Induced pluripotent stem cells derived from somatic cells, cancer cell lines, primary tumors, and individuals with an inherited propensity to develop cancer have shown great potential in cancer modeling, cell therapy, immunotherapy, and understanding tumor progression. This review summarizes the evolution of induced pluripotent stem cells technology and its applications in malignant solid tumor. Additionally, we discuss potential obstacles to induced pluripotent stem cell technology.
Collapse
Affiliation(s)
- Rong He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhijie Weng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunkun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bingzhi Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenxuan Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wanrong Meng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bo Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Longjiang Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
7
|
Krüger J, Fischer A, Breunig M, Allgöwer C, Schulte L, Merkle J, Mulaw MA, Okeke N, Melzer MK, Morgenstern C, Azoitei N, Seufferlein T, Barth TF, Siebert R, Hohwieler M, Kleger A. DNA methylation-associated allelic inactivation regulates Keratin 19 gene expression during pancreatic development and carcinogenesis. J Pathol 2023; 261:139-155. [PMID: 37555362 DOI: 10.1002/path.6156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 04/29/2023] [Accepted: 06/09/2023] [Indexed: 08/10/2023]
Abstract
Within the pancreas, Keratin 19 (KRT19) labels the ductal lineage and is a determinant of pancreatic ductal adenocarcinoma (PDAC). To investigate KRT19 expression dynamics, we developed a human pluripotent stem cell (PSC)-based KRT19-mCherry reporter system in different genetic backgrounds to monitor KRT19 expression from its endogenous gene locus. A differentiation protocol to generate mature pancreatic duct-like organoids was applied. While KRT19/mCherry expression became evident at the early endoderm stage, mCherry signal was present in nearly all cells at the pancreatic endoderm (PE) and pancreatic progenitor (PP) stages. Interestingly, despite homogenous KRT19 expression, mCherry positivity dropped to 50% after ductal maturation, indicating a permanent switch from biallelic to monoallelic expression. DNA methylation profiling separated the distinct differentiation intermediates, with site-specific DNA methylation patterns occurring at the KRT19 locus during ductal maturation. Accordingly, the monoallelic switch was partially reverted upon treatment with a DNA-methyltransferase inhibitor. In human PDAC cohorts, high KRT19 levels correlate with low locus methylation and decreased survival. At the same time, activation of oncogenic KRASG12D signalling in our reporter system reversed monoallelic back to biallelic KRT19 expression in pancreatic duct-like organoids. Allelic reactivation was also detected in single-cell transcriptomes of human PDACs, which further revealed a positive correlation between KRT19 and KRAS expression. Accordingly, KRAS mutant PDACs had higher KRT19 mRNA but lower KRT19 gene locus DNA methylation than wildtype counterparts. KRT19 protein was additionally detected in plasma of PDAC patients, with higher concentrations correlating with shorter progression-free survival in gemcitabine/nabPaclitaxel-treated and opposing trends in FOLFIRINOX-treated patients. Apart from being an important pancreatic ductal lineage marker, KRT19 appears tightly controlled via a switch from biallelic to monoallelic expression during ductal lineage entry and is aberrantly expressed after oncogenic KRASG12D expression, indicating a role in PDAC development and malignancy. Soluble KRT19 might serve as a relevant biomarker to stratify treatment. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jana Krüger
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
| | - Anja Fischer
- Institute of Human Genetics, Ulm University & Ulm University Hospital, Ulm, Germany
| | - Markus Breunig
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
| | - Chantal Allgöwer
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
| | - Lucas Schulte
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | | | - Medhanie A Mulaw
- Unit for Single-cell Genomics, Medical Faculty, Ulm University, Ulm, Germany
| | - Nnamdi Okeke
- Institute of Human Genetics, Ulm University & Ulm University Hospital, Ulm, Germany
| | - Michael K Melzer
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
- Department of Urology, Ulm University Hospital, Ulm, Germany
| | - Clara Morgenstern
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
| | - Ninel Azoitei
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Thomas Fe Barth
- Department of Pathology, Ulm University Hospital, Ulm, Germany
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University & Ulm University Hospital, Ulm, Germany
| | - Meike Hohwieler
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
| | - Alexander Kleger
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
- Organoid Core Facility, Ulm University, Ulm, Germany
| |
Collapse
|
8
|
Mastracci TL, Apte M, Amundadottir LT, Alvarsson A, Artandi S, Bellin MD, Bernal-Mizrachi E, Caicedo A, Campbell-Thompson M, Cruz-Monserrate Z, El Ouaamari A, Gaulton KJ, Geisz A, Goodarzi MO, Hara M, Hull-Meichle RL, Kleger A, Klein AP, Kopp JL, Kulkarni RN, Muzumdar MD, Naren AP, Oakes SA, Olesen SS, Phelps EA, Powers AC, Stabler CL, Tirkes T, Whitcomb DC, Yadav D, Yong J, Zaghloul NA, Pandol SJ, Sander M. Integrated Physiology of the Exocrine and Endocrine Compartments in Pancreatic Diseases: Workshop Proceedings. Diabetes 2023; 72:433-448. [PMID: 36940317 PMCID: PMC10033248 DOI: 10.2337/db22-0942] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/29/2022] [Indexed: 03/22/2023]
Abstract
The Integrated Physiology of the Exocrine and Endocrine Compartments in Pancreatic Diseases workshop was a 1.5-day scientific conference at the National Institutes of Health (Bethesda, MD) that engaged clinical and basic science investigators interested in diseases of the pancreas. This report provides a summary of the proceedings from the workshop. The goals of the workshop were to forge connections and identify gaps in knowledge that could guide future research directions. Presentations were segregated into six major theme areas, including 1) pancreas anatomy and physiology, 2) diabetes in the setting of exocrine disease, 3) metabolic influences on the exocrine pancreas, 4) genetic drivers of pancreatic diseases, 5) tools for integrated pancreatic analysis, and 6) implications of exocrine-endocrine cross talk. For each theme, multiple presentations were followed by panel discussions on specific topics relevant to each area of research; these are summarized here. Significantly, the discussions resulted in the identification of research gaps and opportunities for the field to address. In general, it was concluded that as a pancreas research community, we must more thoughtfully integrate our current knowledge of normal physiology as well as the disease mechanisms that underlie endocrine and exocrine disorders so that there is a better understanding of the interplay between these compartments.
Collapse
Affiliation(s)
- Teresa L. Mastracci
- Department of Biology, Indiana University–Purdue University Indianapolis, Indianapolis, IN
| | - Minoti Apte
- Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | | | - Alexandra Alvarsson
- Diabetes, Obesity, and Metabolism Institute, Mount Sinai Hospital, New York, NY
| | - Steven Artandi
- Department of Internal Medicine, Stanford University, Stanford, CA
| | - Melena D. Bellin
- Departments of Pediatrics and Surgery, University of Minnesota Medical School, Minneapolis, MN
| | - Ernesto Bernal-Mizrachi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Alejandro Caicedo
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
| | - Martha Campbell-Thompson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Zobeida Cruz-Monserrate
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - Kyle J. Gaulton
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Andrea Geisz
- Department of Molecular and Cell Biology, Boston University Henry M. Goldman School of Dental Medicine, Boston, MA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Manami Hara
- Department of Medicine, The University of Chicago, Chicago, IL
| | - Rebecca L. Hull-Meichle
- Department of Medicine, Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA
| | - Alexander Kleger
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University, Ulm, Germany
| | - Alison P. Klein
- Department of Pathology and Medicine, Johns Hopkins School of Medicine, Baltimore MD
| | - Janel L. Kopp
- Department of Cellular & Physiological Sciences, The University of British Columbia, Vancouver, Canada
| | | | - Mandar D. Muzumdar
- Departments of Genetics and Internal Medicine (Oncology), Yale University School of Medicine, New Haven, CT
| | | | - Scott A. Oakes
- Department of Pathology, The University of Chicago, Chicago, IL
| | - Søren S. Olesen
- Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Edward A. Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Alvin C. Powers
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN
| | - Cherie L. Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Temel Tirkes
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN
| | | | - Dhiraj Yadav
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Jing Yong
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Norann A. Zaghloul
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Stephen J. Pandol
- Department of Gastroenterology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Maike Sander
- Department of Pediatrics and Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA
| |
Collapse
|
9
|
Melzer MK, Resheq Y, Navaee F, Kleger A. The application of pancreatic cancer organoids for novel drug discovery. Expert Opin Drug Discov 2023; 18:429-444. [PMID: 36945198 DOI: 10.1080/17460441.2023.2194627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma presents with a dismal prognosis. Personalized therapy is urgently warranted to overcome the treatment limitations of the "one-size-fits-all" scheme. Organoids have emerged as fundamental novel tools to study tumor biology and heterogeneity, hence overcoming limitations of other model systems by better-reflecting tissue heterogeneity and recapitulating in-vivo processes. Besides their crucial role in basic research, they have evolved as tools for translational drug discovery and patient stratification. AREAS COVERED This review highlights the achievements of an organoid-based drug investigation and discovery. The authors present an overview of studies using organoids for drug testing. Further, they pinpoint studies correlating the in vitro prediction of organoids to the actual patient`s response. Furthermore, the authors describe novel model systems and take a thorough overlook of microfluidic chips, synthetic matrices, multicellular systems, bioprinting, and stem cell-derived pancreatic organoid systems. EXPERT OPINION Organoid systems promise great potential for future clinical applications. Indeed, they may be implemented into informed decision-making for guiding therapies. However, validation by randomized trials is mandatory. Additionally, organoids in combination with other cellular compartments may be exploited for drug discovery by studying niche-tumor interaction. Yet, several precautions must be kept in mind, such as standardization and reproducibility.
Collapse
Affiliation(s)
- Michael Karl Melzer
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
- Department of Urology, Ulm University Hospital, Ulm, Germany
| | - Yazid Resheq
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Fatemeh Navaee
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
| | - Alexander Kleger
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, Germany
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine 1, Ulm University Hospital, Ulm, Germany
- Core Facility Organoids, Ulm University, Ulm, Germany
| |
Collapse
|
10
|
Melzer MK, Schirge S, Gout J, Arnold F, Srinivasan D, Burtscher I, Allgöwer C, Mulaw M, Zengerling F, Günes C, Lickert H, Christoffels VM, Liebau S, Wagner M, Seufferlein T, Bolenz C, Moon AM, Perkhofer L, Kleger A. TBX3 is dynamically expressed in pancreatic organogenesis and fine-tunes regeneration. BMC Biol 2023; 21:55. [PMID: 36941669 PMCID: PMC10029195 DOI: 10.1186/s12915-023-01553-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 02/27/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND The reactivation of genetic programs from early development is a common mechanism for injury-induced organ regeneration. T-box 3 (TBX3) is a member of the T-box family of transcription factors previously shown to regulate pluripotency and subsequent lineage commitment in a number of tissues, including limb and lung. TBX3 is also involved in lung and heart organogenesis. Here, we provide a comprehensive and thorough characterization of TBX3 and its role during pancreatic organogenesis and regeneration. RESULTS We interrogated the level and cell specificity of TBX3 in the developing and adult pancreas at mRNA and protein levels at multiple developmental stages in mouse and human pancreas. We employed conditional mutagenesis to determine its role in murine pancreatic development and in regeneration after the induction of acute pancreatitis. We found that Tbx3 is dynamically expressed in the pancreatic mesenchyme and epithelium. While Tbx3 is expressed in the developing pancreas, its absence is likely compensated by other factors after ablation from either the mesenchymal or epithelial compartments. In an adult model of acute pancreatitis, we found that a lack of Tbx3 resulted in increased proliferation and fibrosis as well as an enhanced inflammatory gene programs, indicating that Tbx3 has a role in tissue homeostasis and regeneration. CONCLUSIONS TBX3 demonstrates dynamic expression patterns in the pancreas. Although TBX3 is dispensable for proper pancreatic development, its absence leads to altered organ regeneration after induction of acute pancreatitis.
Collapse
Affiliation(s)
- Michael Karl Melzer
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
- Clinic of Urology, Ulm University Hospital, Ulm, 89081, Germany
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany
| | - Silvia Schirge
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Johann Gout
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany
| | - Frank Arnold
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany
| | - Dharini Srinivasan
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany
| | - Ingo Burtscher
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Chantal Allgöwer
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany
| | - Medhanie Mulaw
- Unit for Single-cell Genomics, Ulm University, 89081, Ulm, Germany
| | | | - Cagatay Günes
- Clinic of Urology, Ulm University Hospital, Ulm, 89081, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Zentrum München, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
- Chair of b-Cell Biology, Technische Universität München, School of Medicine, Klinikum Rechts der Isar, 81675, München, Germany
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 15, 1105AZ, Amsterdam, The Netherlands
| | - Stefan Liebau
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Österbergstrasse 3, 72074, Tübingen, Germany
| | - Martin Wagner
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
| | - Thomas Seufferlein
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
| | | | - Anne M Moon
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville, PA, USA
- Department of Human Genetics (adjunct), University of Utah, Salt Lake City, UT, USA
- The Mindich Child Health and Development Institute, Hess Center for Science and Medicine at Mount Sinai, New York, NY, USA
| | - Lukas Perkhofer
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany
| | - Alexander Kleger
- Clinic of Internal Medicine I, Ulm University Hospital, Ulm, 89081, Germany.
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, Ulm, 89081, Germany.
- Core Facility Organoids, Ulm University, 89081, Ulm, Germany.
| |
Collapse
|
11
|
Protocol to use de-epithelialized porcine urinary bladder as a tissue scaffold for propagation of pancreatic cells. STAR Protoc 2022; 3:101869. [PMID: 36595896 PMCID: PMC9692066 DOI: 10.1016/j.xpro.2022.101869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/03/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022] Open
Abstract
Ex vivo organ culture can be a useful alternative to in vivo models, which can be time-, labor-, and cost-intensive. Here we describe a step-by-step protocol to use de-epithelialized porcine urinary bladders as scaffolds in air-liquid interface in vitro culture systems for a variety of pluripotent stem-cell-derived and patient-derived pancreatic cells and organoids. The scaffold can trigger cell maturation and enable cell-cell interaction and invasion capacity studies. However, this model is limited by the lack of functional vasculature. For complete details on the use and execution of this protocol, please refer to Melzer et al. (2022),1 Breunig et al. (2021),2 and Breunig et al. (2021).3.
Collapse
|
12
|
Melzer MK, Roger E, Kleger A. State-matched organoid models to fight pancreatic cancer. Trends Cancer 2022; 8:445-447. [PMID: 35370114 DOI: 10.1016/j.trecan.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 11/16/2022]
Abstract
As one of the deadliest cancers, pancreatic ductal adenocarcinoma (PDAC) requires sophisticated model systems to dissect disease onset, progression, and therapy resistance, as well as to personalize therapy. In recent years, patient- and pluripotent stem cell-derived organoids have become state-of-the-art systems to refine existing therapeutic strategies and deepen our knowledge of disease pathophysiology.
Collapse
Affiliation(s)
- Michael Karl Melzer
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany; Clinic of Urology, Ulm University Hospital, Ulm, Germany
| | - Elodie Roger
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Alexander Kleger
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany.
| |
Collapse
|
13
|
Melzer MK, Breunig M, Arnold F, Wezel F, Azoitei A, Roger E, Krüger J, Merkle J, Schütte L, Resheq Y, Hänle M, Zehe V, Zengerling F, Azoitei N, Klein L, Penz F, Singh SK, Seufferlein T, Hohwieler M, Bolenz C, Günes C, Gout J, Kleger A. Organoids at the PUB: The Porcine Urinary Bladder Serves as a Pancreatic Niche for Advanced Cancer Modeling. Adv Healthc Mater 2022; 11:e2102345. [PMID: 35114730 PMCID: PMC11468201 DOI: 10.1002/adhm.202102345] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/17/2021] [Indexed: 12/17/2022]
Abstract
Despite intensive research and progress in personalized medicine, pancreatic ductal adenocarcinoma remains one of the deadliest cancer entities. Pancreatic duct-like organoids (PDLOs) derived from human pluripotent stem cells (PSCs) or pancreatic cancer patient-derived organoids (PDOs) provide unique tools to study early and late stage dysplasia and to foster personalized medicine. However, such advanced systems are neither rapidly nor easily accessible and require an in vivo niche to study tumor formation and interaction with the stroma. Here, the establishment of the porcine urinary bladder (PUB) is revealed as an advanced organ culture model for shaping an ex vivo pancreatic niche. This model allows pancreatic progenitor cells to enter the ductal and endocrine lineages, while PDLOs further mature into duct-like tissue. Accordingly, the PUB offers an ex vivo platform for earliest pancreatic dysplasia and cancer if PDLOs feature KRASG12D mutations. Finally, it is demonstrated that PDOs-on-PUB i) resemble primary pancreatic cancer, ii) preserve cancer subtypes, iii) enable the study of niche epithelial crosstalk by spiking in pancreatic stellate and immune cells into the grafts, and finally iv) allow drug testing. In summary, the PUB advances the existing pancreatic cancer models by adding feasibility, complexity, and customization at low cost and high flexibility.
Collapse
Affiliation(s)
- Michael Karl Melzer
- Department of UrologyUlm UniversityUlm89081Germany
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Markus Breunig
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Frank Arnold
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Felix Wezel
- Department of UrologyUlm UniversityUlm89081Germany
| | - Anca Azoitei
- Department of UrologyUlm UniversityUlm89081Germany
| | - Elodie Roger
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Jana Krüger
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Jessica Merkle
- Department of Internal Medicine IUlm UniversityUlm89081Germany
- Core Facility OrganoidsUlm UniversityUlm89081Germany
| | - Lena Schütte
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Yazid Resheq
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Mark Hänle
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Viktor Zehe
- Department of UrologyUlm UniversityUlm89081Germany
| | | | - Ninel Azoitei
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Lukas Klein
- Department of GastroenterologyGastrointestinal Oncology and EndocrinologyUniversity Medicine GoettingenGoettingen37075Germany
| | - Frederike Penz
- Department of GastroenterologyGastrointestinal Oncology and EndocrinologyUniversity Medicine GoettingenGoettingen37075Germany
| | - Shiv K. Singh
- Department of GastroenterologyGastrointestinal Oncology and EndocrinologyUniversity Medicine GoettingenGoettingen37075Germany
| | | | - Meike Hohwieler
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | | | | | - Johann Gout
- Department of Internal Medicine IUlm UniversityUlm89081Germany
| | - Alexander Kleger
- Department of Internal Medicine IUlm UniversityUlm89081Germany
- Core Facility OrganoidsUlm UniversityUlm89081Germany
| |
Collapse
|
14
|
Shi WK, Li YH, Bai XS, Lin GL. The Cell Cycle-Associated Protein CDKN2A May Promotes Colorectal Cancer Cell Metastasis by Inducing Epithelial-Mesenchymal Transition. Front Oncol 2022; 12:834235. [PMID: 35311137 PMCID: PMC8929760 DOI: 10.3389/fonc.2022.834235] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/31/2022] [Indexed: 01/04/2023] Open
Abstract
Colorectal cancer (CRC) is a common gastrointestinal malignancy, and recurrence and metastasis contribute considerably to its high mortality. It is well known that the epithelial-mesenchymal transition (EMT) accelerates the rate of cancer cell dissemination and migration, thus promoting cancer metastasis. Targeted therapy is a common modality for cancer treatment, and it can play a role in inhibiting cancer progression. In this study, bioinformatics was used to search for genes associated with the prognosis of CRC. First, differential analysis was performed on colon and rectal cancer samples to obtain 2,840 and 3,177 differentially expressed genes (DEGs), respectively. A Venn diagram was then used to identify 262 overlapping genes from the two groups of DEGs and EMT-related genes. The overlapping genes were subjected to batch survival analysis and batch expression analysis successively, and nine genes were obtained whose high expression in CRC led to a poor prognosis. The least absolute shrinkage and selection operator (LASSO) prognostic model was then constructed to obtain the risk score formula. A nomogram was constructed to seek prognostic independent factors to obtain CDKN2A. Finally, CCK-8 assay, flow cytometry and western blotting assays were performed to analyze the cellular biological function of CDKN2A. The results showed that knockdown of CDKN2A expression inhibited HT-29 cell proliferation, promoted apoptosis and cell cycle progression, and affected the EMT process in CRC.
Collapse
Affiliation(s)
- Wei-Kun Shi
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yun-Hao Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xue-Shan Bai
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guo-Le Lin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|