1
|
Marquez J, Viviano S, Beckman E, Thies J, Friedland-Little J, Lam CT, Deniz E, Shelkowitz E. Polyamine metabolism is dysregulated in COXFA4-related mitochondrial disease. HGG ADVANCES 2025; 6:100418. [PMID: 39967265 PMCID: PMC11946867 DOI: 10.1016/j.xhgg.2025.100418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
Most of the chemical energy that organisms rely on to support cellular function is generated through oxidative phosphorylation, a metabolic pathway in which electron donors NADH and FADH are oxidized through a series of successive steps to generate adenosine triphosphate. These redox reactions are orchestrated by a series of five protein complexes that sit within the mitochondrial membrane. Deficiency of cytochrome c oxidase, the fourth of these complexes, is a recognized cause of mitochondrial disease. COXFA4 encodes one of the protein subunits of cytochrome c oxidase, and variants in COXFA4 have recently been reported in individuals with a range of symptoms. These symptoms can include feeding difficulties, poor growth, cardiomyopathy, Leigh or Leigh-like disease, and neurodevelopmental delay, although these symptoms vary widely between individuals. However, a mechanistic understanding of the connection between COXFA4 loss and these varied disease manifestations is lacking. Using animal modeling in Xenopus, we explored the ramifications of coxfa4 loss of function on the early developing heart. We then conducted a hypothesis naive analysis of cellular gene expression in the context of COXFA4 deletion and discovered a downstream deficiency in the ornithine decarboxylase pathway. Small-molecule-based modulation of the ornithine decarboxylase pathway in our model modified the extent of disease, including improvement of cardiac function. Our findings point to a mechanism by which COXFA4 dysfunction leads to tissue-specific disease.
Collapse
Affiliation(s)
- Jonathan Marquez
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, USA.
| | - Stephen Viviano
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA; Pediatric Genomics Discovery Program, Yale School of Medicine, New Haven, CT, USA
| | - Erika Beckman
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Jenny Thies
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Joshua Friedland-Little
- Division of Cardiology, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Christina T Lam
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, USA
| | - Engin Deniz
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA; Pediatric Genomics Discovery Program, Yale School of Medicine, New Haven, CT, USA
| | - Emily Shelkowitz
- Division of Genetic Medicine, Department of Pediatrics, University of Washington and Seattle Children's Hospital, Seattle, WA, USA.
| |
Collapse
|
2
|
Wang Z, Li Z, Ji R, Wang W, Li J, Xu W, Li X, Yang X, Du H, Liu D. Sesquiterpene lactone from Artemisia argyi inhibited cancer proliferation by inducing apoptosis and ferroptosis via key cell metabolism enzyme NDUFA4. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156312. [PMID: 39671784 DOI: 10.1016/j.phymed.2024.156312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/25/2024] [Accepted: 12/01/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Artemisia argyi is a well-known medicinal plant. A. argyi has been widely used in clinical for about 3000 years, owing to its extensive pharmacological activity. Among these, its anti-cancer properties are the most reported activity. However, its pharmacodynamic compounds remain unknown. PURPOSE This study aimed to investigate the potential anti-cancer compounds in A. argyi and reveal its molecular mechanisms and targets. METHODS Firstly, A. argyi were extracted with 70 % ethanol, yielding A. argyi EtOH (AAE) crude extracts. AAE was extracted with Ethyl acetate and Butanol successively to yield A. argyi EtOAc (AAEA) and A. argyi Butanol (AAB) sub-fraction. And, AAE, AAEA, and AAB were prepared to assess their anti-cancer ability in vitro and in vivo. Then, the natural products were isolated from active sub-fraction via activity-oriented separation and identification. Meanwhile, all the compounds were evaluated the anti-cancer effect. The anti-proliferation mechanism of representative compounds was explored, based on programmed cell death. Moreover, 4D-data-independent (DIA) quantitative proteomic studies were performed to reveal the underlying targets and mechanism of representative compounds. Finally, the pharmacodynamic compound and key target interaction were identified by the evaluation of targets function, molecular docking, surface plasmon resonance (SPR) assay, and small interfering RNA. In addition, the toxicity of pharmacodynamic compounds were evaluated by in vitro and zebrafish model in vivo. RESULTS AAEA demonstrated stronger inhibitory effects than AAB on various cancer cell lines in vitro. And, AAEA sub-fraction effectively inhibited the tumor growth in vitro and in vivo. Subsequently, we isolated and identified 47 anti-cancer components from AAEA, especially 23 of which were isolated from A. argyi for the first time. Among them, 8 sesquiterpenes compounds showed strong anti-cancer activity. Moreover, compound 3 (moxartenolide) exhibited stronger induction of apoptosis and ferroptosis. Ultimately, a series of studies based on proteomics revealed that Moxartenolide inhibited cancer cell proliferation through the key enzyme NDUFA4. In addition, toxicological evaluation in vivo and in vitro demonstrated the safety of the candidate drug. CONCLUSION These findings reveal the anti-cancer components of A. argyi based on activity-oriented separation and identification for the first time. Specially, Compound 3 (moxartenolide) inhibited cancer proliferation by inducing apoptosis and ferroptosis via key cell metabolism enzyme NDUFA4. Briefly, it suggests that A. argyi has the potential of anti-cancer drug development.
Collapse
Affiliation(s)
- Ziling Wang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Zhouyuan Li
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Rongsheng Ji
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Wenjing Wang
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Peoples R China
| | - Jing Li
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Peoples R China
| | - Wenli Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xiaoxuan Li
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xiaolong Yang
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, Peoples R China.
| | - Hongzhi Du
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| | - Dahui Liu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
3
|
Liu N, Guan M, Ma B, Chu H, Tian G, Zhang Y, Li C, Zheng W, Wang X. Unraveling genetic mysteries: A comprehensive review of GWAS and DNA insights in animal and plant pathosystems. Int J Biol Macromol 2025; 285:138216. [PMID: 39631605 DOI: 10.1016/j.ijbiomac.2024.138216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/13/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
DNA serves as the carrier of genetic information, with sequence variations playing a pivotal role in defining hereditary traits. Genome-Wide Association Studies (GWAS) facilitate the investigation of the links between genetic variations and phenotypes, significantly influencing biological research, particularly in animal and plant pathology. By identifying genetic markers associated with specific traits or diseases, GWAS enhances our understanding of host-pathogen interactions and improves disease-resistant breeding strategies. It has been vital in revealing the genetic basis of disease resistance, pinpointing key genes and DNA loci, which enrich genetic resources for breeding programs and deepen our knowledge of disease resistance mechanisms at the DNA level. Additionally, GWAS contributes to pathogen population genetics, facilitating a thorough exploration of pathogen virulence. Integrating GWAS with marker-assisted selection enhances breeding efficiency and precision in selecting for disease-resistant traits. While previous research has largely focused on host genetics, the genetic variation of pathogens is equally significant. Notably, reports integrating animal and plant pathosystems are still lacking. Given the importance of these systems, this review summarizes key advancements in this field, addresses current challenges, and proposes future directions, thereby offering a vital reference for ongoing research.
Collapse
Affiliation(s)
- Na Liu
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China
| | - Mengxin Guan
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China
| | - Baozhan Ma
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China
| | - Hao Chu
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China
| | - Guangxiang Tian
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China
| | - Yanyan Zhang
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China
| | - Chuang Li
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China; Center of Crop Genome Engineering, College of Agronomy, Henan Agricultural University, 450046 Zhengzhou, China.
| | - Wenming Zheng
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China.
| | - Xu Wang
- Collaborative Innovation Center of Henan Grain Crops/State Key Laboratory of Wheat and Maize Crop Science, College of Life Sciences, Henan Agricultural University, 450046 Zhengzhou, China.
| |
Collapse
|
4
|
Hojo H, Tani S, Ohba S. Modeling of skeletal development and diseases using human pluripotent stem cells. J Bone Miner Res 2024; 40:5-19. [PMID: 39498496 DOI: 10.1093/jbmr/zjae178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/28/2024] [Accepted: 11/02/2024] [Indexed: 01/07/2025]
Abstract
Human skeletal elements are formed from distinct origins at distinct positions of the embryo. For example, the neural crest produces the facial bones, the paraxial mesoderm produces the axial skeleton, and the lateral plate mesoderm produces the appendicular skeleton. During skeletal development, different combinations of signaling pathways are coordinated from distinct origins during the sequential developmental stages. Models for human skeletal development have been established using human pluripotent stem cells (hPSCs) and by exploiting our understanding of skeletal development. Stepwise protocols for generating skeletal cells from different origins have been designed to mimic developmental trails. Recently, organoid methods have allowed the multicellular organization of skeletal cell types to recapitulate complicated skeletal development and metabolism. Similarly, several genetic diseases of the skeleton have been modeled using patient-derived induced pluripotent stem cells and genome-editing technologies. Model-based drug screening is a powerful tool for identifying drug candidates. This review briefly summarizes our current understanding of the embryonic development of skeletal tissues and introduces the current state-of-the-art hPSC methods for recapitulating skeletal development, metabolism, and diseases. We also discuss the current limitations and future perspectives for applications of the hPSC-based modeling system in precision medicine in this research field.
Collapse
Affiliation(s)
- Hironori Hojo
- Division of Clinical Biotechnology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8655, Japan
| | - Shoichiro Tani
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Shinsuke Ohba
- Department of Tissue and Developmental Biology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
5
|
Zhang L, Qin J, Li P. Bioinformatics analysis of potential common pathogenic mechanisms for COVID-19 and venous thromboembolism. Cytokine 2024; 181:156682. [PMID: 38909539 DOI: 10.1016/j.cyto.2024.156682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/20/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND A growing body of research has shown that patients with coronavirus disease 2019 (COVID-19) have significantly higher rates of venous thromboembolism (VTE) than healthy. However, the mechanism remains incompletely elucidated. This study aimed to further investigate the molecular mechanisms underlying the development of this complication. METHODS The gene expression profiles of COVID-19 and VTE were downloaded from the Gene Expression Omnibus (GEO) database. After identifying the common differentially expressed genes (DEGs) for COVID-19 and VTE, functional annotation, a protein-protein interactions (PPI) network, module construction, and hub gene identification were performed. Finally, we constructed a transcription factor (TF)-gene regulatory network and a TF-miRNA regulatory network for hub genes. RESULTS A total of 42 common DEGs were selected for subsequent analyses. Functional analyses showed that biological function and signaling pathways collectively participated in the development and progression of VTE and COVID-19. Finally, 8 significant hub genes were identified using the cytoHubba plugin, including RSL24D1, RPS17, RPS27, HINT1, COX7C, RPL35, RPL34, and NDUFA4, which had preferable values as diagnostic markers for COVID-19 and VTE. CONCLUSIONS Our study revealed the common pathogenesis of COVID-19 and VTE. These common pathways and pivotal genes may provide new ideas for further mechanistic studies.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Jing Qin
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Peiwu Li
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
6
|
Xiong M, Liu Z, Wang B, Sokolich T, Graham N, Chen M, Wang WL, Boldin MP. The epithelial C15ORF48/miR-147-NDUFA4 axis is an essential regulator of gut inflammation, energy metabolism, and the microbiome. Proc Natl Acad Sci U S A 2024; 121:e2315944121. [PMID: 38917002 PMCID: PMC11228508 DOI: 10.1073/pnas.2315944121] [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: 09/19/2023] [Accepted: 05/13/2024] [Indexed: 06/27/2024] Open
Abstract
Chronic inflammation is epidemiologically linked to the pathogenesis of gastrointestinal diseases, including inflammatory bowel disease (IBD) and colorectal cancer (CRC). However, our understanding of the molecular mechanisms controlling gut inflammation remains insufficient, hindering the development of targeted therapies for IBD and CRC. In this study, we uncovered C15ORF48/miR-147 as a negative regulator of gut inflammation, operating through the modulation of epithelial cell metabolism. C15ORF48/miR-147 encodes two molecular products, C15ORF48 protein and miR-147-3p microRNA, which are predominantly expressed in the intestinal epithelium. C15ORF48/miR-147 ablation leads to gut dysbiosis and exacerbates chemically induced colitis in mice. C15ORF48 and miR-147-3p work together to suppress colonocyte metabolism and inflammation by silencing NDUFA4, a subunit of mitochondrial complex IV (CIV). Interestingly, the C15ORF48 protein, a structural paralog of NDUFA4, contains a unique C-terminal α-helical domain crucial for displacing NDUFA4 from CIV and its subsequent degradation. NDUFA4 silencing hinders NF-κB signaling activation and consequently attenuates inflammatory responses. Collectively, our findings have established the C15ORF48/miR-147-NDUFA4 molecular axis as an indispensable regulator of gut homeostasis, bridging mitochondrial metabolism and inflammation.
Collapse
Affiliation(s)
- Min Xiong
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA91010
| | - Ze Liu
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Bintao Wang
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA91010
| | - Thomas Sokolich
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA91010
| | - Natalie Graham
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA91010
| | - Meirong Chen
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu210009, China
| | - Wei-Le Wang
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA91010
| | - Mark P. Boldin
- Department of Systems Biology, Beckman Research Institute, City of Hope, Duarte, CA91010
| |
Collapse
|
7
|
Kimura M, Takebe T. Cellotype-phenotype associations using 'organoid villages'. Trends Endocrinol Metab 2024; 35:462-465. [PMID: 38575442 PMCID: PMC11752945 DOI: 10.1016/j.tem.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024]
Abstract
En masse phenotyping technology, using massively mosaic donor-derived cells and organoids, can offer enriched insights for cellotype-phenotype association in a cell-type-specific regulatory context. This emerging approach will help to discover biomarkers, inform genetic-epigenetic interactions and identify personalized therapeutic targets, offering hope for precision medicine against highly heterogeneous metabolic diseases.
Collapse
Affiliation(s)
- Masaki Kimura
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Gastroenterology, Hepatology and Nutrition, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Takanori Takebe
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Gastroenterology, Hepatology and Nutrition, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan.
| |
Collapse
|
8
|
Cao Z, Kong F, Ding J, Chen C, He F, Deng W. Promoting Alzheimer's disease research and therapy with stem cell technology. Stem Cell Res Ther 2024; 15:136. [PMID: 38715083 PMCID: PMC11077895 DOI: 10.1186/s13287-024-03737-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a prevalent form of dementia leading to memory loss, reduced cognitive and linguistic abilities, and decreased self-care. Current AD treatments aim to relieve symptoms and slow disease progression, but a cure is elusive due to limited understanding of the underlying disease mechanisms. MAIN CONTENT Stem cell technology has the potential to revolutionize AD research. With the ability to self-renew and differentiate into various cell types, stem cells are valuable tools for disease modeling, drug screening, and cell therapy. Recent advances have broadened our understanding beyond the deposition of amyloidβ (Aβ) or tau proteins in AD to encompass risk genes, immune system disorders, and neuron-glia mis-communication, relying heavily on stem cell-derived disease models. These stem cell-based models (e.g., organoids and microfluidic chips) simulate in vivo pathological processes with extraordinary spatial and temporal resolution. Stem cell technologies have the potential to alleviate AD pathology through various pathways, including immunomodulation, replacement of damaged neurons, and neurotrophic support. In recent years, transplantation of glial cells like oligodendrocytes and the infusion of exosomes have become hot research topics. CONCLUSION Although stem cell-based models and therapies for AD face several challenges, such as extended culture time and low differentiation efficiency, they still show considerable potential for AD treatment and are likely to become preferred tools for AD research.
Collapse
Affiliation(s)
- Zimeng Cao
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Fanshu Kong
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jiaqi Ding
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Chunxia Chen
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Fumei He
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China.
- School of Pharmaceutical Sciences, Dali University, Dali, 671000, China.
| | - Wenbin Deng
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
9
|
Yin DE, Palin AC, Lombo TB, Mahon RN, Poon B, Wu DY, Atala A, Brooks KM, Chen S, Coyne CB, D’Souza MP, Fackler OT, Furler O’Brien RL, Garcia-de-Alba C, Jean-Philippe P, Karn J, Majji S, Muotri AR, Ozulumba T, Sakatis MZ, Schlesinger LS, Singh A, Spiegel HM, Struble E, Sung K, Tagle DA, Thacker VV, Tidball AM, Varthakavi V, Vunjak-Novakovic G, Wagar LE, Yeung CK, Ndhlovu LC, Ott M. 3D human tissue models and microphysiological systems for HIV and related comorbidities. Trends Biotechnol 2024; 42:526-543. [PMID: 38071144 PMCID: PMC11065605 DOI: 10.1016/j.tibtech.2023.10.008] [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: 07/03/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 03/03/2024]
Abstract
Three-dimensional (3D) human tissue models/microphysiological systems (e.g., organs-on-chips, organoids, and tissue explants) model HIV and related comorbidities and have potential to address critical questions, including characterization of viral reservoirs, insufficient innate and adaptive immune responses, biomarker discovery and evaluation, medical complexity with comorbidities (e.g., tuberculosis and SARS-CoV-2), and protection and transmission during pregnancy and birth. Composed of multiple primary or stem cell-derived cell types organized in a dedicated 3D space, these systems hold unique promise for better reproducing human physiology, advancing therapeutic development, and bridging the human-animal model translational gap. Here, we discuss the promises and achievements with 3D human tissue models in HIV and comorbidity research, along with remaining barriers with respect to cell biology, virology, immunology, and regulatory issues.
Collapse
|
10
|
Zhou Q, Li X, Zhou H, Zhao J, Zhao H, Li L, Zhou Y. Mitochondrial respiratory chain component NDUFA4: a promising therapeutic target for gastrointestinal cancer. Cancer Cell Int 2024; 24:97. [PMID: 38443961 PMCID: PMC10916090 DOI: 10.1186/s12935-024-03283-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 02/24/2024] [Indexed: 03/07/2024] Open
Abstract
Gastrointestinal cancer, one of the most common cancers, continues to be a major cause of mortality and morbidity globally. Accumulating evidence has shown that alterations in mitochondrial energy metabolism are involved in developing various clinical diseases. NADH dehydrogenase 1 alpha subcomplex 4 (NDUFA4), encoded by the NDUFA4 gene located on human chromosome 7p21.3, is a component of mitochondrial respiratory chain complex IV and integral to mitochondrial energy metabolism. Recent researchers have disclosed that NDUFA4 is implicated in the pathogenesis of various diseases, including gastrointestinal cancer. Aberrant expression of NDUFA4 leads to the alteration in mitochondrial energy metabolism, thereby regulating the growth and metastasis of cancer cells, indicating that it might be a new promising target for cancer intervention. This article comprehensively reviews the structure, regulatory mechanism, and biological function of NDUFA4. Of note, the expression and roles of NDUFA4 in gastrointestinal cancer including colorectal cancer, liver cancer, gastric cancer, and so on were discussed. Finally, the existing problems of NDUFA4-based intervention on gastrointestinal cancer are discussed to provide help to strengthen the understanding of the carcinogenesis of gastrointestinal cancer, as well as the development of new strategies for clinical intervention.
Collapse
Affiliation(s)
- Quanling Zhou
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Department of Physics, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Xiaohui Li
- Department of Physics, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Honglian Zhou
- Department of Physics, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Juanjuan Zhao
- Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000, Guizhou, China
| | - Hailong Zhao
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Lijuan Li
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Ya Zhou
- Department of Pathophysiology, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
- Department of Physics, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
- Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
11
|
Yang L, Han Y, Zhou T, Lacko LA, Saeed M, Tan C, Danziger R, Zhu J, Zhao Z, Cahir C, Giani AM, Li Y, Dong X, Moroziewicz D, The NYSCF Global Stem Cell Array® Team, Paull D, Chen Z, Zhong A, Noggle SA, Rice CM, Qi Q, Evans T, Chen S. Isogenic human trophectoderm cells demonstrate the role of NDUFA4 and associated variants in ZIKV infection. iScience 2023; 26:107001. [PMID: 37534130 PMCID: PMC10391681 DOI: 10.1016/j.isci.2023.107001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/17/2023] [Accepted: 05/25/2023] [Indexed: 08/04/2023] Open
Abstract
Population-based genome-wide association studies (GWAS) normally require a large sample size, which can be labor intensive and costly. Recently, we reported a human induced pluripotent stem cell (hiPSC) array-based GWAS method, identifying NDUFA4 as a host factor for Zika virus (ZIKV) infection. In this study, we extended our analysis to trophectoderm cells, which constitute one of the major routes of mother-to-fetus transmission of ZIKV during pregnancy. We differentiated hiPSCs from various donors into trophectoderm cells. We then infected cells carrying loss of function mutations in NDUFA4, harboring risk versus non-risk alleles of SNPs (rs917172 and rs12386620) or having deletions in the NDUFA4 cis-regulatory region with ZIKV. We found that loss/reduction of NDUFA4 suppressed ZIKV infection in trophectoderm cells. This study validated our published hiPSC array-based system as a useful platform for GWAS and confirmed the role of NDUFA4 as a susceptibility locus for ZIKV in disease-relevant trophectoderm cells.
Collapse
Affiliation(s)
- Liuliu Yang
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Yuling Han
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Ting Zhou
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Stem Cell Research Facility, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Lauretta A. Lacko
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Mohsan Saeed
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
- Department of Biochemistry & Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
| | - Christina Tan
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Ron Danziger
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Jiajun Zhu
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Zeping Zhao
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Clare Cahir
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Alice Maria Giani
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Yang Li
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Xue Dong
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Dorota Moroziewicz
- The New York Stem Cell Foundation Research Institute, 619 West 54th Street, 3Road Floor, New York, NY 10019, USA
| | | | - Daniel Paull
- The New York Stem Cell Foundation Research Institute, 619 West 54th Street, 3Road Floor, New York, NY 10019, USA
| | - Zhengming Chen
- Department of Population Health Sciences, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Aaron Zhong
- Stem Cell Research Facility, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Scott A. Noggle
- The New York Stem Cell Foundation Research Institute, 619 West 54th Street, 3Road Floor, New York, NY 10019, USA
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Qibin Qi
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
- Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| |
Collapse
|
12
|
Vandana JJ, Manrique C, Lacko LA, Chen S. Human pluripotent-stem-cell-derived organoids for drug discovery and evaluation. Cell Stem Cell 2023; 30:571-591. [PMID: 37146581 PMCID: PMC10775018 DOI: 10.1016/j.stem.2023.04.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/27/2023] [Accepted: 04/11/2023] [Indexed: 05/07/2023]
Abstract
Human pluripotent stem cells (hPSCs) and three-dimensional organoids have ushered in a new era for disease modeling and drug discovery. Over the past decade, significant progress has been in deriving functional organoids from hPSCs, which have been applied to recapitulate disease phenotypes. In addition, these advancements have extended the application of hPSCs and organoids for drug screening and clinical-trial safety evaluations. This review provides an overview of the achievements and challenges in using hPSC-derived organoids to conduct relevant high-throughput, high-contentscreens and drug evaluation. These studies have greatly enhanced our knowledge and toolbox for precision medicine.
Collapse
Affiliation(s)
- J Jeya Vandana
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Tri-Institutional PhD Program in Chemical Biology, Weill Cornell Medicine, The Rockefeller University, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Cassandra Manrique
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medical College, New York, NY, USA
| | - Lauretta A Lacko
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Center for Genomic Health, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
13
|
Wells MF, Nemesh J, Ghosh S, Mitchell JM, Salick MR, Mello CJ, Meyer D, Pietilainen O, Piccioni F, Guss EJ, Raghunathan K, Tegtmeyer M, Hawes D, Neumann A, Worringer KA, Ho D, Kommineni S, Chan K, Peterson BK, Raymond JJ, Gold JT, Siekmann MT, Zuccaro E, Nehme R, Kaykas A, Eggan K, McCarroll SA. Natural variation in gene expression and viral susceptibility revealed by neural progenitor cell villages. Cell Stem Cell 2023; 30:312-332.e13. [PMID: 36796362 PMCID: PMC10581885 DOI: 10.1016/j.stem.2023.01.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/28/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023]
Abstract
Human genome variation contributes to diversity in neurodevelopmental outcomes and vulnerabilities; recognizing the underlying molecular and cellular mechanisms will require scalable approaches. Here, we describe a "cell village" experimental platform we used to analyze genetic, molecular, and phenotypic heterogeneity across neural progenitor cells from 44 human donors cultured in a shared in vitro environment using algorithms (Dropulation and Census-seq) to assign cells and phenotypes to individual donors. Through rapid induction of human stem cell-derived neural progenitor cells, measurements of natural genetic variation, and CRISPR-Cas9 genetic perturbations, we identified a common variant that regulates antiviral IFITM3 expression and explains most inter-individual variation in susceptibility to the Zika virus. We also detected expression QTLs corresponding to GWAS loci for brain traits and discovered novel disease-relevant regulators of progenitor proliferation and differentiation such as CACHD1. This approach provides scalable ways to elucidate the effects of genes and genetic variation on cellular phenotypes.
Collapse
Affiliation(s)
- Michael F Wells
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA; Department of Human Genetics, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - James Nemesh
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Sulagna Ghosh
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Jana M Mitchell
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA; Insitro, South San Francisco, CA 94080, USA
| | | | - Curtis J Mello
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel Meyer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Olli Pietilainen
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Federica Piccioni
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | - Ellen J Guss
- Department of Stem Cell and Regenerative Biology, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Kavya Raghunathan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Matthew Tegtmeyer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | - Derek Hawes
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | - Anna Neumann
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA
| | - Kathleen A Worringer
- Department of Neuroscience, Novartis Institute for BioMedical Research, Cambridge, MA 02139, USA
| | - Daniel Ho
- Department of Neuroscience, Novartis Institute for BioMedical Research, Cambridge, MA 02139, USA
| | - Sravya Kommineni
- Department of Neuroscience, Novartis Institute for BioMedical Research, Cambridge, MA 02139, USA
| | - Karrie Chan
- Department of Neuroscience, Novartis Institute for BioMedical Research, Cambridge, MA 02139, USA
| | - Brant K Peterson
- Department of Neuroscience, Novartis Institute for BioMedical Research, Cambridge, MA 02139, USA
| | - Joseph J Raymond
- Department of Neuroscience, Novartis Institute for BioMedical Research, Cambridge, MA 02139, USA
| | - John T Gold
- Department of Stem Cell and Regenerative Biology, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA; Department of Biology, Davidson College, Davidson, NC 28035, USA
| | - Marco T Siekmann
- Department of Stem Cell and Regenerative Biology, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Emanuela Zuccaro
- Department of Stem Cell and Regenerative Biology, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Ralda Nehme
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | | | - Kevin Eggan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, and Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| | - Steven A McCarroll
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard University, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|