1
|
Jiang T, Guo Q, Lyu L, Jing X, Li J, Zuo C, Qi X, Jiang W, Yao J, Wei M. Establishment, identification, and transcriptome analysis of a Sertoli cell line from ovoviviparous black rockfish Sebastes schlegelii. FISH PHYSIOLOGY AND BIOCHEMISTRY 2025; 51:95. [PMID: 40343626 DOI: 10.1007/s10695-025-01509-8] [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: 12/10/2024] [Accepted: 04/29/2025] [Indexed: 05/11/2025]
Abstract
Black rockfish (Sebastes schlegelii) is an economically important species with a unique ovoviviparous reproductive mode, in which reproduction is limited by incomplete fertilization. In order to understand the mechanism of spermatogenesis of black rockfish, a cell line derived from the testis, named SSTC, was successfully established and cultured in L-15 medium at 25 °C, and it was passaged to the 50th generation. The SSTC exhibited fibroblast-like and epithelial-like morphology during in vitro culture, and 62% of the SSTC retained the diploid karyotype with 48 chromosomes by the 30th passage (P30). To evaluate the ability of SSTC to express exogenous genes, lipofection and electrotransfection were performed, achieving transfection efficiencies of 9% and 12%, respectively. Transcriptome analysis showed that SSTC at P15 and P30 scarcely expressed germ cell and Leydig cell marker genes, while only expressing the Sertoli cell marker genes sox9a, amh, krt18 and fasl, indicating that SSTC mainly consists of Sertoli cells. GO and KEGG enrichment analyses revealed that, compared to the primary cells, the MAPK, TGF-β, and Wnt signaling pathways, which are crucial for spermatogenesis in Sertoli cells, were significantly upregulated in SSTC after passaging. Additionally, cell cycle-related pathways were upregulated, while pathways associated with cell adhesion, extracellular matrix, cell communication and membrane signal transduction were significantly downregulated. This study demonstrated that SSTC can be used as a tool for exploring the molecular mechanisms of gonadal differentiation and development in black rockfish, providing an effective platform for research on reproduction and endocrinology in this species.
Collapse
Affiliation(s)
- Tianyu Jiang
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao, 266003, China
| | - Qingqing Guo
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao, 266003, China
| | - Likang Lyu
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao, 266003, China
| | - Xiao Jing
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao, 266003, China
| | - Jianshuang Li
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao, 266003, China
| | - Chenpeng Zuo
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao, 266003, China
| | - Xin Qi
- Key Laboratory of Mariculture, Ministry of Education (KLMME), Ocean University of China, Qingdao, 266003, China
| | - Weiming Jiang
- ASEAN Key Laboratory of Comprehensive Exploitation and Utilization of Aquatic Germplasm Resources, Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquaculture Genetic and Breeding and Healthy Aquaculture of Guangxi, Guangxi Academy of Fishery Sciences, Nanning, 530021, China
| | - Jiuxiang Yao
- ASEAN Key Laboratory of Comprehensive Exploitation and Utilization of Aquatic Germplasm Resources, Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquaculture Genetic and Breeding and Healthy Aquaculture of Guangxi, Guangxi Academy of Fishery Sciences, Nanning, 530021, China
| | - Mingli Wei
- ASEAN Key Laboratory of Comprehensive Exploitation and Utilization of Aquatic Germplasm Resources, Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquaculture Genetic and Breeding and Healthy Aquaculture of Guangxi, Guangxi Academy of Fishery Sciences, Nanning, 530021, China.
| |
Collapse
|
2
|
Krebs T, Bauer J, Graff S, Teich L, Sterneberg M, Gebert M, Seibel H, Seeger B, Hellmann J, Wessel Ø, Rimstad E, Surachetpong W, Steinhagen D, Jung‐Schroers V, Adamek M. Beating Cardiac Cell Cultures From Different Developmental Stages of Rainbow Trout as a Novel Approach for Replication of Cardiac Fish Viruses. JOURNAL OF FISH DISEASES 2025; 48:e14080. [PMID: 39821901 PMCID: PMC11976189 DOI: 10.1111/jfd.14080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/19/2025]
Abstract
Piscine orthoreovirus-1 and 3 (PRV-1, PRV-3) cause highly prevalent infection in cultured salmonids and can induce heart and skeletal muscle inflammation (HSMI) resulting in economic losses in aquaculture. However, to date, PRV-1 and PRV-3 have withstood replication in continuous cell lines. In this study, we used beating heart cell cultures obtained from different developmental stages of rainbow trout (Oncorhynchus mykiss) (RTC-L and RTC-A) and tested their ability to sustain replication of PRV-1 and PRV-3. Furthermore, we compared the replication pattern of the different viruses with those in the newly developed heart fibroblast cell line (RTH-F) and the traditional established rainbow trout gonad cell line (RTG-2). Neither RTCs nor RTH-F cell lines supported replication of PRV-1 and PRV-3. Comparative experiments showed varying susceptibility of the novel cultures to viral haemorrhagic septicaemia virus (VHSV), chum salmon reovirus (CSV), infectious pancreatic necrosis virus (IPNV), piscine myocarditis virus (PMCV), salmonid alphavirus 3 (SAV-3) and tilapia lake virus (TiLV), indicating their usability for work with multiple fish viruses. While confirming the difficulty of replicating PRV-1 and PRV-3, the results demonstrate the potential of novel heart-derived cell cultures as in vitro tools for studying fish viruses.
Collapse
Affiliation(s)
- Torben Krebs
- Fish Disease Research Unit, Centre for Infection MedicineUniversity of Veterinary Medicine HannoverHannoverGermany
| | - Julia Bauer
- Fish Disease Research Unit, Centre for Infection MedicineUniversity of Veterinary Medicine HannoverHannoverGermany
| | - Sarah Graff
- Working Group Fish Health and –Welfare, Section Aquaculture and Aquatic ResourcesFraunhofer Research Institution for Individualized and Cell‐Based Medical Engineering IMTEBüsumGermany
| | - Lukas Teich
- Fish Disease Research Unit, Centre for Infection MedicineUniversity of Veterinary Medicine HannoverHannoverGermany
| | - Markus Sterneberg
- Fish Disease Research Unit, Centre for Infection MedicineUniversity of Veterinary Medicine HannoverHannoverGermany
| | - Marina Gebert
- Working Group Fish Health and –Welfare, Section Aquaculture and Aquatic ResourcesFraunhofer Research Institution for Individualized and Cell‐Based Medical Engineering IMTEBüsumGermany
| | - Henrike Seibel
- Working Group Fish Health and –Welfare, Section Aquaculture and Aquatic ResourcesFraunhofer Research Institution for Individualized and Cell‐Based Medical Engineering IMTEBüsumGermany
| | - Bettina Seeger
- Institute for Food Quality and Food SafetyUniversity of Veterinary Medicine HannoverHannoverGermany
| | - John Hellmann
- Environment and Consumer Protection, Fisheries Ecology and AquacultureNorth Rhine Westphalian State Agency for NatureGermany
| | - Øystein Wessel
- Faculty of Veterinary MedicineNorwegian University of Life SciencesÅsNorway
| | - Espen Rimstad
- Faculty of Veterinary MedicineNorwegian University of Life SciencesÅsNorway
| | - Win Surachetpong
- Department of Veterinary Microbiology and Immunology, Faculty of Veterinary MedicineKasetsart UniversityBangkokThailand
| | - Dieter Steinhagen
- Fish Disease Research Unit, Centre for Infection MedicineUniversity of Veterinary Medicine HannoverHannoverGermany
| | - Verena Jung‐Schroers
- Fish Disease Research Unit, Centre for Infection MedicineUniversity of Veterinary Medicine HannoverHannoverGermany
| | - Mikolaj Adamek
- Fish Disease Research Unit, Centre for Infection MedicineUniversity of Veterinary Medicine HannoverHannoverGermany
| |
Collapse
|
3
|
Elblová P, Anthi J, Liu M, Lunova M, Jirsa M, Stephanopoulos N, Lunov O. DNA Nanostructures for Rational Regulation of Cellular Organelles. JACS AU 2025; 5:1591-1616. [PMID: 40313805 PMCID: PMC12042030 DOI: 10.1021/jacsau.5c00117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/15/2025] [Accepted: 03/20/2025] [Indexed: 05/03/2025]
Abstract
DNA nanotechnology has revolutionized materials science and biomedicine by enabling precise manipulation of matter at the nanoscale. DNA nanostructures (DNs) in particular represent a promising frontier for targeted therapeutics. Engineered DNs offer unprecedented molecular programmability, biocompatibility, and structural versatility, making them ideal candidates for advanced drug delivery, organelle regulation, and cellular function modulation. This Perspective explores the emerging role of DNs in modulating cellular behavior through organelle-targeted interventions. We highlight current advances in nuclear, mitochondrial, and lysosomal targeting, showcasing applications ranging from gene delivery to cancer therapeutics. For instance, DNs have enabled precision mitochondrial disruption in cancer cells, lysosomal pH modulation to enhance gene silencing, and nuclear delivery of gene-editing templates. While DNs hold immense promise for advancing nanomedicine, outstanding challenges include optimizing biological interactions and addressing safety concerns. This Perspective highlights the current potential of DNs for rational control of targeted organelles, which could lead to novel therapeutic strategies and advancement of precision nanomedicines in the future.
Collapse
Affiliation(s)
- Petra Elblová
- FZU
- Institute of Physics of the Czech Academy of Sciences, 182 21 Prague, Czech Republic
- Faculty
of Mathematics and Physics, Charles University, Ke Karlovu 3, 121 16 Prague 2, Czech Republic
| | - Judita Anthi
- FZU
- Institute of Physics of the Czech Academy of Sciences, 182 21 Prague, Czech Republic
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign
Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85281, United States
| | - Minghui Liu
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign
Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85281, United States
| | - Mariia Lunova
- FZU
- Institute of Physics of the Czech Academy of Sciences, 182 21 Prague, Czech Republic
- Institute
for Clinical & Experimental Medicine (IKEM), 14 021 Prague, Czech Republic
| | - Milan Jirsa
- Institute
for Clinical & Experimental Medicine (IKEM), 14 021 Prague, Czech Republic
| | - Nicholas Stephanopoulos
- School
of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign
Center for Molecular Design and Biomimetics, Arizona State University, Tempe, Arizona 85281, United States
| | - Oleg Lunov
- FZU
- Institute of Physics of the Czech Academy of Sciences, 182 21 Prague, Czech Republic
| |
Collapse
|
4
|
Derman ID, Alioglu MA, Moses JC, Chroneos ZC, Yilmaz YO, Banerjee D, Koff J, Rizvi SHA, Klunk DN, Celik N, Pochareddy S, Umstead TM, Namli I, Holton SE, Mikacenic C, Thompson JL, Castaneda DC, Hickey DR, Nagamine M, Warang P, Schotsaert M, Chen P, Peeples ME, Palucka K, Ozbolat IT. A ventilated perfused lung model platform to dissect the response of the lungs to viral infection. Trends Biotechnol 2025:S0167-7799(25)00115-5. [PMID: 40280814 DOI: 10.1016/j.tibtech.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025]
Abstract
In this study, we developed a 3D lung model that incorporated alveolar and vascular components, allowing for the investigation of lung physiology and responses to infection. We investigated the role of ventilation in formation of the alveolar epithelial layer and its response to viral infections. We subjected our perfused model to a continuous respiratory cycle at the air-liquid interface (ALI) for up to 10 days. The results revealed that ventilation increased tight-junction formation with better epithelial barrier function over time. Two viruses, influenza and respiratory syncytial virus (RSV), were tested, where ventilation enhanced infectivity with an increased progression of viral spread over time while sensitizing the epithelium for viral recognition. Ventilation also attenuated the production of key proinflammatory chemokines. Our findings represent a critical step forward in advancing our understanding of lung-specific viral responses and respiratory infections in response to ventilation, shedding light on vital aspects of pulmonary physiology and pathobiology.
Collapse
Affiliation(s)
- I Deniz Derman
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Mecit Altan Alioglu
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Joseph Christakiran Moses
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Zissis C Chroneos
- Department of Pediatrics, Penn State University College of Medicine, Hershey, PA 17033, USA; Department of Cell and Biological Systems, Penn State University College of Medicine, Hershey, PA, USA
| | - Yasar Ozer Yilmaz
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA; Department of Nanoscience and Nanoengineering, Istanbul Technical University, Istanbul, Turkey
| | - Dishary Banerjee
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA; Department of Cardiology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jonathan Koff
- Department of Medicine, Yale University, New Haven, CT 06520, USA
| | - Syed Hasan Askari Rizvi
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Danielle Nicole Klunk
- Biomedical Engineering Department, Penn State University, University Park, PA 16802, USA
| | - Nazmiye Celik
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Sirisha Pochareddy
- Genome Sciences Core, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Todd M Umstead
- Department of Pediatrics, Penn State University College of Medicine, Hershey, PA 17033, USA
| | - Ilayda Namli
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Sarah E Holton
- Translational Immunology Department, Benaroya Research Institute, Seattle, WA 98101, USA; Department of Pulmonary, Critical Care, & Sleep Medicine, University of Washington, Seattle, WA 98195, USA
| | - Carmen Mikacenic
- Translational Immunology Department, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Jessica L Thompson
- Chemistry Department, Penn State University, University Park, PA 16802, USA
| | | | - Danielle Reifsnyder Hickey
- Chemistry Department, Penn State University, University Park, PA 16802, USA; Department of Materials Science and Engineering, Penn State University, University Park, PA 16802, USA; Materials Research Institute, Penn State University, University Park, PA 16802, USA
| | - Momoka Nagamine
- The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA; Chemistry Department, Penn State University, University Park, PA 16802, USA
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Phylip Chen
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | - Mark E Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA; Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Infectious Disease Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Karolina Palucka
- The Jackson laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Ibrahim T Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA; Biomedical Engineering Department, Penn State University, University Park, PA 16802, USA; Materials Research Institute, Penn State University, University Park, PA 16802, USA; Cancer Institute, Penn State University, University Park, PA 16802, USA; Neurosurgery Department, Penn State University, University Park, PA 16802, USA.
| |
Collapse
|
5
|
Meyer A, Khalil B, Iljin M, Bange H, Price LS, Dyubankova N, van Westen GJP, van Vlijmen H, Peters DJM, Artursson P. A Patient-Derived 3D Cyst Model of Polycystic Kidney Disease That Mimics Disease Development and Responds to Repurposing Candidates. Clin Transl Sci 2025; 18:e70214. [PMID: 40235151 PMCID: PMC12000233 DOI: 10.1111/cts.70214] [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: 10/30/2024] [Revised: 02/22/2025] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disease. Its progressively expanding, fluid-filled renal cysts eventually lead to end-stage renal disease. Despite the relatively high prevalence, treatment options are currently limited to a single drug approved by the FDA and EMA. Here, we investigated human ADPKD patient-derived three-dimensional cyst cultures (3DCC) as an in vitro model for ADPKD and drug repurposing research. First, we analyzed the proteomes of 3DCC derived from healthy and diseased tissues. We then compared the protein expression profiles with those of reference tissues, mainly from the same patients. We quantified 290 proteins affecting drug disposition and proposed target proteins for drug treatment. Lastly, we investigated the functional response of the quantified target proteins after exposure to repurposing candidates in the 3DCC. Proteomic profiling of human 3DCC reflected previously reported pathophysiological alterations, including aberrant protein expression in inflammation and metabolic reprogramming. While the 3DCCs largely recapitulated the disease phenotype in vitro, drug transporter expression was reduced compared to in vivo conditions. Target proteins for proposed repurposing candidates showed similar expression in vitro and in tissues. Exposure to these repurposing candidates inhibited cyst swelling in vitro, supporting the suitability of the 3DCC for ADPKD drug screening. In summary, our results provide new insights into the ADPKD proteome and offer a starting point for further research to improve treatment options for affected individuals.
Collapse
Affiliation(s)
- Alina Meyer
- Department of PharmacyUppsala UniversityUppsalaSweden
| | - Bola Khalil
- In Silico Discovery, J&J Innovative MedicineBeerseBelgium
- Division of Medicinal ChemistryLeiden Academic Centre for Drug ResearchLeidenthe Netherlands
| | | | - Hester Bange
- Crown Bioscience Netherlands B.VLeidenthe Netherlands
| | - Leo S. Price
- Crown Bioscience Netherlands B.VLeidenthe Netherlands
| | | | - Gerard J. P. van Westen
- Division of Medicinal ChemistryLeiden Academic Centre for Drug ResearchLeidenthe Netherlands
| | | | - Dorien J. M. Peters
- Department of Human GeneticsLeiden University Medical CenterLeidenthe Netherlands
| | - Per Artursson
- Department of PharmacyUppsala UniversityUppsalaSweden
| |
Collapse
|
6
|
Lindsay S, Hussain M, Binici B, Perrie Y. Exploring the Challenges of Lipid Nanoparticle Development: The In Vitro-In Vivo Correlation Gap. Vaccines (Basel) 2025; 13:339. [PMID: 40333261 PMCID: PMC12031360 DOI: 10.3390/vaccines13040339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND/OBJECTIVES The development of lipid nanoparticles (LNPs) as delivery platforms for nucleic acids has revolutionised possibilities for both therapeutic and vaccine applications. However, emerging studies highlight challenges in achieving reliable in vitro-in vivo correlation (IVIVC), which delays the translation of experimental findings into clinical applications. This study investigates these potential discrepancies by evaluating the physicochemical properties, in vitro efficacy (across three commonly used cell lines), and in vivo performance (mRNA expression and vaccine efficacy) of four LNP formulations. METHODS LNPs composed of DSPC, cholesterol, a PEGylated lipid, and one of four ionizable lipids (SM-102, ALC-0315, MC3, or C12-200) were manufactured using microfluidics. RESULTS All formulations exhibited comparable physicochemical properties, as expected (size 70-100 nm, low PDI, near-neutral zeta potential, and high mRNA encapsulation). In vitro studies demonstrated variable LNP-mediated mRNA expression in both immortalised and immune cells, with SM-102 inducing significantly higher protein expression (p < 0.05) than the other formulations in immortalised and immune cells. However, in vivo results revealed that ALC-0315 and SM-102-based LNPs achieved significantly (p < 0.05) higher protein expression without a significant difference between them, while MC3- and C12-200-based LNPs exhibited lower expression levels. As vaccine formulations, all LNPs elicited strong immune responses with no significant differences among them. CONCLUSIONS These findings highlight the complexities of correlating in vitro and in vivo outcomes in LNP development and demonstrate the importance of holistic evaluation strategies to optimise their clinical translation.
Collapse
Affiliation(s)
| | | | | | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (S.L.); (M.H.); (B.B.)
| |
Collapse
|
7
|
Jin Y, Liang Y, Wu B, Wu S, Liu X, Zhou F. Establishment of CD34 + hematopoietic stem cell-derived xenograft model of hyperleukocytic acute myeloid leukemia. BMC Cancer 2025; 25:499. [PMID: 40102796 PMCID: PMC11917077 DOI: 10.1186/s12885-025-13907-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Hyperleukocytic acute myeloid leukemia (HLL) is marked by high early mortality and presents significant therapeutic challenges. Research on HLL is still in its infancy, and comprehensive development of patient-derived xenograft (PDX) models, especially CD34 + hematopoietic stem cell-derived models, remains limited. METHODS We evaluated the establishment of the HLL model through blood examinations, smear analysis, bone marrow biopsy, flow cytometry, and mutation analysis. Correlation between survival times in mice and patients was assessed using linear regression. RESULTS In the HLL PDX mouse model, leukocyte counts could reach up to 37.35^10⁹/L, and immunophenotyping revealed the presence of hCD45+, hCD15+, and hCD33 + cells in both peripheral blood (PB) and bone marrow (BM) following inoculation with PB-derived cells for the establishment of the HLL PDX model. Similar results were observed with cells derived from the patient's BM. In the CD34 + hematopoietic stem cell-derived xenograft model, extensive infiltration of CD34 + cells into the BM, liver, and spleen was observed. Additionally, human WT1 and NRAS mutations were identified in the liver, spleen, and BM of the mice. A comparative analysis of multiple experiments revealed that shorter survival times were observed in mice receiving a higher irradiation dose of 2.5 Gy and a greater number of cells derived from PB. Additionally, shorter survival times were observed in model mice injected with cells carrying NRAS, DNMT3A, FLT3, or NPM1 gene mutations. Correlation analysis indicated that the survival times of the mice were significantly associated with the survival status of the patients. CONCLUSIONS We successfully established a CD34 + hematopoietic stem cell-derived xenograft model of HLL, providing a valuable tool for mechanistic research, drug screening, individualized therapy, and precision medicine. TRIAL REGISTRATION Not application.
Collapse
Affiliation(s)
- Yanxia Jin
- Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China
- College of Life Sciences, Hubei Normal University, Huangshi, 435002, Hubei, China
| | - Yuxing Liang
- Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China
| | - Balu Wu
- Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China
| | - Sanyun Wu
- Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China
| | - Xiaoyan Liu
- Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China.
| | - Fuling Zhou
- Department of Haematology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, Hubei, China.
- Research Center for Lifeorgdivision Health, Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
8
|
Eliasen JN, Kristiansen U, Kohlmeier KA. Dimethyltryptamine (DMT) and ibogaine elicit membrane effects in HEK cells transiently transfected with the human 5-HT2A receptor. Brain Res 2025; 1850:149425. [PMID: 39732157 DOI: 10.1016/j.brainres.2024.149425] [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: 11/08/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Psychedelics show promise in treating psychiatric disorders. Therapeutic effects appear to involve activation of the 5-Hydroxytryptamine 2A receptor (5-HT2AR), a G protein-coupled receptor (GPCR). Several SNPs of the 5-HT2AR naturally occur, which are associated with differences in receptor function and altered responsiveness to treatments. New compounds suspected to act at the 5-HT2AR are actively being generated. HEK cells are not commonly used to study membrane effects induced by agonists of GPCRs. In this study, for the first time, membrane actions of two psychedelics, dimethyltryptamine (DMT) and ibogaine on HEK cells transiently transfected with either the human wildtype (WT) or the human I197V mutated 5-HT2AR were investigated using whole-cell electrophysiology. Membrane effects were observed in both genotypes and with both drugs in most cells, while no responses were observed in non-transfected HEK cells suggesting that responses were due to 5-HT2AR activation. In HEK cells transfected with the I197V SNP, a significantly shorter duration of the DMT response was observed, however there were no differences in drug-elicited amplitudes between drug or receptor genotype. I-V curves showed a significant effect of drug exposure for both DMT and ibogaine at the highest concentration evaluated. Taken together, our data show transfection of the 5-HT2AR, a GPCR, in HEK cells is able to activate downstream ion channels following exposure to two different 5-HT2AR agonists. Accordingly, investigations of novel compounds suspected to act at 5-HT2ARs can include examination of elicitation of ionic currents in 5-HT2AR transfected HEK cells, and drug effects at SNPs can also be evaluated.
Collapse
Affiliation(s)
- Jannik Nicklas Eliasen
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Uffe Kristiansen
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark
| | - Kristi A Kohlmeier
- Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2100, Denmark.
| |
Collapse
|
9
|
Kim J, Ślęczkowska M, Nobre B, Wieringa P. Study Models for Chlamydia trachomatis Infection of the Female Reproductive Tract. Microorganisms 2025; 13:553. [PMID: 40142446 PMCID: PMC11945960 DOI: 10.3390/microorganisms13030553] [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: 12/23/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
Chlamydia trachomatis (Ct) is a leading cause of sexually transmitted infections globally, often resulting in inflammatory disorders, ectopic pregnancies, and infertility. Studying Ct's pathogenesis remains challenging due to its unique life cycle and host-specific interactions, which require diverse experimental models. Animal studies using mouse, guinea pig, pig, and non-human primate models provide valuable insights into immune responses, hormonal influences, and disease progression. However, they face limitations in terms of translational relevance due to physiological differences, as well as ethical concerns. Complementing these, in vitro systems, ranging from simple monolayer to advanced three-dimensional models, exhibit improved physiological relevance by replicating the human tissue architecture. This includes the detailed investigation of epithelial barrier disruptions, epithelium-stroma interactions, and immune responses at a cellular level. Nonetheless, in vitro models fall short in mimicking the intricate tissue structures found in vivo and, therefore, cannot faithfully replicate the host-pathogen interactions or infection dynamics observed in living organisms. This review presents a comprehensive overview of the in vivo and in vitro models employed over the past few decades to investigate Ct and its pathogenesis, addressing their strengths and limitations. Furthermore, we explore emerging technologies, including organ-on-chip and in silico models, as promising tools to overcome the existing challenges and refine our understanding of Ct infections.
Collapse
Affiliation(s)
| | | | | | - Paul Wieringa
- Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.K.); (M.Ś.); (B.N.)
| |
Collapse
|
10
|
Ilg MM, Lapthorn AR, Harding SL, Minhas T, Koduri G, Bustin SA, Cellek S. Development of a phenotypic screening assay to measure activation of cancer-associated fibroblasts. Front Pharmacol 2025; 16:1526495. [PMID: 40017592 PMCID: PMC11865240 DOI: 10.3389/fphar.2025.1526495] [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: 11/11/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
Background In cancer metastasis, tumor cells condition distant tissues to create a supportive environment, or metastatic niche, by driving the activation of cancer-associated fibroblasts (CAFs). These CAFs remodel the extracellular matrix, creating a microenvironment that supports tumor growth and compromises immune cell function, enabling cancer cells to evade immune detection. Consequently, targeting the activation of CAFs has been proposed as a therapeutic strategy to hinder metastatic spread. Our objective was to develop the first in vitro phenotypic screening assay capable of assessing this activation process. Methods Human primary lung fibroblasts were co-cultured with highly invasive breast cancer cells (MDA-MB-231) to identify changes in the expression of selected genes using RT-qPCR. An In-Cell ELISA (ICE)-based assay using human lung fibroblasts, MDA-MB-231 cells and human monocytes (THP-1 cells) was developed to measure the activation of CAFs. Another ELISA assay was used to measure released osteopontin. Results When lung fibroblast were co-cultured with MDA-MB-231 cells, among the 10 selected genes, the genes for osteopontin (SPP1), insulin like growth factor 1 (IGF1), periostin (POSTN) and α-smooth muscle actin (α-SMA, ACTA2) elicited the greatest fold change (55-, 37-, 8- and 5-fold respectively). Since osteopontin, IGF-1 and periostin are secreted proteins and α-SMA is an intracellular cytoskeleton protein, α-SMA was chosen to be the readout biomarker for the ICE assay. When fibroblasts were co-cultured with MDA-MB-231 cells and monocytes in the 96 well ICE assay, α-SMA expression was increased 2.3-fold yielding a robust Z' of 0.56. A secondary, low throughput assay was developed by measuring the release of osteopontin which showed a 6-fold increase when fibroblasts were co-cultured with MDA-MB-231 cells and monocytes. Discussion This phenotypic assay is the first to measure the activation of CAFs in a 96-well format, making it suitable for medium-to high-throughput screening of potential therapeutic compounds. By focusing on observable cellular phenotypic changes rather than targeting specific molecular pathways, this assay allows for a broader and unbiased identification of compounds capable of modulating CAF activation.
Collapse
Affiliation(s)
- Marcus M. Ilg
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Alice R. Lapthorn
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Sophie L. Harding
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Tariq Minhas
- The Essex Cardiothoracic Centre, Basildon University Hospital, Basildon, United Kingdom
| | - Gouri Koduri
- Southend University Hospital NHS Foundation Trust, Westcliff-on-Sea, United Kingdom
| | - Stephen A. Bustin
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Selim Cellek
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| |
Collapse
|
11
|
Mazzaglia C, Shery Huang YY, Shields JD. Advancing tumor microenvironment and lymphoid tissue research through 3D bioprinting and biofabrication. Adv Drug Deliv Rev 2025; 217:115485. [PMID: 39653084 DOI: 10.1016/j.addr.2024.115485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
Cancer progression is significantly influenced by the complex interactions within the tumor microenvironment (TME). Immune cells, in particular, play a critical role by infiltrating tumors from the circulation and surrounding lymphoid tissues in an attempt to control their spread. However, they often fail in this task. Current in vivo and in vitro preclinical models struggle to fully capture these intricate interactions affecting our ability to understand immune evasion and predict drugs behaviour in the clinic. To address this challenge, biofabrication and particularly 3D bioprinting has emerged as a promising tool for modeling both tumors and the immune system. Its ability to incorporate multiple cell types into 3D matrices, enable tissue compartmentalization with high spatial accuracy, and integrate vasculature makes it a valuable approach. Nevertheless, limited research has focused on capturing the complex tumor-immune interplay in vitro. This review highlights the composition and significance of the TME, the architecture and function of lymphoid tissues, and innovative approaches to modeling their interactions in vitro, while proposing the concept of an extended TME.
Collapse
Affiliation(s)
- Corrado Mazzaglia
- The Nanoscience Centre, University of Cambridge, Cambridge, the United Kingdom of Great Britain and Northern Ireland; Department of Engineering, University of Cambridge, Cambridge, the United Kingdom of Great Britain and Northern Ireland; Center for Life Nano, and Neuro-Science of Istituto Italiano di Tecnologia (IIT), Rome 00161, Italy.
| | - Yan Yan Shery Huang
- The Nanoscience Centre, University of Cambridge, Cambridge, the United Kingdom of Great Britain and Northern Ireland; Department of Engineering, University of Cambridge, Cambridge, the United Kingdom of Great Britain and Northern Ireland
| | - Jacqueline D Shields
- Translational Medical Sciences, School of Medicine, University of Nottingham, Biodiscovery Institute, Nottingham, the United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
12
|
Kala S, Strutz AG, Katt ME. The Rise of Pluripotent Stem Cell-Derived Glia Models of Neuroinflammation. Neurol Int 2025; 17:6. [PMID: 39852770 PMCID: PMC11767680 DOI: 10.3390/neurolint17010006] [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: 11/20/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/26/2025] Open
Abstract
Neuroinflammation is a blanket term that describes the body's complex inflammatory response in the central nervous system (CNS). It encompasses a phenotype shift to a proinflammatory state, the release of cytokines, the recruitment of peripheral immune cells, and a wide variety of other processes. Neuroinflammation has been implicated in nearly every major CNS disease ranging from Alzheimer's disease to brain cancer. Understanding and modeling neuroinflammation is critical for the identification of novel therapeutic targets in the treatment of CNS diseases. Unfortunately, the translation of findings from non-human models has left much to be desired. This review systematically discusses the role of human pluripotent stem cell (hPSC)-derived glia and supporting cells within the CNS, including astrocytes, microglia, oligodendrocyte precursor cells, pericytes, and endothelial cells, to describe the state of the field and hope for future discoveries. hPSC-derived cells offer an expanded potential to study the pathobiology of neuroinflammation and immunomodulatory cascades that impact disease progression. While much progress has been made in the development of models, there is much left to explore in the application of these models to understand the complex inflammatory response in the CNS.
Collapse
Affiliation(s)
- Srishti Kala
- Cancer Cell Biology Graduate Education Program, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| | - Andrew G. Strutz
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| | - Moriah E. Katt
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA
| |
Collapse
|
13
|
Sriram G, Makkar H. Microfluidic organ-on-chip systems for periodontal research: advances and future directions. Front Bioeng Biotechnol 2025; 12:1490453. [PMID: 39840127 PMCID: PMC11747509 DOI: 10.3389/fbioe.2024.1490453] [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: 09/03/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Advances in tissue engineering and microfluidic technologies have enabled the development of sophisticated in vitro models known as organ-on-a-chip (OoC) or microphysiological systems. These systems enable to potential to simulate the dynamic interactions between host tissues and their microenvironment including microbes, biomaterials, mechanical forces, pharmaceutical, and consumer-care products. These fluidic technologies are increasingly being utilized to investigate host-microbe and host-material interactions in oral health and disease. Of interest is their application in understanding periodontal disease, a chronic inflammatory condition marked by the progressive destruction of periodontal tissues, including gingiva, periodontal ligament, and alveolar bone. The pathogenesis of periodontal disease involves a complex interplay between microbial dysbiosis and host immune responses, which can lead to a loss of dental support structures and contribute to systemic conditions such as cardiovascular disease, diabetes, and inflammatory bowel disease. This provides a comprehensive overview of the latest developments in millifluidic and microfluidic systems designed to emulate periodontal host-microbe and host-material interactions. We discuss the critical engineering and biological considerations in designing these platforms, their applications in studying oral biofilms, periodontal tissue responses, and their potential to unravel disease mechanisms and therapeutic targets in periodontal disease.
Collapse
Affiliation(s)
- Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Hardik Makkar
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
- Center for Innovation & Precision Dentistry, School of Dental Medicine and School of Engineering & Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
14
|
Rani S, Ramesh V, Khatoon M, Shijili M, Archana CA, Anand J, Sagar N, Sekar YS, Patil AV, Palavesam A, Barman NN, Patil SS, Hemadri D, Suresh KP. Identification of molecular and cellular infection response biomarkers associated with anthrax infection through comparative analysis of gene expression data. Comput Biol Med 2025; 184:109431. [PMID: 39556915 DOI: 10.1016/j.compbiomed.2024.109431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/16/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024]
Abstract
Bacillus anthracis, a gram-positive bacillus capable of forming spores, causes anthrax in mammals, including humans, and is recognized as a potential biological weapon agent. The diagnosis of anthrax is challenging due to variable symptoms resulting from exposure and infection severity. Despite the availability of a licensed vaccines, their limited long-term efficacy underscores the inadequacy of current human anthrax vaccines, highlighting the urgent need for next-generation alternatives. Our study aimed to identify molecular biomarkers and essential biological pathways for the early detection and accurate diagnosis of human anthrax infection. Using a comparative analysis of Bacillus anthracis gene expression data from the Gene Expression Omnibus (GEO) database, this cost-effective approach enables the identification of shared differentially expressed genes (DEGs) across separate microarray datasets without additional hybridization. Three microarray datasets (GSE34407, GSE14390, and GSE12131) of B. anthracis-infected human cell lines were analyzed via the GEO2R tool to identify shared DEGs. We identified 241 common DEGs (70 upregulated and 171 downregulated) from cell lines treated similarly to lethal toxins. Additionally, 10 common DEGs (5 upregulated and 5 downregulated) were identified across different treatments (lethal toxins and spores) and cell lines. Network meta-analysis identified JUN and GATAD2A as the top hub genes for overexpression, and NEDD4L and GULP1 for underexpression. Furthermore, prognostic analysis and SNP detection of the two identified upregulated hub genes were carried out in conjunction with machine learning classification models, with SVM yielding the best classification accuracy of 87.5 %. Our comparative analysis of Bacillus anthracis infection revealed striking similarities in gene expression 241 profiles across diverse datasets, despite variations in treatments and cell lines. These findings underscore how anthrax infection activates shared genes across different cell types, emphasizing this approach in the discovery of novel gene markers. These markers offer insights into pathogenesis and may lead to more effective therapeutic strategies. By identifying these genetic indicators, we can advance the development of precise immunotherapies, potentially enhancing vaccine efficacy and treatment outcomes.
Collapse
Affiliation(s)
- Swati Rani
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, 560064, India
| | - Varsha Ramesh
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, 560064, India
| | - Mehnaj Khatoon
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, 560064, India
| | - M Shijili
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, 560064, India
| | - C A Archana
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, 560064, India
| | - Jayashree Anand
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, 560064, India
| | - N Sagar
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, 560064, India
| | - Yamini S Sekar
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, 560064, India
| | - Archana V Patil
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, 560064, India
| | - Azhahianambi Palavesam
- Translational Research Platform for Veterinary Biologicals, Centre for Animal Health Studies, Tamil Nadu Veterinary and Animal Sciences University, Chennai, Tamil Nadu, 600051, India
| | - N N Barman
- College of Veterinary Science, Assam Agricultural University, Guwahati, Assam, 781001, India
| | - S S Patil
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, 560064, India
| | - Diwakar Hemadri
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, 560064, India
| | - K P Suresh
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru, Karnataka, 560064, India.
| |
Collapse
|
15
|
Koceva H, Mosig A. Human-Induced Pluripotent Stem Cell-Based Alveolus-on-Chip Model to Study Influenza Virus A Infection. Methods Mol Biol 2025; 2890:225-235. [PMID: 39890730 DOI: 10.1007/978-1-0716-4326-6_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
This chapter introduces an alveolus-on-chip model for studying influenza virus A infection. The model represents a physiologically relevant alternative to animal models and a scalable and cost-effective approach to gaining mechanistic insights into human-related viral infection processes in vitro. We provide a comprehensive protocol for creating a human autologous model from human-induced pluripotent stem cell (hiPSC)-derived alveolar type II cells, endothelial cells, and macrophages. The protocol includes details on on-chip cell culture, microfluidic perfusion, and monitoring influenza A virus infection through immunofluorescence imaging.
Collapse
Affiliation(s)
- Hristina Koceva
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany
| | - Alexander Mosig
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany.
- Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.
| |
Collapse
|
16
|
Periasamy A, Ornelas P, Bausewein T, Mitchell N, Zhao J, Quinn LM, Kuehlbrandt W, Gulbis JM. Structure of an ex vivoDrosophila TOM complex determined by single-particle cryoEM. IUCRJ 2025; 12:49-61. [PMID: 39575538 PMCID: PMC11707698 DOI: 10.1107/s2052252524011011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/13/2024] [Indexed: 01/11/2025]
Abstract
Most mitochondrial precursor proteins are encoded in the cell nucleus and synthesized on cytoplasmic ribosomes. The translocase of the outer membrane (TOM) is the main protein-import pore of mitochondria, recognizing nascent precursors of mitochondrially targeted proteins and transferring them across the outer membrane. A 3.3 Å resolution map and molecular model of a TOM complex from Drosophila melanogaster, obtained by single-particle electron cryomicroscopy, is presented. As the first reported structure of a transgenic protein expressed and purified ex vivo from Drosophila, the method provides impetus for parallel structural and genetic analyses of protein complexes linked to human pathology. The core TOM complex extracted from native membranes of the D. melanogaster retina contains transgenic Tom40 co-assembled with four endogenous TOM components: Tom22, Tom5, Tom6 and Tom7. The Drosophila TOM structure presented here shows that the human and Drosophila TOM are very similar, with small conformational changes at two subunit interfaces attributable to variation in lipid-binding residues. The new structure provides an opportunity to pinpoint general features that differentiate the TOM structures of higher and unicellular eukaryotes. While the quaternary fold of the assembly is retained, local nuances of structural elements implicated in precursor import are indicative of subtle evolutionary change.
Collapse
Affiliation(s)
- Agalya Periasamy
- Structural Biology DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoria3052Australia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoria3052Australia
| | - Pamela Ornelas
- Department of Structural BiologyMax Planck Institute of BiophysicsMax-von-Laue-Strasse 360438Frankfurt am MainGermany
| | - Thomas Bausewein
- Department of Structural BiologyMax Planck Institute of BiophysicsMax-von-Laue-Strasse 360438Frankfurt am MainGermany
| | - Naomi Mitchell
- Department of Cancer Biology and Therapeutics, John Curtin School of Medical ResearchAustralian National University (ANU)CanberraACTAustralia
| | - Jiamin Zhao
- Structural Biology DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoria3052Australia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoria3052Australia
| | - Leonie M. Quinn
- Department of Cancer Biology and Therapeutics, John Curtin School of Medical ResearchAustralian National University (ANU)CanberraACTAustralia
| | - Werner Kuehlbrandt
- Department of Structural BiologyMax Planck Institute of BiophysicsMax-von-Laue-Strasse 360438Frankfurt am MainGermany
| | - Jacqueline M. Gulbis
- Structural Biology DivisionThe Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoria3052Australia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoria3052Australia
| |
Collapse
|
17
|
Gudgeon J, Dannoura A, Chatterjee R, Sidgwick F, Raymond BB, Frey AM, Marin-Rubio JL, Trost M. Mass spectrometry-based proteomic exploration of diverse murine macrophage cellular models. Life Sci Alliance 2025; 8:e202402760. [PMID: 39510801 PMCID: PMC11544424 DOI: 10.26508/lsa.202402760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024] Open
Abstract
Immortalised cell lines that mimic their primary cell counterparts are fundamental to research, particularly when large cell numbers are required. Here, we report that immortalisation of bone marrow-derived macrophages (iBMDMs) using the J2 virus resulted in the loss of a protein of interest, MSR1, in WT cells by an unknown mechanism. This led us to perform an in-depth mass spectrometry-based proteomic characterisation of common murine macrophage cell lines (J774A.1, RAW264.7, and BMA3.1A7), in comparison with the iBMDMs, as well as primary BMDMs from both C57BL/6 and BALB/c mice. This analysis revealed striking differences in protein profiles associated with macrophage polarisation, phagocytosis, pathogen recognition, and interferon signalling. Among the cell lines, J774A.1 cells were the most similar to the gold standard primary BMDM model, whereas BMA3.1A7 cells were the least similar because of the reduction in abundance of several key proteins related closely to macrophage function. This comprehensive proteomic dataset offers valuable insights into the use and suitability of macrophage cell lines for cell signalling and inflammation research.
Collapse
Affiliation(s)
- Jack Gudgeon
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Abeer Dannoura
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ritika Chatterjee
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Frances Sidgwick
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Andrew M Frey
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Matthias Trost
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
18
|
Pant A, Wendel SO, Wallace NA, Yang Z. Sample Preparation for Global Metabolic Profiling of Vaccinia Virus-Infected Primary Human Foreskin Fibroblasts. Methods Mol Biol 2025; 2860:273-285. [PMID: 39621274 DOI: 10.1007/978-1-0716-4160-6_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Vaccinia virus (VACV), the prototype member of the Poxviridae family, has played a crucial role in medicine as a key component in the development of smallpox vaccines, contributing to the eradication of this deadly disease. Beyond its historical significance, VACV continues to be pivotal in researching metabolic alterations induced by viral infections. Studies have revealed that VACV can impact pathways such as glycolysis, the tricarboxylic acid (TCA) cycle, and lipid metabolism in host cells, offering valuable insights into host-virus interactions and broader cellular metabolism. The preference for primary cells, such as human foreskin fibroblasts (HFFs), over cancer cells in metabolic studies is justified for their physiological relevance, representing native cell types with genetic stability. Metabolic profiling is an ideal tool for studying virus-induced metabolic alterations, providing a comprehensive analysis of changes in cellular metabolism triggered by viral infections. This chapter outlines a protocol for extracting HFFs, culturing, infecting them with VACV, and conducting untargeted global metabolic profiling to elucidate detailed metabolic statuses of the infected cells. This protocol may be modified for understanding the intricacies of host-virus interactions at the metabolic interface for other poxviruses and non-poxviruses.
Collapse
Affiliation(s)
- Anil Pant
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | | | | | - Zhilong Yang
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
19
|
Deng Z, Iwasaki K, Peng Y, Honda Y. Mesenchymal Stem Cell Extract Promotes Skin Wound Healing. Int J Mol Sci 2024; 25:13745. [PMID: 39769505 PMCID: PMC11679360 DOI: 10.3390/ijms252413745] [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: 11/05/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Recently, it has been reported that mesenchymal stem cell (MSC)-derived humoral factors promote skin wound healing. As these humoral factors are transiently stored in cytoplasm, we collected them as part of the cell extracts from MSCs (MSC-ext). This study aimed to investigate the effects of MSC-ext on skin wound healing. We examined the effects of MSC-ext on cell proliferation and migration. Additionally, the effect of MSC-ext on skin wound healing was evaluated using a mouse skin defect model. The MSC-ext enhanced the proliferation of dermal fibroblasts, epithelial cells, and endothelial cells. It also increased the number of migrating fibroblasts and epithelial cells. The skin defects treated with MSC-ext demonstrated rapid wound closure compared to those treated with phosphate-buffered saline. The MSC-ext group exhibited a thicker dermis, larger Picrosirius red-positive areas, and a higher number of Ki67-positive cells. Our results indicate that MSC-ext promotes the proliferation and/or migration of fibroblasts, epithelial cells, and endothelial cells, and enhances skin wound healing. This suggests the therapeutic potential of MSC-ext in treating skin defects as a novel cell-free treatment modality.
Collapse
Affiliation(s)
- Zi Deng
- Department of Oral Anatomy, Osaka Dental University, Osaka 573-1121, Japan; (Z.D.); (Y.H.)
| | - Kengo Iwasaki
- Advanced Medicine Research Center, Translational Research Institute for Medical Innovation (TRIMI), Osaka Dental University, Osaka 573-1121, Japan
| | - Yihao Peng
- Department of Periodontology, Osaka Dental University, Osaka 573-1121, Japan;
| | - Yoshitomo Honda
- Department of Oral Anatomy, Osaka Dental University, Osaka 573-1121, Japan; (Z.D.); (Y.H.)
| |
Collapse
|
20
|
Cherkaoui I, Du Q, Egli DM, Dion C, Leitch HG, Sachedina D, Misra S, Rutter GA. Investigating the pathogenicity of the recessive HNF1A p.A251T variant in monogenic diabetes using iPSC-derived beta-like cells. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.10.24318788. [PMID: 39711726 PMCID: PMC11661423 DOI: 10.1101/2024.12.10.24318788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Monogenic diabetes, formerly called Maturity-Onset Diabetes of the Young (MODY), involves single-gene mutations, typically with dominant inheritance, and has been associated with variants in 14 genes. Among these, HNF1A mutations are the most common, and their diagnosis allows the use of alternative therapies, including sulfonylureas. In an earlier study, we described a variant displaying recessive transmission, p.A251T (Misra, S et al, Diabetes Care, 2020). Initial functional studies revealed only a modest impact on protein function. We extend these earlier in vitro studies to demonstrate that beta-like cells derived from pluripotent stem cells from variant carriers show impaired differentiation into insulin-positive cells, whereas differentiation into alpha cells is significantly enhanced. Additionally, mutant cells showed impaired glucose-stimulated insulin secretion but partially preserved responsiveness to treatment with sulfonylureas. Our study provides proof of principle for the utility of using patient-derived stem cells as a platform to assess the pathogenicity of HNF1A variants, and to explore potential treatment strategies.
Collapse
Affiliation(s)
- Ines Cherkaoui
- Centre de Recherche du CHUM, and Faculty of Medicine, University of Montreal, QC, Canada
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, London, UK
| | - Qian Du
- Departments of Pediatrics, Naomi Berrie Diabetes Center, Irving Medical Center, Columbia University, New York, USA
| | - Dieter M. Egli
- Departments of Pediatrics, Naomi Berrie Diabetes Center, Irving Medical Center, Columbia University, New York, USA
| | - Camille Dion
- MRC Laboratory of Medical Sciences, West London, UK
| | - Harry G. Leitch
- MRC Laboratory of Medical Sciences, West London, UK
- Genetics & Genomic Medicine Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Dilshad Sachedina
- Department of Diabetes, Imperial College Healthcare NHS Trust, London, UK
| | - Shivani Misra
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, London, UK
- Department of Diabetes, Imperial College Healthcare NHS Trust, London, UK
| | - Guy A. Rutter
- Centre de Recherche du CHUM, and Faculty of Medicine, University of Montreal, QC, Canada
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, London, UK
- Lee Kong Chian Imperial Medical School, Nanyang Technological University, Singapore
- Research Institute of the McGill University Hospital Centre, Montréal, QC, Canada
| |
Collapse
|
21
|
Awad N, Weidinger D, Greune L, Kronsbein J, Heinen N, Westhoven S, Pfaender S, Taube C, Reuter S, Peters M, Hatt H, Fender A, Knobloch J. Functional characterization of OR51B5 and OR1G1 in human lung epithelial cells as potential drug targets for non-type 2 lung diseases. Cell Biol Toxicol 2024; 40:96. [PMID: 39538061 PMCID: PMC11561009 DOI: 10.1007/s10565-024-09935-9] [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: 03/27/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Hypersensitivity to odorants like perfumes can induce or promote asthma with non-type 2 inflammation for which therapeutic options are limited. Cell death of primary bronchial epithelial cells (PBECs) and the release of the pro-inflammatory cytokines interleukin-6 (IL-6) and IL-8 are key in the pathogenesis. Extra-nasal olfactory receptors (ORs) can influence cellular processes involved in asthma. This study investigated the utility of ORs in epithelial cells as potential drug targets in this context. METHODS We used the A549 cell line and primary bronchial epithelial cells using air-liquid interface culture system (ALI-PBECs). OR expression was investigated by RT-PCR, Western blot, and Immunofluorescence. Effects of OR activation by specific ligands on intracellular calcium concentration, cAMP, Phospholipase C (PLC), cell viability, and IL-6 and IL-8 secretion were analyzed by calcium imaging, enzyme immunoassays, Annexin V/ propidium iodide -based fluorescence-activated cell staining or by ELISA, respectively. RESULTS By screening A549 cells, the OR51B5 agonists Farnesol and Isononyl Alcohol and the OR1G1 agonist Nonanal increased intracellular Ca2 + . OR51B5 and OR1G1 mRNAs and proteins were detected. Both receptors showed a preferential intracellular localization. OR51B5- but not OR1G1-induced Ca2 + dependent on both cAMP and PLC signaling. Farnesol, Isononyl Alcohol, and Nonanal, all reduced cell viability and induced IL-8 and IL-6 release. The data were verified in ALI-PBECs. CONCLUSION ORs in the lung epithelium might be involved in airway-sensitivity to odorants. Their antagonism could represent a promising strategy in treatment of odorant-induced asthma with non-type 2 inflammation.
Collapse
Affiliation(s)
- Noha Awad
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-La-Camp-Platz 1, 44789, Bochum, Germany
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Daniel Weidinger
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-La-Camp-Platz 1, 44789, Bochum, Germany
| | - Lea Greune
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-La-Camp-Platz 1, 44789, Bochum, Germany
| | - Juliane Kronsbein
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-La-Camp-Platz 1, 44789, Bochum, Germany
| | - Natalie Heinen
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Universitätsstraße 150, 44801, Bochum, Germany
| | - Saskia Westhoven
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Universitätsstraße 150, 44801, Bochum, Germany
- Research Unit: Emerging Viruses, Leibniz Institute of Virology (N63), Martinistraße 52, 20251, Hamburg, Germany
| | - Stephanie Pfaender
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Universitätsstraße 150, 44801, Bochum, Germany
- Research Unit: Emerging Viruses, Leibniz Institute of Virology (N63), Martinistraße 52, 20251, Hamburg, Germany
- Institute of Virology and Cell Biology, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Medical Center Essen - Ruhrlandklinik, Tüschener Weg 40, 45239, Essen, Germany
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Medical Center Essen - Ruhrlandklinik, Tüschener Weg 40, 45239, Essen, Germany
- Department of Pneumology, Mainz University Medical Center and Mainz Center for Pulmonary Medicine, Mainz, Germany
| | - Marcus Peters
- Department of Molecular Immunology, Ruhr-University Bochum, Universitätsstraße 150, 44801, Bochum, Germany
| | - Hanns Hatt
- Department of Cell Physiology ND4/35, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Universitaetsstraße 150, 44801, Bochum, Germany
| | - Anke Fender
- Institute of Pharmacology, University Hospital Essen, University Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany
| | - Jürgen Knobloch
- Medical Clinic III for Pneumology, Allergology and Sleep Medicine, Bergmannsheil University Hospital, Ruhr-University Bochum, Bürkle-de-La-Camp-Platz 1, 44789, Bochum, Germany.
| |
Collapse
|
22
|
Mansour F, Parisi L, Rihs S, Schnyder I, La Scala GC, Aliu N, Katsaros C, Degen M. Immortalization of patient-derived lip cells for establishing 3D lip models. Front Cell Dev Biol 2024; 12:1449224. [PMID: 39559739 PMCID: PMC11570282 DOI: 10.3389/fcell.2024.1449224] [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: 06/14/2024] [Accepted: 08/29/2024] [Indexed: 11/20/2024] Open
Abstract
Introduction The lips fulfill various critical physiological roles besides being viewed as a fundamental aesthetic feature contributing to the perception of health and beauty. Therefore, any lip injury, abnormality, or congenital malformation, such as cleft lip, needs special attention in order to restore proper lip function and aesthetics. To achieve this goal, a better understanding of the complex lip anatomy, function, and biology is required, which can only be provided by basic research endeavors. However, the current lack of clinically relevant human lip cells and three-dimensional in vitro lip models, capable of replacing ethically questionable animal experimentations, represents a significant limitation in this area of research. Methods To address these limitations, we aimed to pioneer the introduction of immortalized healthy lip- and cleft lip-derived keratinocytes. Primary keratinocytes were isolated from patients' samples and immortalized by introducing the catalytic domain of telomerase, combined with the targeted knockdown of the cell cycle inhibitor gene, p16 INK4A . We then focused on validating the newly established cell lines by comparing their genetic stability and key phenotypic features with their primary keratinocyte counterparts. Results The newly established immortalized keratinocyte cell lines demonstrated genetic stability and preserved the main phenotypic characteristics of primary keratinocytes, such as cellular morphology and differentiation capacity. Three-dimensional lip models, generated using these cell lines, proved to be effective and convenient platforms for screening applications, including wound healing and microbial infection of the lip epithelium. Discussion The establishment of immortalized keratinocytes derived from healthy and cleft lips represents a significant achievement in lip research. These cell lines and the associated three-dimensional lip models are valuable tools that can be used as convenient screening platforms for various assays in a multitude of lip-related research areas, including dermatology, skin care, wound healing, tissue engineering, and craniofacial anomalies. This work opens new avenues in studying lip abnormalities and provides unique tools for personalized medicine approaches beneficial to patients.
Collapse
Affiliation(s)
- Farah Mansour
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Ludovica Parisi
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| | - Silvia Rihs
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| | - Isabelle Schnyder
- University Clinic for Pediatric Surgery, Bern University Hospital, Bern, Switzerland
| | - Giorgio C. La Scala
- Division of Pediatric Surgery, Department of Pediatrics, University Hospital of Geneva, Geneva, Switzerland
| | - Nijas Aliu
- Department of Human Genetics, Bern University Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Christos Katsaros
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| | - Martin Degen
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| |
Collapse
|
23
|
Cheng C, Cai X, Li J, Zhang X, Xie Y, Zhang J. In Vitro Culture of Human Norovirus in the Last 20 Years. Biomedicines 2024; 12:2442. [PMID: 39595008 PMCID: PMC11592199 DOI: 10.3390/biomedicines12112442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Human noroviruses (HuNoVs) are the main pathogens that cause acute gastroenteritis and lead to huge economic losses annually. Due to the lack of suitable culture systems, the pathogenesis of HuNoVs and the development of vaccines and drugs have progressed slowly. Although researchers have employed various methods to culture HuNoVs in vitro in the last century, problems relating to the irreducibility, low viral titer, and non-infectiousness of the progeny virus should not be ignored. In 2016, researchers achieved the cultivation and successive passaging of some HuNoV genotypes using human intestinal enteroids, initially demonstrating the potential use of organoids in overcoming this challenge. This paper reviews the efforts made in the last 20 years to culture HuNoVs in vitro and discusses the superiority and limitations of employing human intestinal enteroids/organoids as an in vitro culture model for HuNoVs.
Collapse
Affiliation(s)
- Chao Cheng
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; (C.C.); (J.L.); (X.Z.)
| | - Xia Cai
- Biosafety Level 3 Laboratory, Shanghai Medical College, Fudan University, Shanghai 200032, China;
| | - Jingjing Li
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; (C.C.); (J.L.); (X.Z.)
| | - Xiaomeng Zhang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; (C.C.); (J.L.); (X.Z.)
| | - Youhua Xie
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; (C.C.); (J.L.); (X.Z.)
| | - Junqi Zhang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; (C.C.); (J.L.); (X.Z.)
| |
Collapse
|
24
|
Jeong H, Kim NK, Park D, Youn H, Osuji CO, Doh J. Cu(II)-Organic Coordination Polymer Networks for Persistent Nitric Oxide Release in Tumor Therapy. Biomacromolecules 2024; 25:6830-6839. [PMID: 39283833 DOI: 10.1021/acs.biomac.4c01071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Nitric oxide (NO) plays a key role in regulating the immune system by polarizing macrophages toward the proinflammatory M1 phenotype, which is beneficial for cancer immunotherapy. We developed a Cu-organic coordination polymer network to sustainably release NO from endogenous donors. This robust polymer network was constructed through a dual-interaction process: complexation and cross-linking. The carboxylate groups of deprotonated 4-((6-(acryloyloxy)hexyl)oxy)benzoic acid (BA) served as bidentate ligands for the formation of Cu(II) complexes. The acrylate moiety of BA anchored these complexes in the polymer network, forming a cross-linked film. Cu ions within the network catalytically promoted NO release from S-nitrosoglutathione, maintaining this release even after 90 days in a physiological environment. The released NO effectively polarized both resting (M0) and tumor-promoting (M2) macrophages to the M1 phenotype. With their demonstrated physiological stability and sustained NO release performance, BA-Cu films hold potential as anticancer patches capable of continuously promoting antitumoral macrophages.
Collapse
Affiliation(s)
- Hyejoong Jeong
- Research Institute of Advanced Materials (RIAM), Seoul National University, Seoul 08826, Republic of Korea
| | - Na Kyung Kim
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Daehwan Park
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Department of Chemistry & Cosmetics, Jeju National University, 102 Jejudaehak-ro, Jeju-si, Jeju-do 63234, Republic of Korea
| | - Heesoo Youn
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Chinedum O Osuji
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Junsang Doh
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
25
|
Herb M, Schatz V, Hadrian K, Hos D, Holoborodko B, Jantsch J, Brigo N. Macrophage variants in laboratory research: most are well done, but some are RAW. Front Cell Infect Microbiol 2024; 14:1457323. [PMID: 39445217 PMCID: PMC11496307 DOI: 10.3389/fcimb.2024.1457323] [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: 06/30/2024] [Accepted: 09/06/2024] [Indexed: 10/25/2024] Open
Abstract
Macrophages play a pivotal role in the innate immune response. While their most characteristic function is phagocytosis, it is important not to solely characterize macrophages by this activity. Their crucial roles in body development, homeostasis, repair, and immune responses against pathogens necessitate a broader understanding. Macrophages exhibit remarkable plasticity, allowing them to modify their functional characteristics in response to the tissue microenvironment (tissue type, presence of pathogens or inflammation, and specific signals from neighboring cells) swiftly. While there is no single defined "macrophage" entity, there is a diverse array of macrophage types because macrophage ontogeny involves the differentiation of progenitor cells into tissue-resident macrophages, as well as the recruitment and differentiation of circulating monocytes in response to tissue-specific cues. In addition, macrophages continuously sense and respond to environmental cues and tissue conditions, adjusting their functional and metabolic states accordingly. Consequently, it is of paramount importance to comprehend the heterogeneous origins and functions of macrophages employed in in vitro studies, as each available in vitro macrophage model is associated with specific sets of strengths and limitations. This review centers its attention on a comprehensive comparison between immortalized mouse macrophage cell lines and primary mouse macrophages. It provides a detailed analysis of the strengths and weaknesses inherent in these in vitro models. Finally, it explores the subtle distinctions between diverse macrophage cell lines, offering insights into numerous factors beyond the model type that can profoundly influence macrophage function.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Valentin Schatz
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Karina Hadrian
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Deniz Hos
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Bohdan Holoborodko
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, Regensburg, Germany
| | - Jonathan Jantsch
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Natascha Brigo
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
26
|
Harris RM, Whitfield T, Blanton LV, Skaletsky H, Blumen K, Hyland P, McDermott E, Summers K, Hughes JF, Jackson E, Teglas P, Liu B, Chan YM, Page DC. Independent effects of testosterone, estradiol, and sex chromosomes on gene expression in immune cells of trans- and cisgender individuals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617275. [PMID: 39416170 PMCID: PMC11482753 DOI: 10.1101/2024.10.08.617275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The origins of sex differences in human disease are elusive, in part because of difficulties in separating the effects of sex hormones and sex chromosomes. To separate these variables, we examined gene expression in four groups of trans- or cisgender individuals: XX individuals treated with exogenous testosterone (n=21), XY treated with exogenous estradiol (n=13), untreated XX (n=20), and untreated XY (n=15). We performed single-cell RNA-sequencing of 358,426 peripheral blood mononuclear cells. Across the autosomes, 8 genes responded with a significant change in expression to testosterone, 34 to estradiol, and 32 to sex chromosome complement with no overlap between the groups. No sex-chromosomal genes responded significantly to testosterone or estradiol, but X-linked genes responded to sex chromosome complement in a remarkably stable manner across cell types. Through leveraging a four-state study design, we successfully separated the independent actions of testosterone, estradiol, and sex chromosome complement on genome-wide gene expression in humans.
Collapse
Affiliation(s)
- Rebecca M. Harris
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Whitehead Institute, Cambridge, MA 02142, USA
| | | | | | - Helen Skaletsky
- Whitehead Institute, Cambridge, MA 02142, USA
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA
| | - Kai Blumen
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Phoebe Hyland
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Em McDermott
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Kiana Summers
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
| | | | | | | | - Bingrun Liu
- Whitehead Institute, Cambridge, MA 02142, USA
| | - Yee-Ming Chan
- Division of Endocrinology, Boston Children’s Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - David C. Page
- Whitehead Institute, Cambridge, MA 02142, USA
- Howard Hughes Medical Institute, Whitehead Institute, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Lead contact
| |
Collapse
|
27
|
Gaylord EA, Choy HL, Chen G, Briner SL, Doering TL. Sac1 links phosphoinositide turnover to cryptococcal virulence. mBio 2024; 15:e0149624. [PMID: 38953635 PMCID: PMC11323556 DOI: 10.1128/mbio.01496-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 07/04/2024] Open
Abstract
Cryptococcus neoformans is an environmentally acquired fungal pathogen that causes over 140,000 deaths per year. Cryptococcal infection occurs when infectious particles are deposited into the lung, where they encounter host phagocytic cells. C. neoformans may be engulfed by these phagocytes, an important step of infection that leads to outcomes ranging from termination of infection to cryptococcal dissemination. To study this critical process, we screened approximately 4,700 cryptococcal gene deletion mutants for altered uptake, using primary mouse and human phagocytic cells. Among the hits of these two screens, we identified 93 mutants with perturbed uptake in both systems, as well as others with differences in uptake by only one cell type. We further screened the hits for changes in thickness of the capsule, a protective polysaccharide layer around the cell which is an important cryptococcal virulence factor. The combination of our three screens yielded 45 mutants, including one lacking the phosphatidylinositol-4-phosphate phosphatase Sac1. In this work, we implicate Sac1 in both host cell uptake and capsule production. We found that sac1 mutants exhibit lipid trafficking defects, reductions in secretory system function, and changes in capsule size and composition. Many of these changes occur specifically in tissue culture media, highlighting the role of Sac1 phosphatase activity in responding to the stress of host-like conditions. Overall, these findings show how genome-scale screening can identify cellular factors that contribute to our understanding of cryptococcal biology and demonstrate the role of Sac1 in determining fungal virulence.IMPORTANCECryptococcus neoformans is a fungal pathogen with significant impact on global health. Cryptococcal cells inhaled from the environment are deposited into the lungs, where they first contact the human immune system. The interaction between C. neoformans and host cells is critical because this step of infection can determine whether the fungal cells die or proliferate within the human host. Despite the importance of this stage of infection, we have limited knowledge of cryptococcal factors that influence its outcome. In this study, we identify cryptococcal genes that affect uptake by both human and mouse cells. We also identify mutants with altered capsule, a protective coating that surrounds the cells to shield them from the host immune system. Finally, we characterize the role of one gene, SAC1, in these processes. Overall, this study contributes to our understanding of how C. neoformans interacts with and protects itself from host cells.
Collapse
Affiliation(s)
- Elizabeth A. Gaylord
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hau Lam Choy
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Guohua Chen
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sydney L. Briner
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tamara L. Doering
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
28
|
Gottipati MK, D'Amato AR, Saksena J, Popovich PG, Wang Y, Gilbert RJ. Delayed administration of interleukin-4 coacervate alleviates the neurotoxic phenotype of astrocytes and promotes functional recovery after a contusion spinal cord injury. J Neural Eng 2024; 21:046052. [PMID: 39029499 DOI: 10.1088/1741-2552/ad6596] [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: 01/30/2024] [Accepted: 07/19/2024] [Indexed: 07/21/2024]
Abstract
Objective. Macrophages and astrocytes play a crucial role in the aftermath of a traumatic spinal cord injury (SCI). Infiltrating macrophages adopt a pro-inflammatory phenotype while resident astrocytes adopt a neurotoxic phenotype at the injury site, both of which contribute to neuronal death and inhibit axonal regeneration. The cytokine interleukin-4 (IL-4) has shown significant promise in preclinical models of SCI by alleviating the macrophage-mediated inflammation and promoting functional recovery. However, its effect on neurotoxic reactive astrocytes remains to be elucidated, which we explored in this study. We also studied the beneficial effects of a sustained release of IL-4 from an injectable biomaterial compared to bolus administration of IL-4.Approach. We fabricated a heparin-based coacervate capable of anchoring and releasing bioactive IL-4 and tested its efficacyin vitroandin vivo. Main results. We show that IL-4 coacervate is biocompatible and drives a robust anti-inflammatory macrophage phenotype in culture. We also show that IL-4 and IL-4 coacervate can alleviate the reactive neurotoxic phenotype of astrocytes in culture. Finally, using a murine model of contusion SCI, we show that IL-4 and IL-4 coacervate, injected intraspinally 2 d post-injury, can reduce macrophage-mediated inflammation, and alleviate neurotoxic astrocyte phenotype, acutely and chronically, while also promoting neuroprotection with significant improvements in hindlimb locomotor recovery. We observed that IL-4 coacervate can promote a more robust regenerative macrophage phenotypein vitro, as well as match its efficacyin vivo,compared to bolus IL-4.Significance. Our work shows the promise of coacervate as a great choice for local and prolonged delivery of cytokines like IL-4. We support this by showing that the coacervate can release bioactive IL-4, which acts on macrophages and astrocytes to promote a pro-regenerative environment following a SCI leading to robust neuroprotective and functional outcomes.
Collapse
Affiliation(s)
- Manoj K Gottipati
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States of America
- Department of Neuroscience, The Ohio State University, 460 W. 12th Avenue, Columbus, OH 43210, United States of America
- Center for Brain and Spinal Cord Repair, The Ohio State University, 460 W. 12th Avenue, Columbus, OH 43210, United States of America
| | - Anthony R D'Amato
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 134 Hollister Drive, 283 Kimball Hall, Ithaca, NY 14853, United States of America
| | - Jayant Saksena
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States of America
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University, 460 W. 12th Avenue, Columbus, OH 43210, United States of America
- Center for Brain and Spinal Cord Repair, The Ohio State University, 460 W. 12th Avenue, Columbus, OH 43210, United States of America
| | - Yadong Wang
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, 134 Hollister Drive, 283 Kimball Hall, Ithaca, NY 14853, United States of America
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, United States of America
| |
Collapse
|
29
|
Müller D, Loskutov J, Küffer S, Marx A, Regenbrecht CRA, Ströbel P, Regenbrecht MJ. Cell Culture Models for Translational Research on Thymomas and Thymic Carcinomas: Current Status and Future Perspectives. Cancers (Basel) 2024; 16:2762. [PMID: 39123489 PMCID: PMC11312172 DOI: 10.3390/cancers16152762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Cell culture model systems are fundamental tools for studying cancer biology and identifying therapeutic vulnerabilities in a controlled environment. TET cells are notoriously difficult to culture, with only a few permanent cell lines available. The optimal conditions and requirements for the ex vivo establishment and permanent expansion of TET cells have not been systematically studied, and it is currently unknown whether different TET subtypes require different culture conditions or specific supplements. The few permanent cell lines available represent only type AB thymomas and thymic carcinomas, while attempts to propagate tumor cells derived from type B thymomas so far have been frustrated. It is conceivable that epithelial cells in type B thymomas are critically dependent on their interaction with immature T cells or their three-dimensional scaffold. Extensive studies leading to validated cell culture protocols would be highly desirable and a major advance in the field. Alternative methods such as tumor cell organoid models, patient-derived xenografts, or tissue slices have been sporadically used in TETs, but their specific contributions and advantages remain to be shown.
Collapse
Affiliation(s)
- Denise Müller
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany; (S.K.); (C.R.A.R.)
| | | | - Stefan Küffer
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany; (S.K.); (C.R.A.R.)
| | - Alexander Marx
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany; (S.K.); (C.R.A.R.)
| | - Christian R. A. Regenbrecht
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany; (S.K.); (C.R.A.R.)
- CELLphenomics GmbH, 13125 Berlin, Germany (M.J.R.)
- ASC Oncology GmbH, 13125 Berlin, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, 37075 Göttingen, Germany; (S.K.); (C.R.A.R.)
| | - Manuela J. Regenbrecht
- CELLphenomics GmbH, 13125 Berlin, Germany (M.J.R.)
- ASC Oncology GmbH, 13125 Berlin, Germany
- Department for Pneumology, Palliative Medicine, DRK Kliniken Berlin, 14050 Berlin, Germany
| |
Collapse
|
30
|
Rahman MM, Wells G, Rantala JK, Helleday T, Muthana M, Danson SJ. In-vitro assays for immuno-oncology drug efficacy assessment and screening for personalized cancer therapy: scopes and challenges. Expert Rev Clin Immunol 2024; 20:821-838. [PMID: 38546609 DOI: 10.1080/1744666x.2024.2336583] [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: 11/30/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Immunotherapies have revolutionized cancer treatment, but often fail to produce desirable therapeutic outcomes in all patients. Due to the inter-patient heterogeneity and complexity of the tumor microenvironment, personalized treatment approaches are gaining demand. Researchers have long been using a range of in-vitro assays including 2D models, organoid co-cultures, and cancer-on-a-chip platforms for cancer drug screening. A comparative analysis of these assays with their suitability, high-throughput capacity, and clinical translatability is required for optimal translational use. AREAS COVERED The review summarized in-vitro platforms with their comparative advantages and limitations including construction strategies, and translational potential for immuno-oncology drug efficacy assessment. We also discussed end-point analysis strategies so that researchers can contextualize their usefulness and optimally design experiments for personalized immunotherapy efficacy prediction. EXPERT OPINION Researchers developed several in-vitro platforms that can provide information on personalized immunotherapy efficacy from different angles. Image-based assays are undoubtedly more suitable to gather a wide range of information including cellular morphology and phenotypical behaviors but need significant improvement to overcome issues including background noise, sample preparation difficulty, and long duration of experiment. More studies and clinical trials are needed to resolve these issues and validate the assays before they can be used in real-life scenarios.
Collapse
Affiliation(s)
- Md Marufur Rahman
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Directorate General of Health Services, Dhaka, Bangladesh
| | - Greg Wells
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| | - Juha K Rantala
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Misvik Biology Ltd, Turku, Finland
| | - Thomas Helleday
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
- Department of Oncology-Pathology, Karolinska Institutet, Huddinge, Sweden
| | - Munitta Muthana
- Nanobug Oncology Sheffield, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| | - Sarah J Danson
- Sheffield Ex vivo Group, Division of Clinical Medicine, School of Medicine & Population Health, University of Sheffield, Sheffield, UK
| |
Collapse
|
31
|
Harper JM. Primary Cell Culture as a Model System for Evolutionary Molecular Physiology. Int J Mol Sci 2024; 25:7905. [PMID: 39063147 PMCID: PMC11277064 DOI: 10.3390/ijms25147905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Primary cell culture is a powerful model system to address fundamental questions about organismal physiology at the cellular level, especially for species that are difficult, or impossible, to study under natural or semi-natural conditions. Due to their ease of use, primary fibroblast cultures are the dominant model system, but studies using both somatic and germ cells are also common. Using these models, genome evolution and phylogenetic relationships, the molecular and biochemical basis of differential longevities among species, and the physiological consequences of life history evolution have been studied in depth. With the advent of new technologies such as gene editing and the generation of induced pluripotent stem cells (iPSC), the field of molecular evolutionary physiology will continue to expand using both descriptive and experimental approaches.
Collapse
Affiliation(s)
- James M Harper
- Department of Biological Sciences, Sam Houston State University, 1900 Avenue I, Huntsville, TX 77341, USA
| |
Collapse
|
32
|
Vergara EJ, Tran AC, Paul MJ, Harrison T, Cooper A, Reljic R. A modified mycobacterial growth inhibition assay for the functional assessment of vaccine-mediated immunity. NPJ Vaccines 2024; 9:123. [PMID: 38956057 PMCID: PMC11219912 DOI: 10.1038/s41541-024-00906-z] [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: 03/14/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
The Mycobacterial growth inhibition assay (MGIA) is an ex-vivo assay used to measure the overall functional immune response elicited by infection or vaccination. In tuberculosis (TB) vaccine development, MGIA is a potentially important tool for preclinical evaluation of early-stage vaccine candidates to complement existing assays, and to potentially reduce the need for lengthy and costly pathogenic Mycobacterium tuberculosis (Mtb) animal challenge experiments. The conventional method of MGIA in mice entails directly infecting mixed cell cultures, most commonly splenocytes, from immunised mice with mycobacteria. However, this direct infection of mixed cell populations may yield unreliable results and lacks sufficient sensitivity to discriminate well between different vaccines due to the low number of mycobacteria-permissive cells. Here, we modified the assay by inclusion of mycobacteria-infected congenic murine macrophage cell lines as the target cells, and by measuring the total number of killed cells rather than the relative reduction between different groups. Thus, using splenocytes from Mycobacterium bovis BCG immunised mice, and J774 and MH-S (BALB/c background) or BL/6-M (C57Bl/6 background) macrophage cell lines, we demonstrated that the modified assay resulted in at least 26-fold greater mycobacterial killing per set quantity of splenocytes as compared to the conventional method. This increased sensitivity of measuring mycobacterial killing was confirmed using both the standard culture forming unit (CFU) assay and luminescence readings of luciferase-tagged virulent and avirulent mycobacteria. We propose that the modified MGIA can be used as a highly calibrated tool for quantitating the killing capacity of immune cells in preclinical evaluation of vaccine candidates for TB.
Collapse
Affiliation(s)
- Emil Joseph Vergara
- Institute for Infection and Immunity, St. George's University of London, London, UK
| | - Andy Cano Tran
- Institute for Infection and Immunity, St. George's University of London, London, UK
| | - Matthew J Paul
- Institute for Infection and Immunity, St. George's University of London, London, UK
| | - Thomas Harrison
- Institute for Infection and Immunity, St. George's University of London, London, UK
| | - Andrea Cooper
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Rajko Reljic
- Institute for Infection and Immunity, St. George's University of London, London, UK.
| |
Collapse
|
33
|
Daniel J, Schönberger Alvarez AA, te Heesen P, Lehrheuer B, Pischinger S, Hollert H, Roß-Nickoll M, Du M. Air-liquid interface exposure of A549 human lung cells to characterize the hazard potential of a gaseous bio-hybrid fuel blend. PLoS One 2024; 19:e0300772. [PMID: 38913629 PMCID: PMC11195957 DOI: 10.1371/journal.pone.0300772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/06/2024] [Indexed: 06/26/2024] Open
Abstract
Gaseous and semi-volatile organic compounds emitted by the transport sector contribute to air pollution and have adverse effects on human health. To reduce harmful effects to the environment as well as to humans, renewable and sustainable bio-hybrid fuels are explored and investigated in the cluster of excellence "The Fuel Science Center" at RWTH Aachen University. However, data on the effects of bio-hybrid fuels on human health is scarce, leaving a data gap regarding their hazard potential. To help close this data gap, this study investigates potential toxic effects of a Ketone-Ester-Alcohol-Alkane (KEAA) fuel blend on A549 human lung cells. Experiments were performed using a commercially available air-liquid interface exposure system which was optimized beforehand. Then, cells were exposed at the air-liquid interface to 50-2000 ppm C3.7 of gaseous KEAA for 1 h. After a 24 h recovery period in the incubator, cells treated with 500 ppm C3.7 KEAA showed significant lower metabolic activity and cells treated with 50, 250, 500 and 1000 ppm C3.7 KEAA showed significant higher cytotoxicity compared to controls. Our data support the international occupational exposure limits of the single KEAA constituents. This finding applies only to the exposure scenario tested in this study and is difficult to extrapolate to the complex in vivo situation.
Collapse
Affiliation(s)
- Jonas Daniel
- Institute for Environmental Research, RWTH Aachen University, Aachen, Germany
| | | | - Pia te Heesen
- Institute for Environmental Research, RWTH Aachen University, Aachen, Germany
| | - Bastian Lehrheuer
- TME—Chair of Thermodynamics of Mobile Energy Conversion Systems, RWTH Aachen University, Aachen, Germany
| | - Stefan Pischinger
- TME—Chair of Thermodynamics of Mobile Energy Conversion Systems, RWTH Aachen University, Aachen, Germany
| | - Henner Hollert
- Department Evolutionary Ecology & Environmental Toxicology (E3T), Faculty Biological Sciences (FB15), Goethe University Frankfurt, Frankfurt, Germany
- Department Environmental Media Related Ecotoxicology, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Schmallenberg, Germany
| | - Martina Roß-Nickoll
- Institute for Environmental Research, RWTH Aachen University, Aachen, Germany
| | - Miaomiao Du
- Institute for Environmental Research, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
34
|
Elblová P, Lunova M, Dejneka A, Jirsa M, Lunov O. Impact of mechanical cues on key cell functions and cell-nanoparticle interactions. DISCOVER NANO 2024; 19:106. [PMID: 38907808 PMCID: PMC11193707 DOI: 10.1186/s11671-024-04052-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
In recent years, it has been recognized that mechanical forces play an important regulative role in living organisms and possess a direct impact on crucial cell functions, ranging from cell growth to maintenance of tissue homeostasis. Advancements in mechanobiology have revealed the profound impact of mechanical signals on diverse cellular responses that are cell type specific. Notably, numerous studies have elucidated the pivotal role of different mechanical cues as regulatory factors influencing various cellular processes, including cell spreading, locomotion, differentiation, and proliferation. Given these insights, it is unsurprising that the responses of cells regulated by physical forces are intricately linked to the modulation of nanoparticle uptake kinetics and processing. This complex interplay underscores the significance of understanding the mechanical microenvironment in shaping cellular behaviors and, consequently, influencing how cells interact with and process nanoparticles. Nevertheless, our knowledge on how localized physical forces affect the internalization and processing of nanoparticles by cells remains rather limited. A significant gap exists in the literature concerning a systematic analysis of how mechanical cues might bias the interactions between nanoparticles and cells. Hence, our aim in this review is to provide a comprehensive and critical analysis of the existing knowledge regarding the influence of mechanical cues on the complicated dynamics of cell-nanoparticle interactions. By addressing this gap, we would like to contribute to a detailed understanding of the role that mechanical forces play in shaping the complex interplay between cells and nanoparticles.
Collapse
Affiliation(s)
- Petra Elblová
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 3, 121 16, Prague 2, Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic.
| |
Collapse
|
35
|
Gaylord EA, Choy HL, Chen G, Briner SL, Doering TL. Sac1 links phosphoinositide turnover to cryptococcal virulence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576303. [PMID: 38293062 PMCID: PMC10827209 DOI: 10.1101/2024.01.18.576303] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Cryptococcus neoformans is an environmentally-acquired fungal pathogen that causes over 140,000 deaths per year. Cryptococcal infection occurs when infectious particles are deposited into the lung, where they encounter host phagocytic cells. C. neoformans may be engulfed by these phagocytes, an important step of infection that leads to outcomes ranging from termination of infection to cryptococcal dissemination. To study this critical process, we screened approximately 4,700 cryptococcal gene deletion mutants for altered uptake, using primary mouse and human phagocytic cells. Among the hits of these two screens, we identified 93 mutants with perturbed uptake in both systems, as well as others with differences in uptake by only one cell type. We further screened the hits for changes in thickness of the capsule, a protective polysaccharide layer around the cell which is an important cryptococcal virulence factor. The combination of our three screens yielded 45 mutants, including one lacking the phosphatidylinositol-4-phosphate phosphatase Sac1. In this work, we implicate Sac1 in both host cell uptake and capsule production. We found that sac1 mutants exhibit lipid trafficking defects, reductions in secretory system function, and changes in capsule size and composition. Many of these changes occur specifically in tissue culture media, highlighting the role of Sac1 phosphatase activity in responding to the stress of host-like conditions. Overall, these findings show how genome-scale screening can identify cellular factors that contribute to our understanding of cryptococcal biology and demonstrate the role of Sac1 in determining fungal virulence. IMPORTANCE Cryptococcus neoformans is a fungal pathogen with significant impact on global health. Cryptococcal cells inhaled from the environment are deposited into the lungs, where they first contact the human immune system. The interaction between C. neoformans and host cells is critical because this step of infection can determine whether the fungal cells die or proliferate within the human host. Despite the importance of this stage of infection, we have limited knowledge of cryptococcal factors that influence its outcome. In this study, we identify cryptococcal genes that affect uptake by both human and mouse cells. We also identify mutants with altered capsule, a protective coating that surrounds the cells to shield them from the host immune system. Finally, we characterize the role of one gene, SAC1 , in these processes. Overall, this study contributes to our understanding of how C. neoformans interacts with and protects itself from host cells.
Collapse
|
36
|
Alvarez MRS, Moreno PG, Grijaldo-Alvarez SJB, Yadlapati A, Zhou Q, Narciso MP, Completo GC, Nacario RC, Rabajante JF, Heralde FM, Lebrilla CB. The effects of immortalization on the N-glycome and proteome of CDK4-transformed lung cancer cells. Glycobiology 2024; 34:cwae030. [PMID: 38579012 PMCID: PMC11041852 DOI: 10.1093/glycob/cwae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/26/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024] Open
Abstract
Biological experiments are often conducted in vitro using immortalized cells due to their accessibility and ease of propagation compared to primary cells and live animals. However, immortalized cells may present different proteomic and glycoproteomic characteristics from the primary cell source due to the introduction of genes that enhance proliferation (e.g. CDK4) or enable telomere lengthening. To demonstrate the changes in phenotype upon CDK4-transformation, we performed LC-MS/MS glycomic and proteomic characterizations of a human lung cancer primary cell line (DTW75) and a CDK4-transformed cell line (GL01) derived from DTW75. We observed that the primary and CDK4-transformed cells expressed significantly different levels of sialylated, fucosylated, and sialofucosylated N-glycans. Specifically, the primary cells expressed higher levels of hybrid- and complex-type sialylated N-glycans, while CDK4-transformed cells expressed higher levels of complex-type fucosylated and sialofucosylated N-glycans. Further, we compared the proteomic differences between the cell lines and found that CDK4-transformed cells expressed higher levels of RNA-binding and adhesion proteins. Further, we observed that the CDK4-transformed cells changed N-glycosylation after 31 days in cell culture, with a decrease in high-mannose and increase in fucosylated, sialylated, and sialofucosylated N-glycans. Identifying these changes between primary and CDK4-transformed cells will provide useful insight when adapting cell lines that more closely resemble in vivo physiological conditions.
Collapse
Affiliation(s)
- Michael Russelle S Alvarez
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California, 95616, USA
| | - Patrick Gabriel Moreno
- Molecular Diagnostics and Cellular Therapeutics Laboratory, Lung Center of the Philippines, Quezon City, 1100, Philippines
| | - Sheryl Joyce B Grijaldo-Alvarez
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California, 95616, USA
- Institute of Chemistry, College of Arts and Sciences, University of the Philippines Los Baños, 4031, Philippines
| | - Anirudh Yadlapati
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California, 95616, USA
| | - Qingwen Zhou
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California, 95616, USA
| | - Michelle P Narciso
- Institute of Mathematical Sciences and Physics, College of Arts and Sciences, University of the Philippines Los Baños, 4031, Philippines
| | - Gladys Cherisse Completo
- Institute of Chemistry, College of Arts and Sciences, University of the Philippines Los Baños, 4031, Philippines
| | - Ruel C Nacario
- Institute of Chemistry, College of Arts and Sciences, University of the Philippines Los Baños, 4031, Philippines
| | - Jomar F Rabajante
- Institute of Mathematical Sciences and Physics, College of Arts and Sciences, University of the Philippines Los Baños, 4031, Philippines
| | - Francisco M Heralde
- Molecular Diagnostics and Cellular Therapeutics Laboratory, Lung Center of the Philippines, Quezon City, 1100, Philippines
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, 1000, Philippines
| | - Carlito B Lebrilla
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California, 95616, USA
- Department of Chemistry, Biochemistry, Molecular, Cellular and Developmental Biology Group, University of California, Davis, 1 Shields Avenue, Davis, California, 95616, USA
| |
Collapse
|
37
|
Lairikyengbam D, Wetterauer B, Schmiech M, Jahraus B, Kirchgessner H, Wetterauer P, Berschneider K, Beier V, Niesler B, Balta E, Samstag Y. Comparative analysis of whole plant, flower and root extracts of Chamomilla recutita L. and characteristic pure compounds reveals differential anti-inflammatory effects on human T cells. Front Immunol 2024; 15:1388962. [PMID: 38720895 PMCID: PMC11077421 DOI: 10.3389/fimmu.2024.1388962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/21/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction Chronic inflammation is a hallmark of chronic wounds and inflammatory skin diseases. Due to a hyperactive and prolonged inflammation triggered by proinflammatory immune cells, transitioning to the repair and healing phase is halted. T cells may exacerbate the proinflammatory milieu by secreting proinflammatory cytokines. Chamomilla recutita L. (chamomile) has been suggested for use in several inflammatory diseases, implying a capability to modulate T cells. Here, we have characterized and compared the effects of differently prepared chamomile extracts and characteristic pure compounds on the T cell redox milieu as well as on the migration, activation, proliferation, and cytokine production of primary human T cells. Methods Phytochemical analysis of the extracts was carried out by LC-MS/MS. Primary human T cells from peripheral blood (PBTs) were pretreated with aqueous or hydroethanolic chamomile extracts or pure compounds. Subsequently, the effects on intracellular ROS levels, SDF-1α induced T cell migration, T cell activation, proliferation, and cytokine production after TCR/CD3 and CD28 costimulation were determined. Gene expression profiling was performed using nCounter analysis, followed by ingenuity pathway analysis, and validation at protein levels. Results The tested chamomile extracts and pure compounds differentially affected intracellular ROS levels, migration, and activation of T cells. Three out of five differently prepared extracts and two out of three pure compounds diminished T cell proliferation. In line with these findings, LC-MS/MS analysis revealed high heterogeneity of phytochemicals among the different extracts. nCounter based gene expression profiling identified several genes related to T cell functions associated with activation and differentiation to be downregulated. Most prominently, apigenin significantly reduced granzyme B induction and cytotoxic T cell activity. Conclusion Our results demonstrate an anti-inflammatory effect of chamomile- derived products on primary human T cells. These findings provide molecular explanations for the observed anti-inflammatory action of chamomile and imply a broader use of chamomile extracts in T cell driven chronic inflammatory diseases such as chronic wounds and inflammatory skin diseases. Importantly, the mode of extract preparation needs to be considered as the resulting different phytochemicals can result in differential effects on T cells.
Collapse
Affiliation(s)
- Divya Lairikyengbam
- Section Molecular Immunology, Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Bernhard Wetterauer
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Michael Schmiech
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm, Ulm, Germany
| | - Beate Jahraus
- Section Molecular Immunology, Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Henning Kirchgessner
- Section Molecular Immunology, Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Pille Wetterauer
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany
| | - Karina Berschneider
- Section Molecular Immunology, Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Verena Beier
- Section Molecular Immunology, Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Beate Niesler
- Department of Human Molecular Genetics, Heidelberg University Hospital, Heidelberg, Germany
- nCounter Core Facility, Institute of Human Genetics, Heidelberg University Hospital, Heidelberg, Germany
| | - Emre Balta
- Section Molecular Immunology, Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Yvonne Samstag
- Section Molecular Immunology, Institute of Immunology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
38
|
Roberts MD, Ruple BA, Godwin JS, McIntosh MC, Chen SY, Kontos NJ, Agyin-Birikorang A, Michel M, Plotkin DL, Mattingly ML, Mobley B, Ziegenfuss TN, Fruge AD, Kavazis AN. A novel deep proteomic approach in human skeletal muscle unveils distinct molecular signatures affected by aging and resistance training. Aging (Albany NY) 2024; 16:6631-6651. [PMID: 38643460 PMCID: PMC11087122 DOI: 10.18632/aging.205751] [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: 06/12/2023] [Accepted: 03/18/2024] [Indexed: 04/22/2024]
Abstract
The skeletal muscle proteome alterations to aging and resistance training have been reported in prior studies. However, conventional proteomics in skeletal muscle typically yields wide protein abundance ranges that mask the detection of lowly expressed proteins. Thus, we adopted a novel deep proteomics approach whereby myofibril (MyoF) and non-MyoF fractions were separately subjected to protein corona nanoparticle complex formation prior to digestion and Liquid Chromatography Mass Spectrometry (LC-MS). Specifically, we investigated MyoF and non-MyoF proteomic profiles of the vastus lateralis muscle of younger (Y, 22±2 years old; n=5) and middle-aged participants (MA, 56±8 years old; n=6). Additionally, MA muscle was analyzed following eight weeks of resistance training (RT, 2d/week). Across all participants, the number of non-MyoF proteins detected averaged to be 5,645±266 (range: 4,888-5,987) and the number of MyoF proteins detected averaged to be 2,611±326 (range: 1,944-3,101). Differences in the non-MyoF (8.4%) and MyoF (2.5%) proteomes were evident between age cohorts, and most differentially expressed non-MyoF proteins (447/543) were more enriched in MA versus Y. Biological processes in the non-MyoF fraction were predicted to be operative in MA versus Y including increased cellular stress, mRNA splicing, translation elongation, and ubiquitin-mediated proteolysis. RT in MA participants only altered ~0.3% of MyoF and ~1.0% of non-MyoF proteomes. In summary, aging and RT predominantly affect non-contractile proteins in skeletal muscle. Additionally, marginal proteome adaptations with RT suggest more rigorous training may stimulate more robust effects or that RT, regardless of age, subtly alters basal state skeletal muscle protein abundances.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Max Michel
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA
| | | | | | - Brooks Mobley
- School of Kinesiology, Auburn University, Auburn, AL 36849, USA
| | | | - Andrew D. Fruge
- College of Nursing, Auburn University, Auburn, AL 36849, USA
| | | |
Collapse
|
39
|
Ohlsson E, Bolay C, Arabulan S, Galler KM, Buchalla W, Schmalz G, Widbiller M. In-vitro-cytotoxicity of self-adhesive dental restorative materials. Dent Mater 2024; 40:739-746. [PMID: 38403539 DOI: 10.1016/j.dental.2024.02.015] [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: 08/18/2023] [Revised: 01/05/2024] [Accepted: 02/12/2024] [Indexed: 02/27/2024]
Abstract
OBJECTIVES Although the introduction of self-adhesive composites in restorative dentistry is very promising, the innovation of new materials also presents challenges and unknowns. Therefore, the aim of this study was to investigate the cytotoxicity of four different self-adhesive composites (SAC) in vitro and to compare them with resin-modified glass ionomer cements (RM-GIC), a more established group of materials. METHODS Samples of the following materials were prepared according to ISO 7405/10993-12 and eluted in cell culture medium for 24 h at 37 °C: Vertise Flow, Fusio Liquid Dentin, Constic, Surefil One, Photac Fil and Fuji II LC. Primary human pulp cells were obtained from extracted wisdom teeth and cultured for 24 h with the extracts in serial dilutions. Cell viability was evaluated by MTT assay, membrane disruption was quantified by LDH assay and apoptosis was assessed by flow cytometry after annexin/PI staining. RESULTS Two SAC (Constic and Vertise Flow) and one RM-GIC (Photac Fil) significantly reduced cell viability by more than 30% compared to the untreated control (p < 0.001). Disruptive cell morphological changes were observed and the cells showed signs of late apoptosis and necrosis in flow cytometry. Membrane disruption was not observed with any of the investigated materials. CONCLUSION Toxic effects occurred independently of the substance group and need to be considered in the development of materials with regard to clinical implications. CLINICAL SIGNIFICANCE SAC have many beneficial qualities, however, the cytotoxic effects of certain products should be considered when applied in close proximity to the dental pulp, as is often required.
Collapse
Affiliation(s)
- Ella Ohlsson
- Department of Operative Dentistry and Periodontology, Friedrich-Alexander-University Erlangen-Nürnberg, Glückstraße 11, 91054 Erlangen, Germany
| | - Carola Bolay
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Sevgi Arabulan
- Department of Pedodontics, Ege University, Ege University Campus, 35040 Izmir, Turkey
| | - Kerstin M Galler
- Department of Operative Dentistry and Periodontology, Friedrich-Alexander-University Erlangen-Nürnberg, Glückstraße 11, 91054 Erlangen, Germany
| | - Wolfgang Buchalla
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Gottfried Schmalz
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany; Department of Periodontology, University of Bern, 3012 Bern, Switzerland
| | - Matthias Widbiller
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany.
| |
Collapse
|
40
|
Yeo C, Kim H, Jeon WJ, Lee J, Hong JY, Kim H, Lee YJ, Baek SH, Ha IH. Protective effect of Luffa cylindrica Roemer against dexamethasone-induced muscle atrophy in primary rat skeletal muscle cells. J Muscle Res Cell Motil 2024; 45:1-10. [PMID: 37845555 PMCID: PMC10844154 DOI: 10.1007/s10974-023-09661-5] [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: 08/01/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023]
Abstract
Glucocorticoids (GCs) are commonly used in the treatment of chronic inflammatory conditions. However, the administration of high doses and long-term use of GCs can induce muscle atrophy (MA) in patients, leading to a decline in quality of life and increased mortality. MA leads to protein degradation in skeletal muscle, resulting in a reduction of muscle mass. This process is triggered by GCs like dexamethasone (DEX), which induce the expression of E3 ubiquitin ligases, namely Atrogin-1 and muscle RING-finger protein-1 (MuRF1). In this study, we examined the anti-MA potential of Luffa cylindrica Roemer (LCR) on DEX-treated primary skeletal myotubes. Primary skeletal myotubes stimulated with LCR alone resulted in a significant upregulation of myotube development, characterized by an increase in both the number and diameter of myotubes. Contrastingly, combined treatment with LCR and DEX reduced the expression of Atrogin-1, while treatment with DEX alone induced the expression of MuRF1. Furthermore, LCR treatment successfully restored the number and diameter of myotubes that had been diminished by DEX treatment. These findings suggest that LCR holds potential for treating MA, as an accelerating effect on muscle development and anti-MA effects on primary skeletal muscle cells were observed.
Collapse
Affiliation(s)
- Changhwan Yeo
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Hyunseong Kim
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Wan-Jin Jeon
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Junseon Lee
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Jin Young Hong
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Hyun Kim
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Yoon Jae Lee
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea
| | - Seung Ho Baek
- College of Korean Medicine, Dongguk University, 32, Dongguk-ro, Ilsandong-gu, Goyang-si, 10326, Gyeonggi-do, Republic of Korea
| | - In-Hyuk Ha
- Jaseng Spine and Joint Research Institute, Jaseng Medical Foundation, Seoul, 135-896, Republic of Korea.
| |
Collapse
|
41
|
Boukhatem I, Fleury S, Jourdi G, Lordkipanidzé M. The intriguing role of platelets as custodians of brain-derived neurotrophic factor. Res Pract Thromb Haemost 2024; 8:102398. [PMID: 38706782 PMCID: PMC11066552 DOI: 10.1016/j.rpth.2024.102398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/26/2024] [Accepted: 03/18/2024] [Indexed: 05/07/2024] Open
Abstract
A State of the Art lecture titled "Platelets and neurotrophins" was presented at the International Society on Thrombosis and Haemostasis Congress in 2023. Neurotrophins, a family of neuronal growth factors known to support cognitive function, are increasingly recognized as important players in vascular health. Indeed, along with their canonical receptors, neurotrophins are expressed in peripheral tissues, particularly in the vasculature. The better-characterized neurotrophin in vascular biology is the brain-derived neurotrophic factor (BDNF). Its largest extracerebral pool resides within platelets, partly inherited from megakaryocytes and also likely internalized from circulation. Activation of platelets releases vast amounts of BDNF into their milieu and interestingly leads to platelet aggregation through binding of its receptor, the tropomyosin-related kinase B, on the platelet surface. As BDNF is readily available in plasma, a mechanism to preclude excessive platelet activation and aggregation appears critical. As such, binding of BDNF to α2-macroglobulin hinders its ability to bind its receptor and limits its platelet-activating effects to the site of vascular injury. Altogether, addition of BDNF to a forming clot facilitates not only paracrine platelet activation but also binding to fibrinogen, rendering the resulting clot more porous and plasma-permeable. Importantly, release of BDNF into circulation also appears to be protective against adverse cardiovascular and cerebrovascular outcomes, which has been reported in both animal models and epidemiologic studies. This opens an avenue for platelet-based strategies to deliver BDNF to vascular lesions and facilitate wound healing through its regenerative properties. Finally, we summarize relevant new data on this topic presented during the 2023 International Society on Thrombosis and Haemostasis Congress.
Collapse
Affiliation(s)
- Imane Boukhatem
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Samuel Fleury
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Georges Jourdi
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
- Université Paris Cité, Institut National de la Santé Et de la Recherche Médicale, Innovative Therapies in Haemostasis, Paris, France
- Service d’Hématologie Biologique, Assistance Publique : Hôpitaux de Paris, Hôpital Lariboisière, Paris, France
| | - Marie Lordkipanidzé
- Research Center, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
42
|
Toschi P, Viola I, Manenti I, Miretti S, Macchi E, Martignani E, Accornero P, Baratta M. Ovine Trophoblast Cells: Cell Isolation and Culturing from the Placenta at the Early Stage of Pregnancy. Methods Mol Biol 2024; 2749:123-133. [PMID: 38133780 DOI: 10.1007/978-1-0716-3609-1_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Embryo development is dependent upon the exchange of oxygen and nutrients through the placenta, mainly composed of peculiar epithelioid cells, known as trophoblast cells. Normal trophoblast functionality plays a key role during the whole pregnancy, especially in the first stage of placentation. This chapter explains the techniques to obtain sheep primary trophoblast cells from the early placenta. Overall, procedures for cell isolation, culture, characterization, and cryopreservation are described.
Collapse
Affiliation(s)
- Paola Toschi
- Department of Veterinary Sciences, University of Torino, Turin, Italy.
| | - Irene Viola
- Department of Veterinary Sciences, University of Torino, Turin, Italy
| | - Isabella Manenti
- Department of Veterinary Sciences, University of Torino, Turin, Italy
| | - Silvia Miretti
- Department of Veterinary Sciences, University of Torino, Turin, Italy
| | - Elisabetta Macchi
- Department of Veterinary Sciences, University of Torino, Turin, Italy
| | | | - Paolo Accornero
- Department of Veterinary Sciences, University of Torino, Turin, Italy
| | - Mario Baratta
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| |
Collapse
|
43
|
Ramadan Q, Hazaymeh R, Zourob M. Immunity-on-a-Chip: Integration of Immune Components into the Scheme of Organ-on-a-Chip Systems. Adv Biol (Weinh) 2023; 7:e2200312. [PMID: 36866511 DOI: 10.1002/adbi.202200312] [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: 11/18/2022] [Revised: 01/16/2023] [Indexed: 03/04/2023]
Abstract
Studying the immune system in vitro aims to understand how, when, and where the immune cells migrate/differentiate and respond to the various triggering events and the decision points along the immune response journey. It becomes evident that organ-on-a-chip (OOC) technology has a superior capability to recapitulate the cell-cell and tissue-tissue interaction in the body, with a great potential to provide tools for tracking the paracrine signaling with high spatial-temporal precision and implementing in situ real-time, non-destructive detection assays, therefore, enabling extraction of mechanistic information rather than phenotypic information. However, despite the rapid development in this technology, integration of the immune system into OOC devices stays among the least navigated tasks, with immune cells still the major missing components in the developed models. This is mainly due to the complexity of the immune system and the reductionist methodology of the OOC modules. Dedicated research in this field is demanded to establish the understanding of mechanism-based disease endotypes rather than phenotypes. Herein, we systemically present a synthesis of the state-of-the-art of immune-cantered OOC technology. We comprehensively outlined what is achieved and identified the technology gaps emphasizing the missing components required to establish immune-competent OOCs and bridge these gaps.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Rana Hazaymeh
- Almaarefa University, Diriyah, 13713, Kingdom of Saudi Arabia
| | | |
Collapse
|
44
|
Bi W, Kraft A, Engelskircher S, Mischke J, Witte M, Klawonn F, van Ham M, Cornberg M, Wedemeyer H, Hengst J, Jänsch L. Proteomics reveals a global phenotypic shift of NK cells in HCV patients treated with direct-acting antivirals. Eur J Immunol 2023; 53:e2250291. [PMID: 37515498 DOI: 10.1002/eji.202250291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 07/31/2023]
Abstract
Chronic hepatitis C virus (HCV) infections compromise natural killer (NK)-cell immunity. Direct-acting antivirals (DAA) effectively eliminate HCV, but the long-term effects on NK cells in cured patients are debated. We conducted a proteomic study on CD56+ NK cells of chronic HCV-infected patients before and 1 year after DAA therapy. Donor-variation was observed in NK-cell proteomes of HCV-infected patients, with 46 dysregulated proteins restored after DAA therapy. However, 30% of the CD56+ NK-cell proteome remained altered 1 year post-therapy, indicating a phenotypic shift with low donor-variation. NK cells from virus-negative cured patients exhibited global regulation of RNA-processing and pathways related to "stimuli response", "chemokine signaling", and "cytotoxicity regulation". Proteomics identified downregulation of vesicle transport components (CD107a, COPI/II complexes) and altered receptor expression profiles, indicating an inhibited NK-cell phenotype. Yet, activated NK cells from HCV patients before and after therapy effectively upregulated IFN-γ and recruited CD107a. Conversely, reduced surface expression levels of Tim-3 and 2B4 were observed before and after therapy. In conclusion, this study reveals long-term effects on the CD56+ NK-cell compartment in convalescent HCV patients 1 year after therapy, with limited abundance of vesicle transport complexes and surface receptors, associated with a responsive NK-cell phenotype.
Collapse
Affiliation(s)
- Wenjie Bi
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, P. R. China
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Anke Kraft
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, Hannover, Germany
- TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Sophie Engelskircher
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Jasmin Mischke
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, Hannover, Germany
- TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Moana Witte
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Frank Klawonn
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Department of Computer Science, Ostfalia University, Wolfenbüttel, Germany
| | - Marco van Ham
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, Hannover, Germany
- TWINCORE, A Joint Venture Between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany
| | - Heiner Wedemeyer
- Centre for Individualised Infection Medicine (CiiM), A Joint Venture Between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- German Centre for Infection Research (DZIF), Partner site Hannover-Braunschweig, Hannover, Germany
- Cluster of Excellence Resolving Infection Susceptibility (RESIST; EXC 2155), Hannover Medical School, Hannover, Germany
| | - Julia Hengst
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Lothar Jänsch
- Cellular Proteome Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
45
|
Fritch EJ, Mordant AL, Gilbert TSK, Wells CI, Yang X, Barker NK, Madden EA, Dinnon KH, Hou YJ, Tse LV, Castillo IN, Sims AC, Moorman NJ, Lakshmanane P, Willson TM, Herring LE, Graves LM, Baric RS. Investigation of the Host Kinome Response to Coronavirus Infection Reveals PI3K/mTOR Inhibitors as Betacoronavirus Antivirals. J Proteome Res 2023; 22:3159-3177. [PMID: 37634194 DOI: 10.1021/acs.jproteome.3c00182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
Host kinases play essential roles in the host cell cycle, innate immune signaling, the stress response to viral infection, and inflammation. Previous work has demonstrated that coronaviruses specifically target kinase cascades to subvert host cell responses to infection and rely upon host kinase activity to phosphorylate viral proteins to enhance replication. Given the number of kinase inhibitors that are already FDA approved to treat cancers, fibrosis, and other human disease, they represent an attractive class of compounds to repurpose for host-targeted therapies against emerging coronavirus infections. To further understand the host kinome response to betacoronavirus infection, we employed multiplex inhibitory bead mass spectrometry (MIB-MS) following MERS-CoV and SARS-CoV-2 infection of human lung epithelial cell lines. Our MIB-MS analyses revealed activation of mTOR and MAPK signaling following MERS-CoV and SARS-CoV-2 infection, respectively. SARS-CoV-2 host kinome responses were further characterized using paired phosphoproteomics, which identified activation of MAPK, PI3K, and mTOR signaling. Through chemogenomic screening, we found that clinically relevant PI3K/mTOR inhibitors were able to inhibit coronavirus replication at nanomolar concentrations similar to direct-acting antivirals. This study lays the groundwork for identifying broad-acting, host-targeted therapies to reduce betacoronavirus replication that can be rapidly repurposed during future outbreaks and epidemics. The proteomics, phosphoproteomics, and MIB-MS datasets generated in this study are available in the Proteomics Identification Database (PRIDE) repository under project identifiers PXD040897 and PXD040901.
Collapse
Affiliation(s)
- Ethan J Fritch
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7290, United States
| | - Angie L Mordant
- UNC Michael Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Thomas S K Gilbert
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7365, United States
| | - Carrow I Wells
- Structural Genomics Consortium, Department of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7264, United States
| | - Xuan Yang
- Structural Genomics Consortium, Department of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7264, United States
| | - Natalie K Barker
- UNC Michael Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Emily A Madden
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7290, United States
| | - Kenneth H Dinnon
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7290, United States
| | - Yixuan J Hou
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7400, United States
| | - Longping V Tse
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7400, United States
| | - Izabella N Castillo
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7290, United States
| | - Amy C Sims
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7400, United States
| | - Nathaniel J Moorman
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7290, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Premkumar Lakshmanane
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7290, United States
| | - Timothy M Willson
- Structural Genomics Consortium, Department of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7264, United States
| | - Laura E Herring
- UNC Michael Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7365, United States
| | - Lee M Graves
- UNC Michael Hooker Proteomics Core, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7365, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, United States
| | - Ralph S Baric
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7290, United States
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599-7400, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27514, United States
| |
Collapse
|
46
|
Zhu I, Landsman D. Clustered and diverse transcription factor binding underlies cell type specificity of enhancers for housekeeping genes. Genome Res 2023; 33:1662-1672. [PMID: 37884340 PMCID: PMC10691539 DOI: 10.1101/gr.278130.123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/12/2023] [Indexed: 10/28/2023]
Abstract
Housekeeping genes are considered to be regulated by common enhancers across different tissues. Here we report that most of the commonly expressed mouse or human genes across different cell types, including more than half of the previously identified housekeeping genes, are associated with cell type-specific enhancers. Furthermore, the binding of most transcription factors (TFs) is cell type-specific. We reason that these cell type specificities are causally related to the collective TF recruitment at regulatory sites, as TFs tend to bind to regions associated with many other TFs and each cell type has a unique repertoire of expressed TFs. Based on binding profiles of hundreds of TFs from HepG2, K562, and GM12878 cells, we show that 80% of all TF peaks overlapping H3K27ac signals are in the top 20,000-23,000 most TF-enriched H3K27ac peak regions, and approximately 12,000-15,000 of these peaks are enhancers (nonpromoters). Those enhancers are mainly cell type-specific and include those linked to the majority of commonly expressed genes. Moreover, we show that the top 15,000 most TF-enriched regulatory sites in HepG2 cells, associated with about 200 TFs, can be predicted largely from the binding profile of as few as 30 TFs. Through motif analysis, we show that major enhancers harbor diverse and clustered motifs from a combination of available TFs uniquely present in each cell type. We propose a mechanism that explains how the highly focused TF binding at regulatory sites results in cell type specificity of enhancers for housekeeping and commonly expressed genes.
Collapse
Affiliation(s)
- Iris Zhu
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894, USA
| | - David Landsman
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894, USA
| |
Collapse
|
47
|
Adeniyi-Ipadeola G, Nwanosike H, Ramani S. Human intestinal organoids as models to study enteric bacteria and viruses. Curr Opin Microbiol 2023; 75:102362. [PMID: 37536261 PMCID: PMC10529792 DOI: 10.1016/j.mib.2023.102362] [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: 05/18/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 08/05/2023]
Abstract
Laboratory studies of host-microbe interactions have historically been carried out using transformed cell lines and animal models. Although much has been learned from these models, recent advances in the development of multicellular, physiologically active, human intestinal organoid (HIO) cultures are allowing unprecedented discoveries of host-microbe interactions. Here, we review recent literature using HIOs as models to investigate the pathogenesis of clinically important enteric bacteria and viruses and study commensal intestinal microbes. We also discuss limitations of current HIO culture systems and how technical advances and innovative engineering approaches are providing new directions to improve the model. The studies discussed here highlight the potential of HIOs for studying microbial pathogenesis, host-microbe interactions, and for preclinical development of therapeutics and vaccines.
Collapse
Affiliation(s)
- Grace Adeniyi-Ipadeola
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Hephzibah Nwanosike
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
48
|
Yin M, Wang J, Ying X, Fang Z, Zhang X. Long non coding RNA, C8orf49, a novel diagnostic and prognostic biomarker, enhances PTEN/FZD4-mediated cell growth and metastasis by sponging miR-1323 in endometriosis. Mol Cell Endocrinol 2023; 575:112040. [PMID: 37557978 DOI: 10.1016/j.mce.2023.112040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/05/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023]
Abstract
Lack of sensitive biomarkers in the early stages of endometriosis (EMs) results in delayed diagnosis and intervention. Long non-coding RNAs (lncRNAs) have prognostic and diagnostic values in various diseases. However, the prognostic and diagnostic effects of lncRNAs on EMs have rarely been discussed in EMs. In this study, we found that lncRNA C8orf49 was stably overexpressed in EMs tissues/plasma, and its expression greatly influenced dysmenorrhea (p = 2.2605E-9) and the revised American Society for Reproductive Medicine stage (p = 0.040765) of EMs. Multivariate logistic regression results revealed that C8orf49 expression was an independent risk factor for EMs [p = 6.4997E-17, 95% confidence interval (CI) = 0.000559-0.023853]. In primary endometrial stromal cells (ESCs), inhibition of C8orf49 could impede the proliferation and metastasis of ESCs. C8orf49 influenced the expression of PTEN/FZD4 by absorbing miR-1323, thus controlling ESCs activity. The results of a subcutaneous endometriosis animal model showed that the inhibition of C8orf49 restrained endometrial growth. Overall, C8orf49 functioned as an activator of EMs pathogenesis via the C8orf49/miR-1323/PTEN/FZD4 axis.
Collapse
Affiliation(s)
- Meichen Yin
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Jianzhang Wang
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Xue Ying
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Zhou Fang
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Department of Gynecology, The First Affiliated Hospital of Huzhou University, Huzhou, Zhejiang, People's Republic of China
| | - Xinmei Zhang
- Department of Obstetrics and Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China.
| |
Collapse
|
49
|
Sekulic M, Abdollahi N, Graf L, Deigendesch N, Puche R, Bodmer D, Petkovic V. Human blood-labyrinth barrier on a chip: a unique in vitro tool for investigation of BLB properties. RSC Adv 2023; 13:25508-25517. [PMID: 37636514 PMCID: PMC10450574 DOI: 10.1039/d3ra04704k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/14/2023] [Indexed: 08/29/2023] Open
Abstract
Hearing loss is one of the leading causes of disability worldwide, usually as a result of hair cell damage in the inner ear due to aging, acoustic trauma, or exposure to antibiotics or chemotherapy. No drug therapies can protect or restore hearing and current in vitro and animal models used in drug discovery have a very low success rate, mostly due to major differences in anatomy and accessibility of the inner ear environment between species. The blood-labyrinth barrier (BLB) in the stria vascularis is a highly specialized capillary network that controls exchanges between the blood and interstitial space in the cochlea. The BLB is critical for normal hearing, functioning as a physical, transport, and metabolic barrier. To address its complexity and accessibility, we created the first micro-engineered human model of BLB on a chip using autogenous progenitor cells from adult temporal bones. We successfully isolated the BLB from post-mortem human tissue and established an endothelial cell and pericyte culture system on a BLB chip. Using biocompatible materials, we fabricated sustainable two chamber chips. We validated the size-dependent permeability limits of our BLB model by measuring the permeability to daptomycin (molecular weight 1.6 kDa) and midazolam (molecular weight 325.78 Da). Daptomycin did not pass through the BLB layer, whereas midazolam readily passed through the BLB in our system. Thus, our BLB-chip mimicked the integrity and permeability of human stria vascularis capillaries. This represents a major step towards establishing a reliable model for the development of hearing loss treatments.
Collapse
Affiliation(s)
- Marijana Sekulic
- Department of Biomedicine, University Hospital Basel, University of Basel Basel Switzerland
| | - Narjes Abdollahi
- Department of Biomedicine, University Hospital Basel, University of Basel Basel Switzerland
| | - Lukas Graf
- Clinic for Otolaryngology, Head and Neck Surgery, University Hospital Basel Basel Switzerland
| | | | - Raoul Puche
- Department of Biomedicine, University Hospital Basel, University of Basel Basel Switzerland
| | - Daniel Bodmer
- Department of Biomedicine, University Hospital Basel, University of Basel Basel Switzerland
- Clinic for Otolaryngology, Head and Neck Surgery, University Hospital Basel Basel Switzerland
| | - Vesna Petkovic
- Department of Biomedicine, University Hospital Basel, University of Basel Basel Switzerland
| |
Collapse
|
50
|
Yau A, Jogdand A, Chen Y. Blood-brain-barrier modeling with tissue chips for research applications in space and on Earth. FRONTIERS IN SPACE TECHNOLOGIES 2023; 4:1176943. [PMID: 38915909 PMCID: PMC11195916 DOI: 10.3389/frspt.2023.1176943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Tissue chip technology has revolutionized biomedical applications and the medical science field for the past few decades. Currently, tissue chips are one of the most powerful research tools aiding in in vitro work to accurately predict the outcome of studies when compared to monolayer two-dimensional (2D) cell cultures. While 2D cell cultures held prominence for a long time, their lack of biomimicry has resulted in a transition to 3D cell cultures, including tissue chips technology, to overcome the discrepancies often seen in in vitro studies. Due to their wide range of applications, different organ systems have been studied over the years, one of which is the blood brain barrier (BBB) which is discussed in this review. The BBB is an incredible protective unit of the body, keeping out pathogens from entering the brain through vasculature. However, there are some microbes and certain diseases that disrupt the function of this barrier which can lead to detrimental outcomes. Over the past few years, various designs of the BBB have been proposed and modeled to study drug delivery and disease modeling on Earth. More recently, researchers have started to utilize tissue chips in space to study the effects of microgravity on human health. BBB tissue chips in space can be a tool to understand function mechanisms and therapeutics. This review addresses the limitations of monolayer cell culture which could be overcome with utilizing tissue chips technology. Current BBB models on Earth and how they are fabricated as well as what influences the BBB cell culture in tissue chips are discussed. Then, this article reviews how application of these technologies together with incorporating biosensors in space would be beneficial to help in predicting a more accurate physiological response in specific tissue or organ chips. Finally, the current platforms used in space and some solutions to overcome some shortcomings for future BBB tissue chip research are also discussed.
Collapse
Affiliation(s)
| | | | - Yupeng Chen
- Nanomedicine Lab, Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States
| |
Collapse
|