Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.153
Filter
1.
Nat Commun ; 15(1): 7611, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39218970

ABSTRACT

The development of functional neurons is a complex orchestration of multiple signaling pathways controlling cell proliferation and differentiation. Because the balance of antioxidants is important for neuronal survival and development, we hypothesized that ferroptosis must be suppressed to gain neurons. We find that removal of antioxidants diminishes neuronal development and laminar organization of cortical organoids, which is fully restored when ferroptosis is inhibited by ferrostatin-1 or when neuronal differentiation occurs in the presence of vitamin A. Furthermore, iron-overload-induced developmental growth defects in C. elegans are ameliorated by vitamin E and A. We determine that all-trans retinoic acid activates the Retinoic Acid Receptor, which orchestrates the expression of anti-ferroptotic genes. In contrast, retinal and retinol show radical-trapping antioxidant activity. Together, our study reveals an unexpected function of vitamin A in coordinating the expression of essential cellular gatekeepers of ferroptosis, and demonstrates that suppression of ferroptosis by radical-trapping antioxidants or by vitamin A is required to obtain mature neurons and proper laminar organization in cortical organoids.


Subject(s)
Antioxidants , Caenorhabditis elegans , Ferroptosis , Neurons , Vitamin A , Animals , Ferroptosis/drug effects , Vitamin A/pharmacology , Vitamin A/metabolism , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/drug effects , Antioxidants/pharmacology , Neurons/metabolism , Neurons/drug effects , Neurons/cytology , Cyclohexylamines/pharmacology , Cell Differentiation/drug effects , Vitamin E/pharmacology , Receptors, Retinoic Acid/metabolism , Receptors, Retinoic Acid/genetics , Tretinoin/pharmacology , Organoids/drug effects , Organoids/metabolism , Neurogenesis/drug effects , Mice , Humans , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Signal Transduction/drug effects , Phenylenediamines
2.
J Transl Med ; 22(1): 816, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223664

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a neurodegenerative condition characterized by the loss of dopaminergic neurons and the accumulation of Lewy-body protein aggregates containing misfolded α-synuclein (α-syn) in a phosphorylated form. The lack of effective models for drug screens has hindered drug development studies for PD. However, the recent development of in vitro brain-like organoids provides a new opportunity for evaluating therapeutic agents to slow the progression of this chronic disease. METHODS: In this study, we used a 3D brain-like organoid model to investigate the potential of repurposing Tilorone, an anti-viral drug, for impeding the propagation of α-synucleinopathy. We assessed the effect of Tilorone on the uptake of fluorescently labeled α-syn preformed fibrils (sPFF) and sPFF-induced apoptosis using confocal microscopy. We also examined Tilorone's impact on the phosphorylation of endogenous α-syn induced by pathogenic sPFF by immunoblotting midbrain-like organoid extracts. Additionally, quantitative RT-PCR and proteomic profiling of sPFF-treated organoids were conducted to evaluate the global impact of Tilorone treatment on tissue homeostasis in the 3D organoid model. RESULTS: Tilorone inhibits the uptake of sPFF in both mouse primary neurons and human midbrain-like organoids. Tilorone also reduces the phosphorylation of endogenous α-syn induced by pathogenic α-syn fibrils and mitigates α-syn fibril-induced apoptosis in midbrain-like organoids. Proteomic profiling of fibril-treated organoids reveals substantial alterations in lipid homeostasis by α-syn fibrils, which are reversed by Tilorone treatment. Given its safety profile in clinics, Tilorone may be further developed as a therapeutic intervention to alleviate the propagation of synucleinopathy in PD patients.


Subject(s)
Mesencephalon , Organoids , Synucleinopathies , alpha-Synuclein , Mesencephalon/pathology , Mesencephalon/drug effects , Mesencephalon/metabolism , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , Humans , alpha-Synuclein/metabolism , Synucleinopathies/pathology , Synucleinopathies/metabolism , Synucleinopathies/drug therapy , Phosphorylation/drug effects , Models, Biological , Apoptosis/drug effects , Animals , Parkinson Disease/pathology , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Mice , Proteomics
3.
PLoS One ; 19(9): e0307414, 2024.
Article in English | MEDLINE | ID: mdl-39226257

ABSTRACT

Cancer continues to pose a significant global health challenge, with gastrointestinal (GI) cancers among the most prevalent and deadly forms. These cancers often lead to high mortality rates and demand the use of potent cytotoxic chemotherapeutics. For example, 5-fluorouracil (5-FU) forms the backbone of chemotherapy regimens for various GI cancers, including colorectal cancer. While these chemotherapeutics efficiently kill cancer cells, they frequently cause off-target effects such as chemotherapy-induced mucositis (CIM), characterized by debilitating symptoms like pain, nausea, and diarrhoea, necessitating medical intervention. In this study, we elucidated the potential of melatonin and misoprostol to reduce 5-FU-induced small intestinal mucositis. Morphological and cellular changes in the jejunum, along with colonic faecal water content were quantified in rats as markers for CIM. Additionally, the effects of melatonin were investigated in vitro on 5-FU treated murine intestinal organoids. The results showed that melatonin prevented villus atrophy in the rat jejunal mucosa and upheld cell viability in murine intestinal organoids. In contrast, misoprostol alone or in combination with melatonin did not significantly affect CIM caused by 5-FU. These in vivo and in vitro experiments provided promising insights that melatonin may be used as a preventive and/or adjuvant combination therapy to prevent and reduce CIM, holding the potential to enhance cancer treatment outcomes and improve patient quality-of-life.


Subject(s)
Fluorouracil , Intestine, Small , Melatonin , Mucositis , Organoids , Animals , Melatonin/pharmacology , Rats , Organoids/drug effects , Fluorouracil/adverse effects , Fluorouracil/pharmacology , Mice , Intestine, Small/drug effects , Intestine, Small/pathology , Mucositis/chemically induced , Mucositis/pathology , Mucositis/prevention & control , Mucositis/drug therapy , Male , Atrophy/chemically induced , Atrophy/drug therapy , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology
4.
J Vis Exp ; (210)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39158306

ABSTRACT

Intestinal epithelial cell (IEC) death is increased in patients with inflammatory bowel diseases (IBD) such as ulcerative colitis (UC) and Crohn's disease (CD). This can contribute to defects in intestinal barrier function, exacerbation of inflammation, and disease immunopathogenesis. Cytokines and death receptor ligands are partially responsible for this increase in IEC death. IBD-relevant cytokines, such as TNF-α and IFN-γ, are cytotoxic to IECs both independently and in combination. This protocol describes a simple and practical assay to quantify cytokine-induced cytotoxicity in CD patient-derived colonic organoids using a fluorescent cell death dye (SYTOX Green Nucleic Acid Stain), live fluorescence microscopy, and open-source image analysis software. We also demonstrate how to use the Bliss independence mathematical model to calculate a coefficient of perturbagen interaction (CPI) based on organoid cytotoxicity. The CPI can be used to determine if interactions between cytokine combinations or other types of perturbagens are antagonistic, additive, or synergistic. This protocol can be implemented to investigate the cytotoxic activity of cytokines and other perturbagens using patient-derived colonic organoids.


Subject(s)
Colon , Cytokines , Microscopy, Fluorescence , Organoids , Humans , Organoids/cytology , Organoids/drug effects , Colon/cytology , Microscopy, Fluorescence/methods , Cytokines/metabolism , Cytokines/pharmacology , Cell Death/drug effects
5.
Cells ; 13(15)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39120325

ABSTRACT

Neonicotinoids are synthetic, nicotine-derived insecticides used worldwide to protect crops and domestic animals from pest insects. The reported evidence shows that they are also able to interact with mammalian nicotine receptors (nAChRs), triggering detrimental responses in cultured neurons. Exposure to high neonicotinoid levels during the fetal period induces neurotoxicity in animal models. Considering the persistent exposure to these insecticides and the key role of nAChRs in brain development, their potential neurotoxicity on mammal central nervous system (CNS) needs further investigations. We studied here the neurodevelopmental effects of different generations of neonicotinoids on CNS cells in mouse fetal brain and primary cultures and in neuronal cells and organoids obtained from human induced pluripotent stem cells (iPSC). Neonicotinoids significantly affect neuron viability, with imidacloprid (IMI) inducing relevant alterations in synaptic protein expression, neurofilament structures, and microglia activation in vitro, and in the brain of prenatally exposed mouse fetuses. IMI induces neurotoxic effects also on developing human iPSC-derived neurons and cortical organoids. Collectively, the current findings show that neonicotinoids might induce impairment during neuro/immune-development in mouse and human CNS cells and provide new insights in the characterization of risk for the exposure to this class of pesticides.


Subject(s)
Induced Pluripotent Stem Cells , Neonicotinoids , Neurons , Organoids , Animals , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Humans , Neonicotinoids/toxicity , Neurons/drug effects , Neurons/metabolism , Organoids/drug effects , Mice , Pesticides/toxicity , Cells, Cultured , Brain/drug effects , Female , Nitro Compounds/toxicity
6.
Pediatr Surg Int ; 40(1): 214, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39102048

ABSTRACT

PURPOSE: We performed animal and organoid study to evaluate the anti-fibrotic effect of steroid on biliary atresia (BA) and the underlying patho-mechanism. METHODS: BA animal models were created by inoculation of mice on post-natal day 1 with rhesus rotavirus (RRV). They received either 20 µl phosphate-buffered saline (PBS) or steroid from day 21 to day 34. On day 34, their serum samples were collected for hormonal markers. Necrosis, fibrosis and CK 19 expression in the liver were evaluated. Liver organoids were developed and their morphology as well as bulk RNA sequencing data were analyzed. RESULTS: Twenty-four mice developed BA features after RRV injection and were equally divided into steroid and PBS groups. On day 34, the weight gain of steroid group increased significantly than PBS group (p < 0.0001). All mice in the PBS group developed liver fibrosis but only one mouse in the steroid group did. Serum bilirubin and liver parenchymal enzymes were significantly lower in steroid group. The morphology of liver organoids were different between the two groups. A total of 6359 differentially expressed genes were found between steroid group and PBS group. CONCLUSION: Based on our findings obtained from RRV-induced BA animal and organoid models, steroid has the potential to mitigate liver fibrosis in BA.


Subject(s)
Biliary Atresia , Disease Models, Animal , Liver Cirrhosis , Organoids , Animals , Mice , Organoids/drug effects , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Liver/pathology , Liver/drug effects
7.
PLoS One ; 19(8): e0308743, 2024.
Article in English | MEDLINE | ID: mdl-39121095

ABSTRACT

Human retinal organoids have become indispensable tools for retinal disease modeling and drug screening. Despite its versatile applications, the long timeframe for their differentiation and maturation limits the throughput of such research. Here, we successfully shortened this timeframe by accelerating human retinal organoid development using unique pharmacological approaches. Our method comprised three key steps: 1) a modified self-formed ectodermal autonomous multizone (SEAM) method, including dual SMAD inhibition and bone morphogenetic protein 4 treatment, for initial neural retinal induction; 2) the concurrent use of a Sonic hedgehog agonist SAG, activin A, and all-trans retinoic acid for rapid retinal cell specification; and 3) switching to SAG treatment alone for robust retinal maturation and lamination. The generated retinal organoids preserved typical morphological features of mature retinal organoids, including hair-like surface structures and well-organized outer layers. These features were substantiated by the spatial immunostaining patterns of several retinal cell markers, including rhodopsin and L/M opsin expression in the outermost layer, which was accompanied by reduced ectopic cone photoreceptor generation. Importantly, our method required only 90 days for retinal organoid maturation, which is approximately two-thirds the time necessary for other conventional methods. These results indicate that thoroughly optimized pharmacological interventions play a pivotal role in rapid and precise photoreceptor development during human retinal organoid differentiation and maturation. Thus, our present method may expedite human retinal organoid research, eventually contributing to the development of better treatment options for various degenerative retinal diseases.


Subject(s)
Activins , Cell Differentiation , Hedgehog Proteins , Organoids , Retina , Signal Transduction , Tretinoin , Humans , Activins/pharmacology , Activins/metabolism , Organoids/drug effects , Organoids/metabolism , Organoids/cytology , Hedgehog Proteins/metabolism , Tretinoin/pharmacology , Retina/metabolism , Retina/cytology , Retina/drug effects , Signal Transduction/drug effects , Cell Differentiation/drug effects , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
8.
PLoS Comput Biol ; 20(8): e1012256, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39093897

ABSTRACT

Patient-derived tumor organoids (PDTOs) are novel cellular models that maintain the genetic, phenotypic and structural features of patient tumor tissue and are useful for studying tumorigenesis and drug response. When integrated with advanced 3D imaging and analysis techniques, PDTOs can be used to establish physiologically relevant high-throughput and high-content drug screening platforms that support the development of patient-specific treatment strategies. However, in order to effectively leverage high-throughput PDTO observations for clinical predictions, it is critical to establish a quantitative understanding of the basic properties and variability of organoid growth dynamics. In this work, we introduced an innovative workflow for analyzing and understanding PDTO growth dynamics, by integrating a high-throughput imaging deep learning platform with mathematical modeling, incorporating flexible growth laws and variable dormancy times. We applied the workflow to colon cancer organoids and demonstrated that organoid growth is well-described by the Gompertz model of growth. Our analysis showed significant intrapatient heterogeneity in PDTO growth dynamics, with the initial exponential growth rate of an organoid following a lognormal distribution within each dataset. The level of intrapatient heterogeneity varied between patients, as did organoid growth rates and dormancy times of single seeded cells. Our work contributes to an emerging understanding of the basic growth characteristics of PDTOs, and it highlights the heterogeneity in organoid growth both within and between patients. These results pave the way for further modeling efforts aimed at predicting treatment response dynamics and drug resistance timing.


Subject(s)
Organoids , Humans , Organoids/growth & development , Organoids/drug effects , Organoids/pathology , Models, Biological , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/pathology , Colonic Neoplasms/drug therapy , Computational Biology , Deep Learning , Models, Theoretical , Imaging, Three-Dimensional/methods
9.
Stem Cell Res Ther ; 15(1): 268, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39183338

ABSTRACT

BACKGROUND: The KCNJ16 gene has been associated with a novel kidney tubulopathy phenotype, viz. disturbed acid-base homeostasis, hypokalemia and altered renal salt transport. KCNJ16 encodes for Kir5.1, which together with Kir4.1 constitutes a potassium channel located at kidney tubular cell basolateral membranes. Preclinical studies provided mechanistic links between Kir5.1 and tubulopathy, however, the disease pathology remains poorly understood. Here, we aimed at generating and characterizing a novel advanced in vitro human kidney model that recapitulates the disease phenotype to investigate further the pathophysiological mechanisms underlying the tubulopathy and potential therapeutic interventions. METHODS: We used CRISPR/Cas9 to generate KCNJ16 mutant (KCNJ16+/- and KCNJ16-/-) cell lines from healthy human induced pluripotent stem cells (iPSC) KCNJ16 control (KCNJ16WT). The iPSCs were differentiated following an optimized protocol into kidney organoids in an air-liquid interface. RESULTS: KCNJ16-depleted kidney organoids showed transcriptomic and potential functional impairment of key voltage-dependent electrolyte and water-balance transporters. We observed cysts formation, lipid droplet accumulation and fibrosis upon Kir5.1 function loss. Furthermore, a large scale, glutamine tracer flux metabolomics analysis demonstrated that KCNJ16-/- organoids display TCA cycle and lipid metabolism impairments. Drug screening revealed that treatment with statins, particularly the combination of simvastatin and C75, prevented lipid droplet accumulation and collagen-I deposition in KCNJ16-/- kidney organoids. CONCLUSIONS: Mature kidney organoids represent a relevant in vitro model for investigating the function of Kir5.1. We discovered novel molecular targets for this genetic tubulopathy and identified statins as a potential therapeutic strategy for KCNJ16 defects in the kidney.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors , Induced Pluripotent Stem Cells , Organoids , Potassium Channels, Inwardly Rectifying , Humans , Organoids/metabolism , Organoids/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Induced Pluripotent Stem Cells/metabolism , Kidney/metabolism , Kidney/pathology , Kidney/drug effects , Lipid Metabolism/drug effects
10.
Cells ; 13(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39195202

ABSTRACT

Cancer is a leading cause of death worldwide. Around one-third of the total global cancer incidence and mortality are related to gastrointestinal (GI) cancers. Over the past few years, rapid developments have been made in patient-derived organoid (PDO) models for gastrointestinal cancers. By closely mimicking the molecular properties of their parent tumors in vitro, PDOs have emerged as powerful tools in personalized medicine and drug discovery. Here, we review the current literature on the application of PDOs of common gastrointestinal cancers in the optimization of drug treatment strategies in the clinic and their rising importance in pre-clinical drug development. We discuss the advantages and limitations of gastrointestinal cancer PDOs and outline the microfluidics-based strategies that improve the throughput of PDO models in order to extract the maximal benefits in the personalized medicine and drug discovery process.


Subject(s)
Gastrointestinal Neoplasms , Organoids , Precision Medicine , Humans , Precision Medicine/methods , Organoids/drug effects , Organoids/pathology , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/pathology , Drug Evaluation, Preclinical/methods , Drug Discovery/methods , Drug Screening Assays, Antitumor/methods
11.
J Transl Med ; 22(1): 754, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39135062

ABSTRACT

BACKGROUND: Organoids are approved by the US FDA as an alternative to animal experiments to guide drug development and for sensitivity screening. Stable organoids models of gastric cancer are desirable for personalized medicine and drug screening. METHODS: Tumor tissues from a primary cancer of the stomach and metastatic cancer of the lymph node were collected for 3D culture. By long-term culture for over 50 generations in vitro, we obtained stably growing organoid lines. We analyzed short tandem repeats (STRs) and karyotypes of cancer cells, and tumorigenesis of the organoids in nude mice, as well as multi-omics profiles of the organoids. A CCK8 method was used to determine the drugs sensitivity to fluorouracil (5-Fu), platinum and paclitaxel. RESULTS: Paired organoid lines from primary cancer (SPDO1P) and metastatic lymph node (SPDO1LM) were established with unique STRs and karyotypes. The organoid lines resulted in tumorigenesis in vivo and had clear genetic profiles. Compared to SPDO1P from primary cancer, upregulated genes of SPDO1LM from the metastatic lymph node were enriched in pathways of epithelial-mesenchymal transition and angiogenesis with stronger abilities of cell migration, invasion, and pro-angiogenesis. Based on drug sensitivity analysis, the SOX regimen (5-Fu plus oxaliplatin) was used for chemotherapy with an optimal clinical outcome. CONCLUSIONS: The organoid lines recapitulate the drug sensitivity of the parental tissues. The paired organoid lines present a step-change toward living biobanks for further translational usage.


Subject(s)
Lymphatic Metastasis , Mice, Nude , Organoids , Precision Medicine , Stomach Neoplasms , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/drug therapy , Organoids/drug effects , Organoids/pathology , Humans , Animals , Lymphatic Metastasis/pathology , Gene Expression Regulation, Neoplastic/drug effects , Cell Line, Tumor , Carcinogenesis/pathology , Carcinogenesis/genetics , Carcinogenesis/drug effects , Mice , Microsatellite Repeats/genetics
12.
Commun Biol ; 7(1): 972, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39122786

ABSTRACT

Dental pulp stem cells (DPSC) have shown osteogenic and bone regenerative potential. Improving the in situ bone regeneration potential of DPSC is crucial for their application as seed cells during bone defect reconstruction in clinics. This study aimed to develop DPSC-derived organoid-like microspheroids as effective seeds for bone tissue engineering applications. DPSC osteogenic microspheroids (70 µm diameter) were cultured in a polydimethylsiloxane-mold-based agarose-gel microwell-culture-system with or without cannabidiol (CBD)-treatment. Results of in vitro studies showed higher osteogenic differentiation potential of microspheroids compared with 2D-cultured-DPSC. CBD treatment further improved the osteogenic differentiation potential of microspheroids. The effect of CBD treatment in the osteogenic differentiation of microspheroids was more pronounced compared with that of CBD-treated 2D-cultured-DPSC. Microspheroids showed a higher degree of bone regeneration in nude mice calvarial bone defect compared to 2D-cultured-DPSC. CBD-treated microspheroids showed the most robust in situ bone regenerative potential compared with microspheroids or CBD-treated 2D-cultured-DPSC. According to mRNA sequencing, bioinformatic analysis, and confirmation study, the higher osteogenic potential of CBD-treated microspheroids was mainly attributed to WNT6 upregulation. Taken together, DPSC microspheroids have robust osteogenic potential and can effectively translate the effect of in vitro osteoinductive stimulation during in situ bone regeneration, indicating their application potential during bone defect reconstruction in clinics.


Subject(s)
Cannabidiol , Cell Differentiation , Dental Pulp , Osteogenesis , Stem Cells , Up-Regulation , Osteogenesis/drug effects , Animals , Cannabidiol/pharmacology , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/cytology , Mice , Up-Regulation/drug effects , Dental Pulp/cytology , Dental Pulp/drug effects , Cell Differentiation/drug effects , Organoids/drug effects , Organoids/metabolism , Humans , Mice, Nude , Cells, Cultured , Bone Regeneration/drug effects
13.
Stem Cell Res Ther ; 15(1): 269, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39183353

ABSTRACT

BACKGROUND: Liver disease imposes a significant medical burden that persists due to a shortage of liver donors and an incomplete understanding of liver disease progression. Hepatobiliary organoids (HBOs) could provide an in vitro mini-organ model to increase the understanding of the liver and may benefit the development of regenerative medicine. METHODS: In this study, we aimed to establish HBOs with bile duct (BD) structures and mature hepatocytes (MHs) using human chemically induced liver progenitor cells (hCLiPs). hCLiPs were induced in mature cryo-hepatocytes using a small-molecule cocktail of TGF-ß inhibitor (A-83-01, A), GSK3 inhibitor (CHIR99021, C), and 10% FBS (FAC). HBOs were then formed by seeding hCLiPs into ultralow attachment plates and culturing them with a combination of small molecules of Rock-inhibitor (Y-27632) and AC (YAC). RESULTS: These HBOs exhibited bile canaliculi of MHs connected to BD structures, mimicking bile secretion and transportation functions of the liver. The organoids showed gene expression patterns consistent with both MHs and BD structures, and functional assays confirmed their ability to transport the bile analogs of rhodamine-123 and CLF. Functional patient-specific HBOs were also successfully created from hCLiPs sourced from cirrhotic liver tissues. CONCLUSIONS: This study demonstrated the potential of human HBOs as an efficient model for studying hepatobiliary diseases, drug discovery, and personalized medicine.


Subject(s)
Bile Ducts , Liver , Organoids , Pyridines , Stem Cells , Humans , Organoids/metabolism , Organoids/drug effects , Bile Ducts/metabolism , Stem Cells/metabolism , Stem Cells/drug effects , Stem Cells/cytology , Pyridines/pharmacology , Liver/drug effects , Liver/metabolism , Hepatocytes/metabolism , Hepatocytes/drug effects , Hepatocytes/cytology , Pyrimidines/pharmacology , Amides/pharmacology , Cell Differentiation/drug effects , Pyrazoles , Thiosemicarbazones
14.
Biofabrication ; 16(4)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39189069

ABSTRACT

Recent years have seen the creation and popularization of various complexin vitromodels (CIVMs), such as organoids and organs-on-chip, as a technology with the potential to reduce animal usage in pharma while also enhancing our ability to create safe and efficacious drugs for patients. Public awareness of CIVMs has increased, in part, due to the recent passage of the FDA Modernization Act 2.0. This visibility is expected to spur deeper investment in and adoption of such models. Thus, end-users and model developers alike require a framework to both understand the readiness of current models to enter the drug development process, and to assess upcoming models for the same. This review presents such a framework for model selection based on comparative -omics data (which we term model-omics), and metrics for qualification of specific test assays that a model may support that we term context-of-use (COU) assays. We surveyed existing healthy tissue models and assays for ten drug development-critical organs of the body, and provide evaluations of readiness and suggestions for improving model-omics and COU assays for each. In whole, this review comes from a pharma perspective, and seeks to provide an evaluation of where CIVMs are poised for maximum impact in the drug development process, and a roadmap for realizing that potential.


Subject(s)
Organoids , Humans , Animals , Organoids/drug effects , Organoids/metabolism , Drug Evaluation, Preclinical , Drug Industry
15.
Int J Mol Sci ; 25(14)2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39063166

ABSTRACT

Mammalian embryos are very vulnerable to environmental toxicants (ETs) exposure. Bisphenol A (BPA), one of the most diffused ETs, exerts endocrine-disrupting effects through estro-gen-mimicking and hormone-like properties, with detrimental health effects, including on reproduction. However, its impact during the peri-implantation stages is still unclear. This study, using gastruloids as a 3D stem cell-based in vitro model of embryonic development, showed that BPA exposure arrests their axial elongation when present during the Wnt/ß-catenin pathway activation period by ß-catenin protein reduction. Gastruloid reshaping might have been impeded by the downregulation of Snail, Slug and Twist, known to suppress E-cadherin expression and to activate the N-cadherin gene, and by the low expression of the N-cadherin protein. Also, the lack of gastruloids elongation might be related to altered exit of BPA-exposed cells from the pluripotency condition and their following differentiation. In conclusion, here we show that the inhibition of gastruloids' axial elongation by BPA might be the result of the concomitant Wnt/ß-catenin perturbation, reduced N-cadherin expression and Oct4, T/Bra and Cdx2 altered patter expression, which all together concur in the impaired development of mouse gastruloids.


Subject(s)
Benzhydryl Compounds , Phenols , Wnt Signaling Pathway , beta Catenin , Animals , Phenols/toxicity , Benzhydryl Compounds/toxicity , Mice , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , beta Catenin/genetics , Cadherins/metabolism , Cadherins/genetics , Organoids/metabolism , Organoids/drug effects , Gene Expression Regulation, Developmental/drug effects , Embryonic Development/drug effects , Cell Differentiation/drug effects , Endocrine Disruptors/toxicity
16.
Front Immunol ; 15: 1418703, 2024.
Article in English | MEDLINE | ID: mdl-39044831

ABSTRACT

Introduction: Salivary gland dysfunction, often resulting from salivary gland obstruction-induced inflammation, is a prevalent condition. Corticosteroid, known for its anti-inflammatory and immunomodulatory properties, is commonly prescribed in clinics. This study investigates the therapeutic implications and potential side effects of dexamethasone on obstructive sialadenitis recovery using duct ligation mice and salivary gland organoid models. Methods: Functional and pathological changes were assessed after administering dexamethasone to the duct following deligation 2 weeks after maintaining ligation of the mouse submandibular duct. Additionally, lipopolysaccharide- and tumor necrosis factor-induced salivary gland organoid inflammation models were established to investigate the effects and underlying mechanisms of action of dexamethasone. Results: Dexamethasone administration facilitated SG function restoration, by increasing salivary gland weight and saliva volume while reducing saliva lag time. Histological evaluation revealed, reduced acinar cell atrophy and fibrosis with dexamethasone treatment. Additionally, dexamethasone suppressed pro-inflammatory cytokines IL-1ß and TNF expression. In a model of inflammation in salivary gland organoids induced by inflammatory substances, dexamethasone restored acinar markers such as AQP5 gene expression levels, while inhibiting pro-inflammatory cytokines TNF and IL6, as well as chemokines CCL2, CXCL5, and CXCL12 induction. Macrophages cultured in inflammatory substance-treated media from salivary gland organoid cultures exhibited pro-inflammatory polarization. However, treatment with dexamethasone shifted them towards an anti-inflammatory phenotype by reducing M1 markers (Tnf, Il6, Il1b, and Cd86) and elevating M2 markers (Ym1, Il10, Cd163, and Klf4). However, high-dose or prolonged dexamethasone treatment induced acino-ductal metaplasia and had side effects in both in vivo and in vitro models. Conclusions: Our findings suggest the effectiveness of corticosteroids in treating obstructive sialadenitis-induced salivary gland dysfunction by regulating pro-inflammatory cytokines.


Subject(s)
Dexamethasone , Kruppel-Like Factor 4 , Sialadenitis , Animals , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Dexamethasone/administration & dosage , Mice , Sialadenitis/drug therapy , Sialadenitis/pathology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , Organoids/drug effects , Cytokines/metabolism , Mice, Inbred C57BL , Salivary Glands/drug effects , Salivary Glands/pathology , Salivary Glands/metabolism , Salivary Glands/immunology , Aquaporin 5/metabolism , Aquaporin 5/genetics , Male , Macrophages/drug effects , Macrophages/immunology , Acinar Cells/drug effects , Acinar Cells/metabolism , Acinar Cells/pathology , Humans
17.
PLoS One ; 19(7): e0307818, 2024.
Article in English | MEDLINE | ID: mdl-39058662

ABSTRACT

INTRODUCTION: High grade astrocytic glioma (HGG) is a lethal solid malignancy with high recurrence rates and limited survival. While several cytotoxic agents have demonstrated efficacy against HGG, drug sensitivity testing platforms to aid in therapy selection are lacking. Patient-derived organoids (PDOs) have been shown to faithfully preserve the biological characteristics of several cancer types including HGG, and coupled with the experimental-analytical hybrid platform Quadratic Phenotypic Optimization Platform (QPOP) which evaluates therapeutic sensitivity at a patient-specific level, may aid as a tool for personalized medical decisions to improve treatment outcomes for HGG patients. METHODS: This is an interventional, non-randomized, open-label study, which aims to enroll 10 patients who will receive QPOP-guided chemotherapy at the time of first HGG recurrence following progression on standard first-line therapy. At the initial presentation of HGG, tumor will be harvested for primary PDO generation during the first biopsy/surgery. At the point of tumor recurrence, patients will be enrolled onto the main study to receive systemic therapy as second-line treatment. Subjects who undergo surgery at the time of recurrence will have a second harvest of tissue for PDO generation. Established PDOs will be subject to QPOP analyses to determine their therapeutic sensitivities to specific panels of drugs. A QPOP-guided treatment selection algorithm will then be used to select the most appropriate drug combination. The primary endpoint of the study is six-month progression-free survival. The secondary endpoints include twelve-month overall survival, RANO criteria and toxicities. In our radiological biomarker sub-study, we plan to evaluate novel radiopharmaceutical-based neuroimaging in determining blood-brain barrier permeability and to assess in vivo drug effects on tumor vasculature over time. TRIAL REGISTRATION: This trial was registered on 8th September 2022 with ClinicalTrials.gov Identifier: NCT05532397.


Subject(s)
Brain Neoplasms , Neoplasm Recurrence, Local , Humans , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/diagnostic imaging , Astrocytoma/drug therapy , Astrocytoma/pathology , Astrocytoma/diagnostic imaging , Organoids/drug effects , Organoids/pathology , Organoids/diagnostic imaging , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasm Grading
18.
Anticancer Res ; 44(8): 3481-3491, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060051

ABSTRACT

BACKGROUND/AIM: Precision medicine aims to revolutionize healthcare by tailoring treatment regimens. This study aimed to integrate comprehensive tumor genomic profiling (CTGP) by targeted-gene panel sequencing and drug screening by circulating tumor cell-derived organoids (CTOs) into clinical practice for the treatment of gastrointestinal (GI) cancers. PATIENTS AND METHODS: Nine patients with various GI cancers underwent CTGP and CTO drug sensitivity testing. CTGP results guided targeted therapy and immunotherapy, while CTO drug sensitivity predicted response to chemotherapy and targeted agents. The drug recommendations from two platforms were correlated with the treatment response to the suggested medications retrospectively. RESULTS: Five patients received therapies aligned with CTGP, including HER2-targeted treatment, immunotherapy, and BRAF/MEK dual inhibition, showing positive responses. CTO drug sensitivity predicted progression under regorafenib (low potential benefit) and good response to chemotherapy with high potential benefit. The combination of CTGP and CTO drug sensitivity may exhibit significant correlation with clinical outcomes during treatment with candidate drugs, demonstrating a sensitivity of 79% and an accuracy of 75%. This encompasses various treatment modalities, such as chemotherapy, targeted therapy, and immunotherapy. CONCLUSION: The present investigation elucidated the integration of CTGP and CTO drug sensitivity screening into clinical practice in a complementary manner, showcasing a significant correlation between treatment response and testing outcomes. Additional prospective evaluation of these two testing modalities in a large cohort is warranted to confirm whether the inclusion of CTO drug sensitivity screening confers enhanced survival benefits compared to utilizing CTGP alone.


Subject(s)
Gastrointestinal Neoplasms , Neoplastic Cells, Circulating , Organoids , Humans , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/pathology , Gastrointestinal Neoplasms/blood , Female , Male , Organoids/pathology , Organoids/drug effects , Middle Aged , Aged , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/drug effects , Precision Medicine/methods , Genomics/methods , Retrospective Studies , Gene Expression Profiling/methods , Biomarkers, Tumor/genetics , Molecular Targeted Therapy/methods , Adult , Drug Screening Assays, Antitumor/methods
19.
Biomolecules ; 14(7)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39062508

ABSTRACT

Many drug and therapeutic modalities have emerged over the past few years. However, successful commercialization is dependent on their safety and efficacy evaluations. Several preclinical models are available for drug-screening and safety evaluations, including cellular- and molecular-level models, tissue and organoid models, and animal models. Organoids are three-dimensional cell cultures derived from primary tissues or stem cells that are structurally and functionally similar to the original organs and can self-renew, and they are used to establish various disease models. Human hepatobiliary organoids have been used to study the pathogenesis of diseases, such as hepatitis, liver fibrosis, hepatocellular carcinoma, primary sclerosing cholangitis and biliary tract cancer, as they retain the physiological and histological characteristics of the liver and bile ducts. Here, we review recent research progress in validating drug toxicity, drug screening and personalized therapy for hepatobiliary-related diseases using human hepatobiliary organoid models, discuss the challenges encountered in current research and evaluate the possible solutions.


Subject(s)
Drug Evaluation, Preclinical , Liver , Organoids , Humans , Organoids/drug effects , Organoids/pathology , Drug Evaluation, Preclinical/methods , Liver/drug effects , Liver/pathology , Animals , Biliary Tract/drug effects , Biliary Tract/pathology
20.
Sci Total Environ ; 947: 174478, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-38964381

ABSTRACT

Perfluorooctane sulfonate (PFOS), a class of synthetic chemicals detected in various environmental compartments, has been associated with dysfunctions of the human central nervous system (CNS). However, the underlying neurotoxicology of PFOS exposure is largely understudied due to the lack of relevant human models. Here, we report bioengineered human midbrain organoid microphysiological systems (hMO-MPSs) to recapitulate the response of a fetal human brain to multiple concurrent PFOS exposure conditions. Each hMO-MPS consists of an hMO on a fully 3D printed holder device with a perfusable organoid adhesion layer for enhancing air-liquid interface culturing. Leveraging the unique, simply-fabricated holder devices, hMO-MPSs are scalable, easy to use, and compatible with conventional well-plates, and allow easy transfer onto a multiple-electrode array (MEA) system for plug-and-play measurement of neural activity. Interestingly, the neural activity of hMO-MPSs initially increased and subsequently decreased by exposure to a concentration range of 0, 30, 100, to 300 µM of PFOS. Furthermore, PFOS exposure impaired neural development and promoted neuroinflammation in the engineered hMO-MPSs. Along with PFOS, our platform is broadly applicable for studies toxicology of various other environmental pollutants.


Subject(s)
Alkanesulfonic Acids , Fluorocarbons , Mesencephalon , Organoids , Fluorocarbons/toxicity , Humans , Alkanesulfonic Acids/toxicity , Organoids/drug effects , Mesencephalon/drug effects , Environmental Pollutants/toxicity , Pregnancy , Female , Microphysiological Systems
SELECTION OF CITATIONS
SEARCH DETAIL