1
|
Nicholls LA, Zeile KA, Scotto LD, Ryznar RJ. Timing of dietary effects on the epigenome and their potential protective effects against toxins. Epigenetics 2025; 20:2451495. [PMID: 39825851 DOI: 10.1080/15592294.2025.2451495] [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: 04/15/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/20/2025] Open
Abstract
Exposure to toxins causes lasting damaging effects on the body. Numerous studies in humans and animals suggest that diet has the potential to modify the epigenome and these modifications can be inherited transgenerationally, but few studies investigate how diet can protect against negative effects of toxins. Potential evidence in the primary literature supports that caloric restriction, high-fat diets, high protein-to-carbohydrate ratios, and dietary supplementation protect against environmental toxins and strengthen these effects on their offspring's epigenome. Most notably, the timing when dietary interventions are given - during a parent's early development, pregnancy, and/or lifetime - result in similar transgenerational epigenetic durations. This implies the existence of multiple opportunities to strategically fortify the epigenome. This narrative review explores how to best utilize dietary modifications to modify the epigenome to protect future generations against negative health effects of persistent environmental toxins. Furthermore, by suggesting an ideal diet with specific micronutrients, macronutrients, and food groups, epigenetics can play a key role in the field of preventive medicine. Based on these findings, longitudinal research should be conducted to determine if a high protein, high-fat, and low-carbohydrate diet during a mother's puberty or pregnancy can epigenetically protect against alcohol, tobacco smoke, and air pollution across multiple generations.
Collapse
Affiliation(s)
- Lynnea A Nicholls
- Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
| | - Kendall A Zeile
- Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
| | - London D Scotto
- Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
| | - Rebecca J Ryznar
- Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
- Department of Biomedical Sciences, Rocky Vista University College of Osteopathic Medicine, Parker, CO, USA
| |
Collapse
|
2
|
Feng L, Fu X, Du Z, Guo Y, Zhuo L, Yang Y, Cao D, Yao X. MultiCTox: Empowering Accurate Cardiotoxicity Prediction through Adaptive Multimodal Learning. J Chem Inf Model 2025; 65:3517-3528. [PMID: 40145660 DOI: 10.1021/acs.jcim.5c00022] [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: 03/28/2025]
Abstract
Cardiotoxicity refers to the inhibitory effects of drugs on cardiac ion channels. Accurate prediction of cardiotoxicity is crucial yet challenging, as it directly impacts the evaluation of cardiac drug efficacy and safety. Numerous methods have been developed to predict cardiotoxicity, yet their performance remains limited. A key limitation is that these methods often rely solely on single-modal data, making multimodal data integration challenging. As a result, we present a multimodal method integrating molecular SMILES, structure, and fingerprint to enhance cardiotoxicity prediction. First, we designed a fusion layer to unify representations from different modalities. During training, the model maximizes intramodal similarity for the same molecule while minimizing intermolecular similarity, ensuring consistent cross-modal representations. This study evaluates the inhibitory effects of candidate drugs on voltage-gated potassium (hERG), sodium (Nav1.5), and calcium (Cav1.2) channels. Experimental results demonstrate that the proposed model significantly outperforms existing state-of-the-art methods in cardiotoxicity prediction. We anticipate that this model will contribute significantly to the development and safety evaluation of cardiac drugs, reducing cardiotoxicity-related risks.
Collapse
Affiliation(s)
- Lin Feng
- School of Data Science and Artificial Intelligence, Wenzhou University of Technology, Wenzhou 325027, China
| | - Xiangzheng Fu
- College of Information Science and Engineering, Hunan University, Changsha 410000, China
| | - Zhenya Du
- School of Nursing, Teaching and Research Department of Public Medical Courses, Guangzhou xinhua University, Guangzhou 510520, China
| | - Yuting Guo
- School of Data Science and Artificial Intelligence, Wenzhou University of Technology, Wenzhou 325027, China
| | - Linlin Zhuo
- School of Data Science and Artificial Intelligence, Wenzhou University of Technology, Wenzhou 325027, China
| | - Yan Yang
- School of Data Science and Artificial Intelligence, Wenzhou University of Technology, Wenzhou 325027, China
| | - Dongsheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410003, China
| | - Xiaojun Yao
- Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China
| |
Collapse
|
3
|
Santos AM, Vieira EM, de Jesus JR, Santana Júnior CC, Nascimento Júnior JAC, Oliveira AMS, Araújo AADS, Picot L, Alves IA, Serafini MR. Development and characterization of farnesol complexed in β- and hydroxypropyl-β-cyclodextrin and their antibacterial activity. Carbohydr Res 2025; 550:109406. [PMID: 39864120 DOI: 10.1016/j.carres.2025.109406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Farnesol (FAR) belongs to terpenes group and is a sesquiterpene alcohol and a hydrophobic compound, which can be extracted from natural sources or obtained by organic chemical or biological synthesis. Recent advances in the field of nanotechnology allow the drawbacks of low drug solubility, which can improve the drug therapeutic index. Therefore, this study aimed to prepare the FAR inclusion complexes with β-cyclodextrin (β-CD) and hydroxypropyl-β-cyclodextrin (HP-β-CD) through freeze-drying method, proposing their physicochemical characterization, comparing their toxicity, and evaluating their in vitro antibacterial activity. Initially, physical mixture and freeze-dried inclusion complexes of FAR/β-CD and FAR/HP-β-CD were obtained in the molar ratio (1:1). The samples were characterized by DSC, TG/DTG, FTIR, PXRD, SEM, pHPZC, and the complexation efficiency were performed by HPLC. In vivo toxicity assay was performed using Tenebrio molitor larvae to determine the LD50 and toxic dose of the samples. Also, it was proposed that the evaluation of the fluorescence suppression of Bovine Serum Albumin and the antibacterial activity. The complexation of FAR was evidenced with β-CD and HP-β-CD by the characterization techniques analyzed. The complexation efficiency of FAR/β-CD and FAR/HP-β-CD were 73,53 % and 74.12 %, respectively. The inclusion complexes demonstrated a reduction in toxicity, as evidenced by lower toxic and LD50 doses compared to the free FAR. The inclusion complexes induced conformational changes in BSA, suggesting that they reached the subdomains containing tryptophan residues. In terms of antibacterial activity, FAR/β-CD and FAR/HP-β-CD did not exhibit significant MIC results compared to free FAR, except for FAR/HP-β-CD against S. aureus ATCC 25923.
Collapse
Affiliation(s)
- Anamaria Mendonça Santos
- Postgraduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil.
| | - Edileuza Marcelo Vieira
- Research Laboratory in Biomaterials, LPBio, Department of Chemistry, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Jemmyson Romário de Jesus
- Research Laboratory in Biomaterials, LPBio, Department of Chemistry, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | | | | | - Ana Maria Santos Oliveira
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | | | - Laurent Picot
- La Rochelle Université, UMR CNRS 7266 LIENSs, La Rochelle, France
| | - Izabel Almeida Alves
- Postgraduate Program in Pharmaceutical Sciences, University of the State of Bahia and Faculty of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Mairim Russo Serafini
- Postgraduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil; Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil.
| |
Collapse
|
4
|
Boonsom S, Chamnansil P, Boonseng S, Srisongkram T. ToxSTK: A multi-target toxicity assessment utilizing molecular structure and stacking ensemble learning. Comput Biol Med 2025; 185:109480. [PMID: 39644580 DOI: 10.1016/j.compbiomed.2024.109480] [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: 09/05/2024] [Revised: 11/04/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
Drug registration requires risk assessment of new active pharmaceutical ingredients or excipients to ensure they are safe for human health and the environment. However, traditional risk assessment is expensive and relies heavily on animal testing. Machine learning (ML) has been used as a risk assessment tool, providing less time, money, and involved animals than in vivo experiments. Despite that, the ML models often rely on a single model, which may introduce bias and unreliable prediction. Stacking ensemble learning is an ML framework that makes predictions based on multimodal outcomes. This framework performs well in quantitative structure-activity relationship (QSAR) studies. In this study, we developed ToxSTK, a multi-target toxicity assessment using stacking ensemble learning. We aimed to create an ML tool that facilitates toxicity assessments more affordably with reduced reliance on animal models. We focused on four key targets generally assessed in early-stage drug development: hERG toxicity, mTOR toxicity, PBMCs toxicity, and mutagenicity. Our model integrated 12 molecular fingerprints with 3 ML algorithms, generating 36 novel predictive features (PFs). These PFs were then combined to construct the final meta-decision model. Our results demonstrated that the ToxSTK model surpasses standard regression and classification metrics, ensuring it is highly reliable and accurate in predicting chemical toxicities within its application domain. This model passed the y-randomization test, confirming that the identified QSAR is robust and not due to random chance. Additionally, this model outperforms the existing ML methods for these endpoints, suggesting its effectiveness for risk assessment applications. We recommend incorporating this stacking ensemble learning framework into the chemical risk assessment pipeline to improve model generalization, accuracy, robustness, and reliability.
Collapse
Affiliation(s)
- Surapong Boonsom
- Department of Chemistry, Mahidol Wittayanusorn School, Phutthamonthon, Nakhon Pathom, Thailand
| | - Panisara Chamnansil
- Department of Chemistry, Mahidol Wittayanusorn School, Phutthamonthon, Nakhon Pathom, Thailand
| | - Sarote Boonseng
- Department of Chemistry, Mahidol Wittayanusorn School, Phutthamonthon, Nakhon Pathom, Thailand
| | - Tarapong Srisongkram
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Thailand.
| |
Collapse
|
5
|
Agarwal S, Bochkova J, Mohamed MK, Schaefer ML, Zhou A, Skinner J, Johns RA. Disruption of Extracellular Signal-Regulated Kinase Partially Mediates Neonatal Isoflurane Anesthesia-Induced Changes in Dendritic Spines and Cognitive Function in Juvenile Mice. Int J Mol Sci 2025; 26:981. [PMID: 39940749 PMCID: PMC11817073 DOI: 10.3390/ijms26030981] [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: 12/23/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
There is a growing concern worldwide about the potential harmful effects of anesthesia on brain development, based on studies in both humans and animals. In infants, repeated anesthesia exposure is linked to learning disabilities and attention disorders. Similarly, laboratory studies in mice show that neonates exposed to general anesthesia experience long-term cognitive and behavioral impairments. Inhaled anesthetics affect the postsynaptic density (PSD)-95, discs large homolog, and zona occludens-1 (PDZ) domains. The disruption of the synaptic PSD95-PDZ2 domain-mediated protein interactions leads to a loss of spine plasticity and cognitive deficits in juvenile mice. The nitric oxide-mediated protein kinase-G signaling pathway enhances synaptic plasticity also by activating extracellular signal-regulated kinase, which subsequently phosphorylates cAMP-response element binding protein, a crucial transcription factor for memory formation. Exposure to isoflurane or postsynaptic density-95-PDZ2-wildtype peptides results in decreased levels of phosphorylated extracellular signal-regulated kinase (p-ERK) and phosphorylated cAMP-response element binding protein (p-CREB), which are critical for synaptic plasticity and memory formation. Pizotifen treatment after isoflurane or postsynaptic density-95-PDZ2-wildtype peptide exposure in mice prevented decline in p-ERK levels, preserved learning and memory functions at 5 weeks of age, and maintained mushroom spine density at 7 weeks of age. Protein kinase-G activation by components of the nitric oxide signaling pathway leads to the stabilization of dendritic spines and synaptic connections. Concurrently, the ERK/CREB pathway, which is crucial for synaptic plasticity and memory consolidation, is supported and maintained by pizotifen, thereby preventing cognitive deficits caused in response to isoflurane or postsynaptic density-95-PDZ2-wildtype peptide exposure. Activation of ERK signaling cascade by pizotifen helps to prevent cognitive impairment and spine loss in response to postsynaptic density-95-PDZ2 domain disruption.
Collapse
Affiliation(s)
- Swati Agarwal
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (S.A.); (M.L.S.); (J.S.)
| | - Jacqueline Bochkova
- Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA; (J.B.); (M.K.M.); (A.Z.)
| | - Mazen K. Mohamed
- Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA; (J.B.); (M.K.M.); (A.Z.)
| | - Michele L. Schaefer
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (S.A.); (M.L.S.); (J.S.)
| | - Annika Zhou
- Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA; (J.B.); (M.K.M.); (A.Z.)
| | - John Skinner
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (S.A.); (M.L.S.); (J.S.)
| | - Roger A. Johns
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (S.A.); (M.L.S.); (J.S.)
| |
Collapse
|
6
|
Kannan S, Hammamieh R, Chakraborty N. A Machine Learning Pipeline to Screen Large In Vivo Molecular Data to Curate Disease Signatures of High Translational Potential. Methods Mol Biol 2025; 2880:331-344. [PMID: 39900768 DOI: 10.1007/978-1-0716-4276-4_17] [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: 02/05/2025]
Abstract
A significantly low success rate of human clinical studies has long been attributed to a capability gap, namely, an ineffective translation of the animal data to the human context. To bridge this capability gap, several correcting measures have been evaluated; using a strict guideline to select animal models for a given disease and implementing alternative models such as tissues-on-chip are some of them. Current hypothesis tells that there is a basic similarity in responding to a stress between human and those mammals that precede human in the phylogenetic tree; however, the corresponding molecular mechanisms are not exactly the same across these species. Therefore, strategic manipulations are necessary to curate those candidates from animal data that would have high translational potential. Hence, we developed an analytical tool that can screen the in vivo results, such as genomic, proteomic, epigenomic data with two primary objectives. The first objective is to identify those molecules that are sequentially conserved across the phylogenetic tree. The second objective is to find those molecules that would similarly perturb across the phylogenetic tree in responding to a stress of interest. A machine learning (ML) algorithm converges these two sets of molecules to curate the common features, which would demonstrate phylogenetic homology in their molecular makeups and characteristic similarity across the phylogenetic tree. This ML-pipeline would be most beneficial in those scenarios, such as the rare diseases or chemical-biological-radiation-nuclear (CBRN)-exposed samples, where the inventory of human samples is minimum. This strategy is surely at a risk in overlooking the human-exclusive signatures; nevertheless, this ML-approach is poised to refine the animal data to generate results of high translational potential with minimum false positive and false negative entries.
Collapse
Affiliation(s)
- Swapna Kannan
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Rasha Hammamieh
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Nabarun Chakraborty
- Medical Readiness Systems Biology, CMPN, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| |
Collapse
|
7
|
Culiat C, Soni D, Malkes W, Wienhold M, Zhang LH, Henry E, Dragan M, Kar S, Angeles DM, Eaker S, Biswas R. NELL1 variant protein (NV1) modulates hyper-inflammation, Th-1 mediated immune response, and the HIF-1α hypoxia pathway to promote healing in viral-induced lung injury. Biochem Biophys Res Commun 2025; 744:151198. [PMID: 39706056 DOI: 10.1016/j.bbrc.2024.151198] [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: 10/16/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Research underscores the urgent need for technological innovations to treat lung tissue damage from viral infections and the lasting impact of COVID-19. Our study demonstrates the effectiveness of recombinant human NV1 protein in promoting a pro-healing extracellular matrix that regulates homeostasis in response to excessive tissue reactions caused by infection and injury. NV1 achieves this by calibrating multiple biological mechanisms, including reducing hyperinflammatory cytokine levels (e.g., IFN-γ, TNF-α, IL-10, and IP-10), enhancing the production of proteins involved in viral inactivation and clearance through endocytosis and phagocytosis (e.g., IL-9, IL-1α), regulating pro-clotting and thrombolytic pathways (e.g., downregulates SERPINE 1 and I-TAC during Th1-mediated inflammation), maintaining cell survival under hypoxic conditions via HIF-1α regulation through the M3K5-JNK-AP-1 and TSC2-mTOR pathways, and promoting blood vessel formation. Our findings reveal NV1 as a potential therapeutic candidate for treating severe lung injuries caused by inflammatory and hypoxic conditions from viral infections and related diseases.
Collapse
Affiliation(s)
| | - Dharmendra Soni
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | | | - Mark Wienhold
- NellOne Therapeutics Inc., Knoxville, TN, 37931, USA
| | | | | | | | | | | | - Shannon Eaker
- NellOne Therapeutics Inc., Knoxville, TN, 37931, USA
| | - Roopa Biswas
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| |
Collapse
|
8
|
Goyal K, Afzal M, Altamimi ASA, Babu MA, Ballal S, Kaur I, Kumar S, Kumar MR, Chauhan AS, Ali H, Shahwan M, Gupta G. Chronic kidney disease and aging: dissecting the p53/p21 pathway as a therapeutic target. Biogerontology 2024; 26:32. [PMID: 39725742 DOI: 10.1007/s10522-024-10173-z] [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: 11/09/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024]
Abstract
Chronic kidney diseases (CKD) are a group of multi-factorial disorders that markedly impair kidney functions with progressive renal deterioration. Aging contributes to age-specific phenotypes in kidneys, which undergo several structural and functional alterations, such as a decline in regenerative capacity and increased fibrosis, inflammation, and tubular atrophy, all predisposing them to disease and increasing their susceptibility to injury while impeding their recovery. A central feature of these age-related processes is the activation of the p53/p21 pathway signaling. The pathway is a key player in cellular senescence, apoptosis, and cell cycle regulation, which are all key to maintaining the health of the kidney. P53 is a transcription factor and a tumor suppressor protein that responds to cell stress and damage. Persistent activation of cell p53 can lead to the expression of p21, an inhibitor of the cell cycle known as a cyclin-dependent kinase. This causes cells to cease dividing and leads to senescence, where cells can no longer increase. The accumulation of senescent cells in the aging kidney impairs kidney function by altering the microenvironment. As the number of senescent cells increases, the capacity of the kidney to recover from injury decreases, accelerating the progression of end-stage renal disease. This article review extensively explores the relationship between the p53/p21 pathway and cellular senescence within an aging kidney and the emerging therapeutic strategies that target it to overcome the impacts of cellular senescence on CKD.
Collapse
Affiliation(s)
- Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | | | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - M Ravi Kumar
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab, 140307, India
| | - Ashish Singh Chauhan
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Haider Ali
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Moyad Shahwan
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
9
|
Zhao Y, Zhang C, Liang C, Lv X, Zhou D, Deng Y, Zhang R. On Multicell-Interaction Chip: In Situ Observing the Interactions between the Astrocytes with Lysosomal Dysfunction and BBB Cells. Anal Chem 2024; 96:20057-20064. [PMID: 39642021 DOI: 10.1021/acs.analchem.4c04893] [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: 12/08/2024]
Abstract
Lysosomes in astrocytes play vital roles in toxic protein degradation in the brain. Lysosomal dysfunction can lead to abnormal protein deposits, which further induce damage to neurons and the blood-brain barrier (BBB), and thereby affect the interaction between the nervous and vascular systems. Therefore, investigating the interactions between astrocytes with lysosomal dysfunction and BBB cells is of significant importance. However, the lack of effective in vitro models hinders the study of this complex system. Herein, an 8-well arrayed microfence multicell interculture chip (AMMIC) with a hydrophilically optimized surface is introduced for investigating the interactions between astrocytes and BBB cells. Then, a novel lysosome-targeted photosensitizer, IVQ-2Br, is synthesized for inducing controllable oxidative stress damage in the lysosomes of astrocytes. By the combination of the 8-well AMMIC and IVQ-2Br, a model for studying the interactions between astrocytes with lysosomal dysfunction and BBB cells has been constructed. Particularly, severe secondary injuries to BBB cells brought about by oxidative stress, including alterations in cell morphology and activity as well as notable DNA damage, are in situ observed on the 8-well AMMIC. The mediators involved in this oxidative stress injury-mediated intercellular communication are validated to be reactive oxygen species (ROS) and exosomes. This work not only presents an in vitro modeling method for studying cell-cell interactions but also demonstrates the potential of in vitro models constructed through the integration of complex microfluidic chip techniques and photosensitizers for advancing biomedical research.
Collapse
Affiliation(s)
- Yimeng Zhao
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Chen Zhang
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Chaohui Liang
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Xuefei Lv
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Di Zhou
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Yulin Deng
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| | - Ruoyao Zhang
- School of Medical Technology, Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
10
|
Winner G J, Jain S, Gupta D. Unveiling novel molecules and therapeutic targets in hypertension - A narrative review. Eur J Pharmacol 2024; 984:177053. [PMID: 39393666 DOI: 10.1016/j.ejphar.2024.177053] [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: 04/25/2024] [Revised: 09/18/2024] [Accepted: 10/07/2024] [Indexed: 10/13/2024]
Abstract
Hypertension is a prevalent non-communicable disease with serious cardiovascular complications, including heart failure, myocardial infarction, and stroke, often resulting from uncontrolled hypertension. While current treatments primarily target the renin-angiotensin-aldosterone pathway, the therapeutic response remains modest in many patients, with some developing resistant hypertension. Newer therapeutic approaches aim to address hypertension from various aspects beyond conventional drugs, including targeting central nervous system pathways, inflammatory pathways, vascular smooth muscle function, and baroreceptors. Despite these advancements, each therapy faces unique clinical and mechanistic challenges that influence its clinical translatability and long-term viability. This review explores the mechanisms of novel molecules in preclinical and clinical development, highlights potential therapeutic targets, and discusses the challenges and ethical considerations related to hypertension therapeutics and their development.
Collapse
Affiliation(s)
| | - Surbhi Jain
- Aligarh Muslim University, Uttar Pradesh, India
| | | |
Collapse
|
11
|
Tagaras N, Song H, Sahar S, Tong W, Mao Z, Buerki‐Thurnherr T. Safety Landscape of Therapeutic Nanozymes and Future Research Directions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407816. [PMID: 39445544 PMCID: PMC11633477 DOI: 10.1002/advs.202407816] [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: 07/10/2024] [Revised: 09/20/2024] [Indexed: 10/25/2024]
Abstract
Oxidative stress and inflammation are at the root of a multitude of diseases. Treatment of these conditions is often necessary but current standard therapies to fight excessive reactive oxygen species (ROS) and inflammation are often ineffective or complicated by substantial safety concerns. Nanozymes are emerging nanomaterials with intrinsic enzyme-like properties that hold great promise for effective cancer treatment, bacterial elimination, and anti-inflammatory/anti-oxidant therapy. While there is rapid progress in tailoring their catalytic activities as evidenced by the recent integration of single-atom catalysts (SACs) to create next-generation nanozymes with superior activity, selectivity, and stability, a better understanding and tuning of their safety profile is imperative for successful clinical translation. This review outlines the current applied safety assessment approaches and provides a comprehensive summary of the safety knowledge of therapeutic nanozymes. Overall, nanozymes so far show good in vitro and in vivo biocompatibility despite considerable differences in their composition and enzymatic activities. However, current safety investigations mostly cover a limited set of basic toxicological endpoints, which do not allow for a thorough and deep assessment. Ultimately, remaining research gaps that should be carefully addressed in future studies are highlighted, to optimize the safety profile of therapeutic nanozymes early in their pre-clinical development.
Collapse
Affiliation(s)
- Nikolaos Tagaras
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
- Department of Health Sciences and TechnologyETH ZurichZurich8093Switzerland
| | - Haihan Song
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University866 Yuhangtang RdHangzhou310058China
| | - Shafaq Sahar
- College of Chemical and Biological EngineeringMOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University866 Yuhangtang RdHangzhou310058China
| | - Weijun Tong
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University866 Yuhangtang RdHangzhou310058China
| | - Zhengwei Mao
- College of Chemical and Biological EngineeringMOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University866 Yuhangtang RdHangzhou310058China
| | - Tina Buerki‐Thurnherr
- Laboratory for Particles‐Biology InteractionsSwiss Federal Laboratories for Materials Science and Technology (Empa)St. Gallen9014Switzerland
| |
Collapse
|
12
|
Corbalan JJ, Jagadeesan P, Frietze KK, Taylor R, Gao GL, Gallagher G, Nickels JT. Humanized monoacylglycerol acyltransferase 2 mice develop metabolic dysfunction-associated steatohepatitis. J Lipid Res 2024; 65:100695. [PMID: 39505262 DOI: 10.1016/j.jlr.2024.100695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Mice lacking monoacylglycerol acyltransferase 2 (mMGAT21) are resistant to diet-induced fatty liver, suggesting hMOGAT2 inhibition is a viable option for treating metabolic dysfunction-associated steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH). We generated humanized hMOGAT2 mice (HuMgat2) for use in pre-clinical studies testing the efficacy of hMOGAT2 inhibitors for treating MASLD/MASH. HuMgat2 mice developed MASH when fed a steatotic diet. Computer-aided histology revealed the presence of hepatocyte cell ballooning, immune cell infiltration, and fibrosis. Hepatocytes accumulated Mallory-Denk bodies containing phosphorylated p62/sequestosome-1-ubiquitinated protein aggregates likely caused by defects in autophagy. Metainflammation and apoptotic cell death were seen in the livers of HuMgat2 mice. Treating HuMgat2 mice with elafibranor reduced several MASH phenotypes. RNASeq analysis predicted changes in bile acid transporter expression that correlated with altered bile acid metabolism indicative of cholestasis. Our results suggest that HuMgat2 mice will serve as a pre-clinical model for testing hMOGAT2 inhibitor efficacy and toxicity and allow for the study of hMOGAT2 in the context of MASH.
Collapse
Affiliation(s)
- J Jose Corbalan
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Pranavi Jagadeesan
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Karla K Frietze
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Rulaiha Taylor
- Department of Pharmacology and Toxicology, Earnest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Grace L Gao
- Department of Pharmacology and Toxicology, Earnest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, USA
| | - Grant Gallagher
- Oncoveda, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Joseph T Nickels
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
13
|
Lee SJ, Jeong W, Atala A. 3D Bioprinting for Engineered Tissue Constructs and Patient-Specific Models: Current Progress and Prospects in Clinical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408032. [PMID: 39420757 PMCID: PMC11875024 DOI: 10.1002/adma.202408032] [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: 06/05/2024] [Revised: 09/24/2024] [Indexed: 10/19/2024]
Abstract
Advancements in bioprinting technology are driving the creation of complex, functional tissue constructs for use in tissue engineering and regenerative medicine. Various methods, including extrusion, jetting, and light-based bioprinting, have their unique advantages and drawbacks. Over the years, researchers and industry leaders have made significant progress in enhancing bioprinting techniques and materials, resulting in the production of increasingly sophisticated tissue constructs. Despite this progress, challenges still need to be addressed in achieving clinically relevant, human-scale tissue constructs, presenting a hurdle to widespread clinical translation. However, with ongoing interdisciplinary research and collaboration, the field is rapidly evolving and holds promise for personalized medical interventions. Continued development and refinement of bioprinting technologies have the potential to address complex medical needs, enabling the development of functional, transplantable tissues and organs, as well as advanced in vitro tissue models.
Collapse
Affiliation(s)
| | | | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States
| |
Collapse
|
14
|
Polis B, Cuda CM, Putterman C. Animal models of neuropsychiatric systemic lupus erythematosus: deciphering the complexity and guiding therapeutic development. Autoimmunity 2024; 57:2330387. [PMID: 38555866 DOI: 10.1080/08916934.2024.2330387] [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: 02/07/2024] [Accepted: 03/10/2024] [Indexed: 04/02/2024]
Abstract
Systemic lupus erythematosus (SLE) poses formidable challenges due to its multifaceted etiology while impacting multiple tissues and organs and displaying diverse clinical manifestations. Genetic and environmental factors contribute to SLE complexity, with relatively limited approved therapeutic options. Murine models offer insights into SLE pathogenesis but do not always replicate the nuances of human disease. This review critically evaluates spontaneous and induced animal models, emphasizing their validity and relevance to neuropsychiatric SLE (NPSLE). While these models undoubtedly contribute to understanding disease pathophysiology, discrepancies persist in mimicking some NPSLE intricacies. The lack of literature addressing this issue impedes therapeutic progress. We underscore the urgent need for refining models that truly reflect NPSLE complexities to enhance translational fidelity. We encourage a comprehensive, creative translational approach for targeted SLE interventions, balancing scientific progress with ethical considerations to eventually improve the management of NPSLE patients. A thorough grasp of these issues informs researchers in designing experiments, interpreting results, and exploring alternatives to advance NPSLE research.
Collapse
Affiliation(s)
- Baruh Polis
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
| | - Carla M Cuda
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Chaim Putterman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
- Division of Rheumatology and Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
15
|
Imran M, Moyle PM, Kamato D, Mohammed Y. Advances in, and prospects of, 3D preclinical models for skin drug discovery. Drug Discov Today 2024; 29:104208. [PMID: 39396673 DOI: 10.1016/j.drudis.2024.104208] [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: 05/20/2024] [Revised: 09/25/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
The skin has an important role in regulating homeostasis and protecting the body from endogenous and exogenous microenvironments. Although 3D models for drug discovery have been extensively studied, there is a growing demand for more advanced 3D skin models to enhance skin research. The use of these advanced skin models holds promise across domains such as cosmetics, skin disease treatments, and toxicity testing of new therapeutics. Recent advances include the development of skin-on-a-chip, spheroids, reconstructed skin, organoids, and computational approaches, including quantitative structure-activity relationship (QSAR) and quantitative structure-property relationship (QSPR) research. These innovations are bridging the gap between traditional 2D and advanced 3D models, moving progress from research to clinical applications. In this review, we highlight in vitro and computational skin models with advanced drug discovery for skin-related applications.
Collapse
Affiliation(s)
- Mohammad Imran
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Peter Michael Moyle
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia; School of Environment and Science, Institute for Biomedicine and Glycomics, Griffith University, Nathan, QLD 4111, Australia
| | - Yousuf Mohammed
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4102, Australia; School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia.
| |
Collapse
|
16
|
Sinmez CC, Yaşar A, van den Hoven R. A Comparison of European Union and Turkish Reports on the Statistics of Experimental Animal Use in Toxicity Testing. Altern Lab Anim 2024; 52:301-315. [PMID: 39365923 DOI: 10.1177/02611929241286885] [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: 10/06/2024]
Abstract
The data on the number of animals used for experimental purposes, with a particular focus on regulatory toxicity studies, in Turkey and EU member states plus Norway (EU+), were compared for the period 2015 to 2020 (UK data after Brexit for 2020 were included too). The total number of listed vertebrates used for regulatory toxicity testing in the full 6-year reporting period in Turkey was 3.6% of that reported for the EU+. However, these numbers showed an increasing trend over the reporting period in Turkey, while the trend decreased in the EU+. In the top three most-used species for regulatory toxicology purposes in Turkey, 41% were fish (68,758 animals), followed by rats (29%; 49,975 animals), and then mice (20%; 33,292 animals). Turkey used low numbers of cats and dogs, while the corresponding number for the EU+ was more than 70-fold higher. Non-human primates were not used at all in Turkey, whereas about 34,000 were used in EU+ laboratories. The majority of toxicity tests (57%) performed in Turkey were categorised as 'Other' toxicity tests, and 27% were acute and sub-acute toxicity tests. Successful replacement alternatives (e.g. in vitro and in silico approaches) are already used in a wide range of research areas. However, although this new research and testing paradigm is underpinned by scientific evidence, the legislation and pace of acceptance of these new techniques in Turkey is considerably lagging behind other countries.
Collapse
Affiliation(s)
- Cagri Caglar Sinmez
- Department of History of Veterinary Medicine and Deontology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Aşkın Yaşar
- Department of History of Veterinary Medicine and Deontology, Faculty of Veterinary Medicine, Selçuk University, Konya, Turkey
| | - Rene van den Hoven
- Clinic of Equine Internal Medicine, Veterinary University of Vienna, Vienna, Austria
| |
Collapse
|
17
|
Pramudito MA, Fuadah YN, Qauli AI, Marcellinus A, Lim KM. Explainable artificial intelligence (XAI) to find optimal in-silico biomarkers for cardiac drug toxicity evaluation. Sci Rep 2024; 14:24045. [PMID: 39402077 PMCID: PMC11473646 DOI: 10.1038/s41598-024-71169-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 08/26/2024] [Indexed: 10/17/2024] Open
Abstract
The Comprehensive In-vitro Proarrhythmia Assay (CiPA) initiative aims to refine the assessment of drug-induced torsades de pointes (TdP) risk, utilizing computational models to predict cardiac drug toxicity. Despite advancements in machine learning applications for this purpose, the specific contribution of in-silico biomarkers to toxicity risk levels has yet to be thoroughly elucidated. This study addresses this gap by implementing explainable artificial intelligence (XAI) to illuminate the impact of individual biomarkers in drug toxicity prediction. We employed the Markov chain Monte Carlo method to generate a detailed dataset for 28 drugs, from which twelve in-silico biomarkers of 12 drugs were computed to train various machine learning models, including Artificial Neural Networks (ANN), Support Vector Machines (SVM), Random Forests (RF), XGBoost, K-Nearest Neighbors (KNN), and Radial Basis Function (RBF) networks. Our study's innovation is leveraging XAI, mainly through the SHAP (SHapley Additive exPlanations) method, to dissect and quantify the contributions of biomarkers across these models. Furthermore, the model performance was evaluated using the test set from 16 drugs. We found that the ANN model coupled with the eleven most influential in-silico biomarkers namelydVm dt repol , dVm dt max , APD 90 , APD 50 , APD tri , CaD 90 , CaD 50 , Ca tri , Ca Diastole , q I n w a r d , a n d q N e t showed the highest classification performance among all classifiers with Area Under the Curve (AUC) scores of 0.92 for predicting high-risk, 0.83 for intermediate-risk, and 0.98 for low-risk drugs. We also found that the optimal in silico biomarkers selected based on SHAP analysis may be different for various classification models. However, we also found that the biomarker selection only sometimes improved the performance; therefore, evaluating various classifiers is still essential to obtain the desired classification performance. Our proposed method could provide a systematic way to assess the best classifier with the optimal in-silico biomarkers for predicting the TdP risk of drugs, thereby advancing the field of cardiac safety evaluations.
Collapse
Affiliation(s)
- Muhammad Adnan Pramudito
- Computational Medicine Lab, Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, 39177, Republic of Korea
| | - Yunendah Nur Fuadah
- Computational Medicine Lab, Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, 39177, Republic of Korea
- School of Electrical Engineering, Telkom University, Bandung, 40257, Indonesia
| | - Ali Ikhsanul Qauli
- Computational Medicine Lab, Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, 39177, Republic of Korea
- Department of Engineering, Faculty of Advanced Technology and Multidiscipline, Universitas Airlangga, Surabaya, 60115, Jawa Timur, Indonesia
| | - Aroli Marcellinus
- Computational Medicine Lab, Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, 39177, Republic of Korea
| | - Ki Moo Lim
- Computational Medicine Lab, Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, 39177, Republic of Korea.
- Computational Medicine Lab, Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi, 39177, Republic of Korea.
- Meta Heart Co., Ltd., Gumi, 39253, Republic of Korea.
| |
Collapse
|
18
|
Moustakas D, Mani I, Pouliakis A, Iacovidou N, Xanthos T. The Effects of IRL-1620 in Post-ischemic Brain Injury: A Systematic Review and Meta-analysis of Experimental Studies. Neurocrit Care 2024; 41:665-680. [PMID: 38724864 DOI: 10.1007/s12028-024-01994-4] [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: 11/15/2023] [Accepted: 04/02/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND Sovateltide (IRL-1620), an endothelin B receptor agonist, has previously demonstrated neuroprotective and neuroregenerative effects in animal models of acute ischemic stroke. Recently, clinical trials indicated that it could also be effective in humans with stroke. Here, we systematically investigate whether IRL-1620 may be used for the treatment of ischemia-induced brain injury. METHODS A systematic review was performed following the Preferred Reporting Items for Systematic reviews and Meta-Analyses guidelines. MEDLINE (PubMed) and Scopus databases were searched for eligible studies up to December 2022. The databases ClinicalTrials.gov and Pharmazz Inc. were screened for unpublished or ongoing trials. Only studies in English were evaluated for eligibility. Meta-analysis of the included studies was also conducted. RESULTS Finally, seven studies were included in the review, all in animal rat models because of scarcity of clinical trials. Six studies, all in middle cerebral artery occlusion (MCAO) models, were selected for meta-analysis. In the two studies assessing mortality, no deaths were reported in the IRL-1620 group 24 h after MCAO, whereas the vehicle group had almost a five times higher mortality risk (risk ratio 5.3, 95% confidence interval 0.7-40.1, I2 = 0%). In all five studies evaluating outcome on day 7 after MCAO, IRL-1620 was associated with statistically significantly lower neurological deficit and improved motor performance compared with the vehicle. Infract volume, differentiation potential of neuronal progenitor cells, and mitochondrial fate also improved with IRL-1620 administration. CONCLUSIONS According to the above, in animal MCAO models, IRL-1620 enhanced neurogenesis and neuroprotection and improved outcome. Future studies are needed to expand our understanding of its effects in human study participants with acute ischemic stroke as well as in other common causes of cerebral ischemia including cardiac arrest.
Collapse
Affiliation(s)
- Dimitris Moustakas
- Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Iliana Mani
- 2d Department of Internal Medicine, Medical School, Hippokration General Hospital, National and Kapodistrian University of Athens, Vas. Sofias 114, 11527, Athens, Greece.
| | - Abraham Pouliakis
- 2d Department of Pathology, Attikon University General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikoletta Iacovidou
- Neonatal Department, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros Xanthos
- School of Health Sciences, University of West Attica, Athens, Greece
| |
Collapse
|
19
|
Yang E, Lu W, Muñoz-Vergara D, Goldfinger E, Kaptchuk TJ, Napadow V, Ahn AC, Wayne PM. Acupoint Sensitivity in Health and Disease: A Systematic Review. JOURNAL OF INTEGRATIVE AND COMPLEMENTARY MEDICINE 2024; 30:925-939. [PMID: 38748400 DOI: 10.1089/jicm.2023.0204] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2024]
Abstract
Introduction: The concept of acupoints is a key defining feature of acupuncture, yet the scientific basis of acupoints remains unclear. In recent years, there has been an emerging body of animal studies demonstrating an association between cutaneous sensitivity and visceral pathophysiology, through which acupoints over the skin are sensitized in pathologic conditions. Several studies with humans have also been conducted to assess whether the sensitivity of acupoints is distinct in healthy versus clinical populations. However, no systematic review has been conducted to collate and synthesize the status and quality of human studies on this topic. Methods: A systematic review was conducted in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA). Literature search was performed by combining variations of search terms related to acupoints and pain sensitivity in PubMed, EMBASE, and Alt HealthWatch (EBSCOHost). Screening of titles and abstracts and review of full-text articles for eligibility were performed by two independent investigators. Using a predefined template, information on subject characteristics, pathologic conditions, names of assessed acupoints, and relevant main findings were extracted from the included studies. The methodological quality of included studies was assessed using a modified Newcastle-Ottawa Scale (NOS) for case-control studies. A quality assessment checklist was also developed by the present authors to examine the quality of reporting of experimental variables that were considered important for evaluating acupoint sensitivity. Results: A total of 3453 studies were identified from the database search, of which 11 met the eligibility criteria to be included in this review. Six studies examined the mechanical sensitivity of body acupoints, and the remaining five studies examined the mechanical sensitivity of auricular points. Overall, findings suggest that the sensitivity of acupoints may be distinct in healthy versus clinical populations. However, there were various potential sources of bias and substantial heterogeneity across included studies in clinical conditions and acupoints. Conclusion: There is at present insufficient evidence to support or refute that acupoints in humans are sensitized in pathologic conditions. There were various methodological issues, including small sample size and poor reporting of experimental design and variables, which limit the ability to draw a definitive conclusion on this topic. It is also largely unclear whether it is the general body regions rather than specific acupoints that may be sensitized, as most studies did not include nonacupoint location(s) for comparison. Thus, further rigorous research is warranted.
Collapse
Affiliation(s)
- EunMee Yang
- Osher Center for Integrative Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Weidong Lu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Dennis Muñoz-Vergara
- Osher Center for Integrative Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Esme Goldfinger
- Osher Center for Integrative Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Ted J Kaptchuk
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Vitaly Napadow
- Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Andrew C Ahn
- Labfront Inc.., Boston, MA, USA
- Department of Medicine, Veteran Affairs Boston Healthcare System, West Roxbury, MA, USA
| | - Peter M Wayne
- Osher Center for Integrative Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
20
|
Standley A, Xie J, Lau AW, Grote L, Gifford AJ. Working with Miraculous Mice: Mus musculus as a Model Organism. Curr Protoc 2024; 4:e70021. [PMID: 39435766 DOI: 10.1002/cpz1.70021] [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: 10/23/2024]
Abstract
The laboratory mouse has been described as a "miracle" model organism, providing a window by which we may gain an understanding of ourselves. Since the first recorded mouse experiment in 1664, the mouse has become the most used animal model in biomedical research. Mice are ideally suited as a model organism because of their small size, short gestation period, large litter size, and genetic similarity to humans. This article provides a broad overview of the laboratory mouse as a model organism and is intended for undergraduates and those new to working with mice. We delve into the history of the laboratory mouse and outline important terminology to accurately describe research mice. The types of laboratory mice available to researchers are reviewed, including outbred stocks, inbred strains, immunocompromised mice, and genetically engineered mice. The critical role mice have played in advancing knowledge in the areas of oncology, immunology, and pharmacology is highlighted by examining the significant contribution of mice to Nobel Prize winning research. International mouse mutagenesis programs and accurate phenotyping of mouse models are outlined. We also explain important considerations for working with mice, including animal ethics; the welfare principles of replacement, refinement, and reduction; and the choice of mouse model in experimental design. Finally, we present practical advice for maintaining a mouse colony, which involves adequate training of staff, the logistics of mouse housing, monitoring colony health, and breeding strategies. Useful resources for working with mice are also listed. The aim of this overview is to equip the reader with a broad appreciation of the enormous potential and some of the complexities of working with the laboratory mouse in a quest to improve human health. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Anick Standley
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Jinhan Xie
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Angelica Wy Lau
- Garvan Institute of Medical Research, St Vincent's Clinical School, Darlinghurst, NSW, Australia
| | - Lauren Grote
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
| | - Andrew J Gifford
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- Anatomical Pathology, NSW Heath Pathology, Prince of Wales Hospital, Randwick, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
21
|
Tang MC, Wong KH, Azman AS, Lani R. Applications and advancements in animal models for antiviral research on mosquito-borne arboviruses. Animal Model Exp Med 2024; 7:673-684. [PMID: 38987937 PMCID: PMC11528384 DOI: 10.1002/ame2.12471] [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: 01/31/2024] [Accepted: 06/27/2024] [Indexed: 07/12/2024] Open
Abstract
Vector-borne diseases caused by arthropod-borne viruses (arboviruses) are a considerable challenge to public health globally. Mosquito-borne arboviruses, such as Chikungunya, Dengue, and Zika viruses, cause a range of human illnesses and may be fatal. Currently, efforts to control these diseases still face challenges due to growing vector resistance towards insecticides, urbanization, and limited effective antiviral treatments and vaccines. Animal models are crucial in antiviral research on mosquito-borne arboviruses, playing a role in understanding disease mechanisms, vaccine development, and toxicity testing, but the application of animal models still faces the challenges of ethical considerations and animal-to-human translational success. Genetically engineered mouse models, hamster models and non-human primate (NHP) are currently used in arbovirus research, but new models such as tree shrews and novel humanized mice are emerging. In the context of Malaysian research, the use of long-tailed macaques as potential NHP models for arbovirus research is possible; however, it faces the ethical dilemma of using an endangered species for scientific purposes. Overall, animal models play a crucial role in advancing infectious disease research, but a balance between medical research and species conservation must be upheld.
Collapse
Affiliation(s)
| | - Ka Heng Wong
- School of ScienceMonash University MalaysiaSunwaySelangorMalaysia
| | | | - Rafidah Lani
- Department of Medical Microbiology, Faculty of MedicineUniversiti MalayaKuala LumpurMalaysia
| |
Collapse
|
22
|
Kim H, Hwang J, Park C, Park R. Redox system and ROS-related disorders in peroxisomes. Free Radic Res 2024; 58:662-675. [PMID: 39550761 DOI: 10.1080/10715762.2024.2427088] [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: 07/29/2024] [Revised: 10/10/2024] [Accepted: 11/01/2024] [Indexed: 11/19/2024]
Abstract
Peroxisomes are essential organelles that help mitigate the oxidative damage caused by reactive oxygen species (ROS) through their antioxidant systems. They perform functions such as α-oxidation, β-oxidation, and the synthesis of cholesterol and ether phospholipids. During the breakdown of specific metabolites, peroxisomes generate ROS as byproducts, which can either be neutralized or contribute to oxidative stress. The relationship between peroxisomal metabolism and ROS-related disorders, including neurodegenerative diseases and cancers, has been studied for decades; however, the exact mechanisms remain unclear. Our review will provide recent insights into the peroxisomal redox system and its association with oxidative stress-related diseases.
Collapse
Affiliation(s)
- Hyunsoo Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jaetaek Hwang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Channy Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| |
Collapse
|
23
|
Ahkam AH, Susilawati Y, Sumiwi SA. Peronema canescens as a Source of Immunomodulatory Agents: A New Opportunity and Perspective. BIOLOGY 2024; 13:744. [PMID: 39336171 PMCID: PMC11428267 DOI: 10.3390/biology13090744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/01/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024]
Abstract
Immunomodulators are pivotal in managing various health conditions by regulating the immune response by either enhancing or suppressing it to maintain homeostasis. The growing interest in natural sources of immunomodulatory agents has spurred the investigation of numerous medicinal plants, including Peronema canescens, commonly known in Asia as sungkai. Traditionally used for its medicinal properties in Southeast Asia, Peronema canescens belongs to the Verbenaceae family and has garnered significant attention. This review discusses the immunomodulatory activity of the active compounds in Peronema canescens and explores the potential directions for future research.
Collapse
Affiliation(s)
- Ahmad Hafidul Ahkam
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang 45363, Indonesia
| | - Yasmiwar Susilawati
- Department of Biology Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang 45363, Indonesia
- The Herbal Studies Center, Faculty of Pharmacy, Padjadjaran University, Sumedang 45363, Indonesia
| | - Sri Adi Sumiwi
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Padjadjaran University, Sumedang 45363, Indonesia
| |
Collapse
|
24
|
Chowdhury R, Bhuia MS, Al Hasan MS, Hossain Snigdha S, Afrin S, Büsselberg D, Habtemariam S, Sönmez Gürer E, Sharifi‐Rad J, Ahmed Aldahish A, Аkhtayeva N, Islam MT. Anticancer potential of phytochemicals derived from mangrove plants: Comprehensive mechanistic insights. Food Sci Nutr 2024; 12:6174-6205. [PMID: 39554337 PMCID: PMC11561795 DOI: 10.1002/fsn3.4318] [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: 02/24/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 11/19/2024] Open
Abstract
Cancer is a collection of illnesses characterized by aberrant cellular proliferation that can infiltrate or metastasize to distant anatomical sites, posing a notable threat to human well-being due to its substantial morbidity and death rates worldwide. The potential of plant-derived natural compounds as anticancer medicines has been assessed owing to their favorable attributes of few side effects and significant antitumor activity. Mangrove plants and their derived compounds have been scientifically shown to exhibit many significant beneficial biological activities, such as anti-inflammatory, immunomodulatory, antioxidant, neuroprotective, cardioprotective, and hepatoprotective properties. This study summarized mangrove plants and their derived compounds as potential anticancer agents, with an emphasis on the underlying molecular mechanisms. To explore this, we gathered data on the preclinical (in vivo and in vitro) anticancer effects of mangrove plants and their derived compounds from reputable literature spanning 2000 to 2023. We conducted thorough searches in various academic databases, including PubMed, ScienceDirect, Wiley Online, SpringerLink, Google Scholar, Scopus, and the Web of Science. The results demonstrated that mangrove plants and their derived compounds have promising anticancer properties in preclinical pharmacological test systems through various molecular mechanisms, including induction of oxidative stress and mitochondrial dysfunction, cytotoxicity, genotoxicity, cell cycle arrest, apoptosis, autophagy, antiproliferative, antimetastatic, and other miscellaneous actions. Upon thorough observation of the pertinent information, it is suggested that mangrove plants and their derived chemicals may serve as a potential lead in the development of novel drugs for cancer therapy.
Collapse
Affiliation(s)
- Raihan Chowdhury
- Department of PharmacyBangabandhu Sheikh Mujibur Rahman Science and Technology UniversityGopalganjBangladesh
- Phytochemistry and Biodiversity Research LaboratoryBioLuster Research CenterGopalganjBangladesh
| | - Md. Shimul Bhuia
- Department of PharmacyBangabandhu Sheikh Mujibur Rahman Science and Technology UniversityGopalganjBangladesh
- Phytochemistry and Biodiversity Research LaboratoryBioLuster Research CenterGopalganjBangladesh
| | - Md. Sakib Al Hasan
- Department of PharmacyBangabandhu Sheikh Mujibur Rahman Science and Technology UniversityGopalganjBangladesh
| | | | - Sadia Afrin
- Pharmacy DisciplineKhulna UniversityKhulnaBangladesh
| | | | | | - Eda Sönmez Gürer
- Faculty of Pharmacy, Department of PharmacognosySivas Cumhuriyet UniversitySivasTurkey
| | - Javad Sharifi‐Rad
- Department of Biomedical SciencesCollege of Medicine, Korea UniversitySeoulRepublic of Korea
| | - Afaf Ahmed Aldahish
- Department of Pharmacology, College of PharmacyKing Khalid UniversityAbhaSaudi Arabia
| | - Nursulu Аkhtayeva
- Department of Biodiversity and Bioresources of Al‐Farabi Kazakh National UniversityAlmatyKazakhstan
| | - Muhammad Torequl Islam
- Department of PharmacyBangabandhu Sheikh Mujibur Rahman Science and Technology UniversityGopalganjBangladesh
- Phytochemistry and Biodiversity Research LaboratoryBioLuster Research CenterGopalganjBangladesh
- Pharmacy DisciplineKhulna UniversityKhulnaBangladesh
| |
Collapse
|
25
|
Kumar P, Verma A, Ashique S, Bhowmick M, Mohanto S, Singh A, Gupta M, Gupta A, Haider T. Unlocking the role of herbal cosmeceutical in anti-ageing and skin ageing associated diseases. Cutan Ocul Toxicol 2024; 43:211-226. [PMID: 39024063 DOI: 10.1080/15569527.2024.2380326] [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: 04/01/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/20/2024]
Abstract
The process of skin ageing is a natural biological phenomenon characterised by the emergence of wrinkles, age spots, sagging skin, and dryness over time. The increasing significance of skin in physical attractiveness has heightened skincare concerns. Anti-ageing cosmetics play a pivotal role in nurturing the skin, enhancing its quality, and promoting overall health. Today, cosmetics have evolved beyond mere aesthetics and are now integral to individual wellness. The contemporary quest for perpetual youth has intensified, prompting a deeper exploration into the skin ageing process. This comprehensive exploration delves into various elements involved in skin ageing, encompassing cells such as stem and endothelial cells, blood vessels, soft tissues, and signalling pathways. The molecular basis of skin ageing, including biochemical factors like reactive oxygen species, damaged DNA, free radicals, ions, and proteins (mRNA), is scrutinised alongside relevant animal models. The article critically analyzes the outcomes of utilising herbal components, emphasising their advantageous anti-ageing properties. The factors contributing to skin ageing, mechanistic perspectives, management approaches involving herbal cosmeceutical, and associated complications (especially cardiovascular diseases, Parkinson's, Alzheimer's, etc.) are succinctly addressed. In addition, the manuscript further summarises the recent patented innovations and toxicity of the herbal cosmeceuticals for anti-ageing and ageing associated disorders. Despite progress, further research is imperative to unlock the full potential of herbal components as anti-ageing agents.
Collapse
Affiliation(s)
- Prashant Kumar
- SRM University Delhi-NCR Campus, Ghaziabad, UPttarpradesh, India
| | - Anurag Verma
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, UPttarpradesh, India
| | - Sumel Ashique
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences & Research, Durgapur, West Bengal, India
| | - Mithun Bhowmick
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences & Research, Durgapur, West Bengal, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Anita Singh
- Department of Pharmaceutical Sciences, Kumaun University Campus, Bhimtal, Uttarakhand, India
| | - Madhu Gupta
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Abhishek Gupta
- Department of Pharmacognosy and phytochemistry, Hygia Institute of Pharmaceutical Education & Research, Lucknow, Uttar Pradesh, India
| | - Tanweer Haider
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University Madhya Pradesh, Gwalior, India
| |
Collapse
|
26
|
Rahman M, Sahoo A, Almalki WH, Salman Almujri S, Aodah A, Alnofei AA, Alhamyani A. Three-dimensional cell culture: Future scope in cancer vaccine development. Drug Discov Today 2024; 29:104114. [PMID: 39067612 DOI: 10.1016/j.drudis.2024.104114] [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: 03/25/2024] [Revised: 07/10/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Three-dimensional (3D) cell culture techniques, which are superior to 2D methods in viability and functionality, are being used to develop innovative cancer vaccines. Tumor spheroids, which are structurally and functionally similar to actual tumors, can be developed using 3D cell culture. These spheroid vaccines have shown superior antitumor immune responses to 2D cell-based vaccines. Dendritic cell vaccines can also be produced more efficiently using 3D cell culture. Personalized cancer vaccines are being developed using 3D cell culture, providing substantial benefits over 2D methods. The more natural conditions of 3D cell culture might promote the expression of tumor antigens not expressed in 2D culture, potentially allowing for more targeted vaccines by co-culturing tumor cells with other cell types. Advanced cancer vaccines using 3D cell cultures are expected soon.
Collapse
Affiliation(s)
- Mahfoozur Rahman
- Department of Pharmaceutical Sciences, Shalom Institute of Health & Allied Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad 211007, Uttar Pradesh, India.
| | - Ankit Sahoo
- College of Pharmacy, J.S. University, Shikohabad, Firozabad, Uttar Pradesh, 283135, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Salem Salman Almujri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Asir-Abha 61421, Saudi Arabia
| | - Alhussain Aodah
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Abdulrahman A Alnofei
- Psychological Measurement and Evaluation, Department of Psychology, Faculty of Education, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdulrahman Alhamyani
- Pharmaceuticals Chemistry Department, Faculty of Clinical Pharmacy, Al Baha University, Al Baha 65779, Saudi Arabia
| |
Collapse
|
27
|
Park G, Rim YA, Sohn Y, Nam Y, Ju JH. Replacing Animal Testing with Stem Cell-Organoids : Advantages and Limitations. Stem Cell Rev Rep 2024; 20:1375-1386. [PMID: 38639829 PMCID: PMC11319430 DOI: 10.1007/s12015-024-10723-5] [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] [Accepted: 04/08/2024] [Indexed: 04/20/2024]
Abstract
Various groups including animal protection organizations, medical organizations, research centers, and even federal agencies such as the U.S. Food and Drug Administration, are working to minimize animal use in scientific experiments. This movement primarily stems from animal welfare and ethical concerns. However, recent advances in technology and new studies in medicine have contributed to an increase in animal experiments throughout the years. With the rapid increase in animal testing, concerns arise including ethical issues, high cost, complex procedures, and potential inaccuracies.Alternative solutions have recently been investigated to address the problems of animal testing. Some of these technologies are related to stem cell technologies, such as organ-on-a-chip, organoids, and induced pluripotent stem cell models. The aim of the review is to focus on stem cell related methodologies, such as organoids, that can serve as an alternative to animal testing and discuss its advantages and limitations, alongside regulatory considerations.Although stem cell related methodologies has shortcomings, it has potential to replace animal testing. Achieving this requires further research on stem cells, with potential societal and technological benefits.
Collapse
Affiliation(s)
- Guiyoung Park
- School of Biopharmaceutical and Medical Sciences, Health & Wellness College, Sungshin Women's University, 55, Dobong-ro 76ga-gil, Gangbuk-gu, Seoul, Republic of Korea
| | - Yeri Alice Rim
- CiSTEM laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, 4 3, Seoul, 06591, Republic of Korea
- Department of Biomedicine & Health Sciences, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yeowon Sohn
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
| | - Yoojun Nam
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea.
- Yipscell Inc, L2 Omnibus Park, Banpo-dearo 222, Seocho-gu, Seoul, Korea.
| | - Ji Hyeon Ju
- CiSTEM laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, 4 3, Seoul, 06591, Republic of Korea.
- Department of Biomedicine & Health Sciences, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Yipscell Inc, L2 Omnibus Park, Banpo-dearo 222, Seocho-gu, Seoul, Korea.
| |
Collapse
|
28
|
Farani PSG, Jones KM, Poveda C. Treatments and the Perspectives of Developing a Vaccine for Chagas Disease. Vaccines (Basel) 2024; 12:870. [PMID: 39203996 PMCID: PMC11359273 DOI: 10.3390/vaccines12080870] [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: 05/21/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 09/03/2024] Open
Abstract
Chagas disease (CD) treatment and vaccine development are critical due to the significant health burden caused by the disease, especially in Latin America. Current treatments include benznidazole and nifurtimox, which are most effective in the acute phase of the disease but less so in the chronic phase, often with significant side effects. Here, using the available literature, we summarize the progress in vaccine development and new treatments that promise to reduce CD incidence and improve the quality of life for those at risk, particularly in endemic regions. New treatment options, such as posaconazole and fexinidazole, are being explored to improve efficacy and reduce adverse effects. Vaccine development for CD remains a high priority. The complex life stages and genetic diversity of Trypanosoma cruzi present challenges, but several promising vaccine candidates are under investigation. These efforts focus on stimulating a protective immune response through various innovative approaches.
Collapse
Affiliation(s)
- Priscila Silva Grijó Farani
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79902, USA
| | - Kathryn Marie Jones
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristina Poveda
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
29
|
Mulay AR, Hwang J, Kim DH. Microphysiological Blood-Brain Barrier Systems for Disease Modeling and Drug Development. Adv Healthc Mater 2024; 13:e2303180. [PMID: 38430211 PMCID: PMC11338747 DOI: 10.1002/adhm.202303180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 02/22/2024] [Indexed: 03/03/2024]
Abstract
The blood-brain barrier (BBB) is a highly controlled microenvironment that regulates the interactions between cerebral blood and brain tissue. Due to its selectivity, many therapeutics targeting various neurological disorders are not able to penetrate into brain tissue. Pre-clinical studies using animals and other in vitro platforms have not shown the ability to fully replicate the human BBB leading to the failure of a majority of therapeutics in clinical trials. However, recent innovations in vitro and ex vivo modeling called organs-on-chips have shown the potential to create more accurate disease models for improved drug development. These microfluidic platforms induce physiological stressors on cultured cells and are able to generate more physiologically accurate BBBs compared to previous in vitro models. In this review, different approaches to create BBBs-on-chips are explored alongside their application in modeling various neurological disorders and potential therapeutic efficacy. Additionally, organs-on-chips use in BBB drug delivery studies is discussed, and advances in linking brain organs-on-chips onto multiorgan platforms to mimic organ crosstalk are reviewed.
Collapse
Affiliation(s)
- Atharva R. Mulay
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
| | - Jihyun Hwang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland 21218
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Center for Microphysiological Systems, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, 21218
| |
Collapse
|
30
|
Rudroff T. Artificial Intelligence as a Replacement for Animal Experiments in Neurology: Potential, Progress, and Challenges. Neurol Int 2024; 16:805-820. [PMID: 39195562 DOI: 10.3390/neurolint16040060] [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: 06/05/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
Animal experimentation has long been a cornerstone of neurology research, but it faces growing scientific, ethical, and economic challenges. Advances in artificial intelligence (AI) are providing new opportunities to replace animal testing with more human-relevant and efficient methods. This article explores the potential of AI technologies such as brain organoids, computational models, and machine learning to revolutionize neurology research and reduce reliance on animal models. These approaches can better recapitulate human brain physiology, predict drug responses, and uncover novel insights into neurological disorders. They also offer faster, cheaper, and more ethical alternatives to animal experiments. Case studies demonstrate AI's ability to accelerate drug discovery for Alzheimer's, predict neurotoxicity, personalize treatments for Parkinson's, and restore movement in paralysis. While challenges remain in validating and integrating these technologies, the scientific, economic, practical, and moral advantages are driving a paradigm shift towards AI-based, animal-free research in neurology. With continued investment and collaboration across sectors, AI promises to accelerate the development of safer and more effective therapies for neurological conditions while significantly reducing animal use. The path forward requires the ongoing development and validation of these technologies, but a future in which they largely replace animal experiments in neurology appears increasingly likely. This transition heralds a new era of more humane, human-relevant, and innovative brain research.
Collapse
Affiliation(s)
- Thorsten Rudroff
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA 52242, USA
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| |
Collapse
|
31
|
Bock M, Hong SJ, Zhang S, Yu Y, Lee S, Shin H, Choi BH, Han I. Morphogenetic Designs, and Disease Models in Central Nervous System Organoids. Int J Mol Sci 2024; 25:7750. [PMID: 39062993 PMCID: PMC11276855 DOI: 10.3390/ijms25147750] [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: 06/24/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Since the emergence of the first cerebral organoid (CO) in 2013, advancements have transformed central nervous system (CNS) research. Initial efforts focused on studying the morphogenesis of COs and creating reproducible models. Numerous methodologies have been proposed, enabling the design of the brain organoid to represent specific regions and spinal cord structures. CNS organoids now facilitate the study of a wide range of CNS diseases, from infections to tumors, which were previously difficult to investigate. We summarize the major advancements in CNS organoids, concerning morphogenetic designs and disease models. We examine the development of fabrication procedures and how these advancements have enabled the generation of region-specific brain organoids and spinal cord models. We highlight the application of these organoids in studying various CNS diseases, demonstrating the versatility and potential of organoid models in advancing our understanding of complex conditions. We discuss the current challenges in the field, including issues related to reproducibility, scalability, and the accurate recapitulation of the in vivo environment. We provide an outlook on prospective studies and future directions. This review aims to provide a comprehensive overview of the state-of-the-art CNS organoid research, highlighting key developments, current challenges, and prospects in the field.
Collapse
Affiliation(s)
- Minsung Bock
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Sung Jun Hong
- Research Competency Milestones Program, School of Medicine, CHA University, Seongnam-si 13488, Republic of Korea;
- Department of Medicine, School of Medicine, CHA University, Seongnam-si 13496, Republic of Korea
| | - Songzi Zhang
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Yerin Yu
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Somin Lee
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Haeeun Shin
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Byung Hyune Choi
- Department of Biomedical Science, Inha University College of Medicine, Incheon 22212, Republic of Korea;
| | - Inbo Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
- Advanced Regenerative Medicine Research Center, CHA Future Medicine Research Institute, Seongnam-si 13488, Republic of Korea
| |
Collapse
|
32
|
Myung Y, de Sá AGC, Ascher DB. Deep-PK: deep learning for small molecule pharmacokinetic and toxicity prediction. Nucleic Acids Res 2024; 52:W469-W475. [PMID: 38634808 PMCID: PMC11223837 DOI: 10.1093/nar/gkae254] [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: 02/14/2024] [Revised: 03/20/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024] Open
Abstract
Evaluating pharmacokinetic properties of small molecules is considered a key feature in most drug development and high-throughput screening processes. Generally, pharmacokinetics, which represent the fate of drugs in the human body, are described from four perspectives: absorption, distribution, metabolism and excretion-all of which are closely related to a fifth perspective, toxicity (ADMET). Since obtaining ADMET data from in vitro, in vivo or pre-clinical stages is time consuming and expensive, many efforts have been made to predict ADMET properties via computational approaches. However, the majority of available methods are limited in their ability to provide pharmacokinetics and toxicity for diverse targets, ensure good overall accuracy, and offer ease of use, interpretability and extensibility for further optimizations. Here, we introduce Deep-PK, a deep learning-based pharmacokinetic and toxicity prediction, analysis and optimization platform. We applied graph neural networks and graph-based signatures as a graph-level feature to yield the best predictive performance across 73 endpoints, including 64 ADMET and 9 general properties. With these powerful models, Deep-PK supports molecular optimization and interpretation, aiding users in optimizing and understanding pharmacokinetics and toxicity for given input molecules. The Deep-PK is freely available at https://biosig.lab.uq.edu.au/deeppk/.
Collapse
Affiliation(s)
- Yoochan Myung
- School of Chemistry and Molecular Biosciences, The Australian Centre for Ecogenomics, The University of Queensland, Brisbane, Queensland 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Alex G C de Sá
- School of Chemistry and Molecular Biosciences, The Australian Centre for Ecogenomics, The University of Queensland, Brisbane, Queensland 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - David B Ascher
- School of Chemistry and Molecular Biosciences, The Australian Centre for Ecogenomics, The University of Queensland, Brisbane, Queensland 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
33
|
Kumar P, Verma A, Ashique S, Bhowmick M, Mohanto S, Singh A, Gupta M, Gupta A, Haider T. Unlocking the role of herbal cosmeceutical in anti-ageing and skin ageing associated diseases. Cutan Ocul Toxicol 2024; 43:211-226. [DOI: https:/doi.org/10.1080/15569527.2024.2380326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 04/05/2025]
Affiliation(s)
- Prashant Kumar
- SRM University Delhi-NCR Campus, Ghaziabad, UPttarpradesh, India
| | - Anurag Verma
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, UPttarpradesh, India
| | - Sumel Ashique
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences & Research, Durgapur, West Bengal, India
| | - Mithun Bhowmick
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences & Research, Durgapur, West Bengal, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Anita Singh
- Department of Pharmaceutical Sciences, Kumaun University Campus, Bhimtal, Uttarakhand, India
| | - Madhu Gupta
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Abhishek Gupta
- Department of Pharmacognosy and phytochemistry, Hygia Institute of Pharmaceutical Education & Research, Lucknow, Uttar Pradesh, India
| | - Tanweer Haider
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University Madhya Pradesh, Gwalior, India
| |
Collapse
|
34
|
Hauben M. A Pharmacovigilance Florilegium. Clin Ther 2024; 46:520-523. [PMID: 39030077 DOI: 10.1016/j.clinthera.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 07/21/2024]
Affiliation(s)
- Manfred Hauben
- Department of Family and Community Medicine, New York Medical College, Valhalla, New York; Truliant Consulting, Baltimore, Maryland.
| |
Collapse
|
35
|
Vashishat A, Patel P, Das Gupta G, Das Kurmi B. Alternatives of Animal Models for Biomedical Research: a Comprehensive Review of Modern Approaches. Stem Cell Rev Rep 2024; 20:881-899. [PMID: 38429620 DOI: 10.1007/s12015-024-10701-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2024] [Indexed: 03/03/2024]
Abstract
Biomedical research has long relied on animal models to unravel the intricacies of human physiology and pathology. However, concerns surrounding ethics, expenses, and inherent species differences have catalyzed the exploration of alternative avenues. The contemporary alternatives to traditional animal models in biomedical research delve into three main categories of alternative approaches: in vitro models, in vertebrate models, and in silico models. This unique approach to artificial intelligence and machine learning has been a keen interest to be used in different biomedical research. The main goal of this review is to serve as a guide to researchers seeking novel avenues for their investigations and underscores the importance of considering alternative models in the pursuit of scientific knowledge and medical breakthroughs, including showcasing the broad spectrum of modern approaches that are revolutionizing biomedical research and leading the way toward a more ethical, efficient, and innovative future. Models can insight into cellular processes, developmental biology, drug interaction, assessing toxicology, and understanding molecular mechanisms.
Collapse
Affiliation(s)
- Abhinav Vashishat
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India.
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
36
|
Ifediora N, Canoll P, Hargus G. Human stem cell transplantation models of Alzheimer's disease. Front Aging Neurosci 2024; 16:1354164. [PMID: 38450383 PMCID: PMC10915253 DOI: 10.3389/fnagi.2024.1354164] [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: 12/12/2023] [Accepted: 02/06/2024] [Indexed: 03/08/2024] Open
Abstract
Alzheimer's disease (AD) is the most frequent form of dementia. It is characterized by pronounced neuronal degeneration with formation of neurofibrillary tangles and deposition of amyloid β throughout the central nervous system. Animal models have provided important insights into the pathogenesis of AD and they have shown that different brain cell types including neurons, astrocytes and microglia have important functions in the pathogenesis of AD. However, there are difficulties in translating promising therapeutic observations in mice into clinical application in patients. Alternative models using human cells such as human induced pluripotent stem cells (iPSCs) may provide significant advantages, since they have successfully been used to model disease mechanisms in neurons and in glial cells in neurodegenerative diseases in vitro and in vivo. In this review, we summarize recent studies that describe the transplantation of human iPSC-derived neurons, astrocytes and microglial cells into the forebrain of mice to generate chimeric transplantation models of AD. We also discuss opportunities, challenges and limitations in using differentiated human iPSCs for in vivo disease modeling and their application for biomedical research.
Collapse
Affiliation(s)
- Nkechime Ifediora
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Peter Canoll
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
| | - Gunnar Hargus
- Department of Pathology and Cell Biology, Columbia University, New York, NY, United States
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, United States
| |
Collapse
|
37
|
Tawade P, Mastrangeli M. Integrated Electrochemical and Optical Biosensing in Organs-on-Chip. Chembiochem 2024; 25:e202300560. [PMID: 37966365 DOI: 10.1002/cbic.202300560] [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: 08/09/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/16/2023]
Abstract
Demand for biocompatible, non-invasive, and continuous real-time monitoring of organs-on-chip has driven the development of a variety of novel sensors. However, highest accuracy and sensitivity can arguably be achieved by integrated biosensing, which enables in situ monitoring of the in vitro microenvironment and dynamic responses of tissues and miniature organs recapitulated in organs-on-chip. This paper reviews integrated electrical, electrochemical, and optical sensing methods within organ-on-chip devices and platforms. By affording precise detection of analytes and biochemical reactions, these methods expand and advance the monitoring capabilities and reproducibility of organ-on-chip technology. The integration of these sensing techniques allows a deeper understanding of organ functions, and paves the way for important applications such as drug testing, disease modeling, and personalized medicine. By consolidating recent advancements and highlighting challenges in the field, this review aims to foster further research and innovation in the integration of biosensing in organs-on-chip.
Collapse
Affiliation(s)
- Pratik Tawade
- Electronic Components, Technology and Materials, Department of Microelectronics, Delft University of Technology, Mekelweg 4, 2628CD, Delft, Netherlands
| | - Massimo Mastrangeli
- Electronic Components, Technology and Materials, Department of Microelectronics, Delft University of Technology, Mekelweg 4, 2628CD, Delft, Netherlands
| |
Collapse
|
38
|
Bai H, Olson KNP, Pan M, Marshall T, Singh H, Ma J, Gilbride P, Yuan Y, McCormack J, Si L, Maharjan S, Huang D, Qian X, Livermore C, Zhang YS, Xie X. Rapid Prototyping of Thermoplastic Microfluidic 3D Cell Culture Devices by Creating Regional Hydrophilicity Discrepancy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304332. [PMID: 38032118 PMCID: PMC10870023 DOI: 10.1002/advs.202304332] [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: 06/28/2023] [Revised: 10/02/2023] [Indexed: 12/01/2023]
Abstract
Microfluidic 3D cell culture devices that enable the recapitulation of key aspects of organ structures and functions in vivo represent a promising preclinical platform to improve translational success during drug discovery. Essential to these engineered devices is the spatial patterning of cells from different tissue types within a confined microenvironment. Traditional fabrication strategies lack the scalability, cost-effectiveness, and rapid prototyping capabilities required for industrial applications, especially for processes involving thermoplastic materials. Here, an approach to pattern fluid guides inside microchannels is introduced by establishing differential hydrophilicity using pressure-sensitive adhesives as masks and a subsequent selective coating with a biocompatible polymer. Optimal coating conditions are identified using polyvinylpyrrolidone, which resulted in rapid and consistent hydrogel flow in both the open-chip prototype and the fully bonded device containing additional features for medium perfusion. The suitability of the device for dynamic 3D cell culture is tested by growing human hepatocytes in the device under controlled fluid flow for a 14-day period. Additionally, the study demonstrated the potential of using the device for pharmaceutical high-throughput screening applications, such as predicting drug-induced liver injury. The approach offers a facile strategy of rapid prototyping thermoplastic microfluidic organ chips with varying geometries, microstructures, and substrate materials.
Collapse
Affiliation(s)
| | | | - Ming Pan
- Xellar BiosystemsCambridgeMA02458USA
| | | | | | | | | | | | | | - Longlong Si
- CAS Key Laboratory of Quantitative Engineering BiologyShenzhen Institute of Synthetic BiologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Sushila Maharjan
- Division of Engineering in MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolCambridgeMA02142USA
| | - Di Huang
- Research Center for Nano‐biomaterials & Regenerative MedicineCollege of Biomedical EngineeringTaiyuan University of TechnologyTaiyuan030024P. R. China
| | | | - Carol Livermore
- Department of Mechanical and Industrial EngineeringNortheastern UniversityBostonMA02115USA
| | - Yu Shrike Zhang
- Division of Engineering in MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolCambridgeMA02142USA
| | - Xin Xie
- Xellar BiosystemsCambridgeMA02458USA
| |
Collapse
|
39
|
Lancheros Porras KD, Alves IA, Novoa DMA. PBPK Modeling as an Alternative Method of Interspecies Extrapolation that Reduces the Use of Animals: A Systematic Review. Curr Med Chem 2024; 31:102-126. [PMID: 37031391 DOI: 10.2174/0929867330666230408201849] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/03/2023] [Accepted: 02/03/2023] [Indexed: 04/10/2023]
Abstract
INTRODUCTION Physiologically based pharmacokinetic (PBPK) modeling is a computational approach that simulates the anatomical structure of the studied species and presents the organs and tissues as compartments interconnected by arterial and venous blood flows. AIM The aim of this systematic review was to analyze the published articles focused on the development of PBPK models for interspecies extrapolation in the disposition of drugs and health risk assessment, presenting to this modeling an alternative to reduce the use of animals. METHODS For this purpose, a systematic search was performed in PubMed using the following search terms: "PBPK" and "Interspecies extrapolation". The revision was performed according to PRISMA guidelines. RESULTS In the analysis of the articles, it was found that rats and mice are the most commonly used animal models in the PBPK models; however, most of the physiological and physicochemical information used in the reviewed studies were obtained from previous publications. Additionally, most of the PBPK models were developed to extrapolate pharmacokinetic parameters to humans and the main application of the models was for toxicity testing. CONCLUSION PBPK modeling is an alternative that allows the integration of in vitro and in silico data as well as parameters reported in the literature to predict the pharmacokinetics of chemical substances, reducing in large quantity the use of animals that are required in traditional studies.
Collapse
|
40
|
Kim J, Shin SA, Lee CS, Chung HJ. An Improved In Vitro Blood-Brain Barrier Model for the Evaluation of Drug Permeability Using Transwell with Shear Stress. Pharmaceutics 2023; 16:48. [PMID: 38258059 PMCID: PMC10820479 DOI: 10.3390/pharmaceutics16010048] [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: 12/10/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
The development of drugs targeting the central nervous system (CNS) is challenging because of the presence of the Blood-Brain barrier (BBB). Developing physiologically relevant in vitro BBB models for evaluating drug permeability and predicting the activity of drug candidates is crucial. The transwell model is one of the most widely used in vitro BBB models. However, this model has limitations in mimicking in vivo conditions, particularly in the absence of shear stress. This study aimed to overcome the limitations of the transwell model using immortalized human endothelial cells (hCMEC/D3) by developing a novel dish design for an orbital shaker, providing shear stress. During optimization, we assessed cell layer integrity using trans-endothelial electrical resistance measurements and the % diffusion of lucifer yellow. The efflux transporter activity and mRNA expression of junctional proteins (claudin-5, occludin, and VE-cadherin) in the newly optimized model were verified. Additionally, the permeability of 14 compounds was evaluated and compared with published in vivo data. The cell-layer integrity was substantially increased using the newly designed annular shaking-dish model. The results demonstrate that our model provided robust conditions for evaluating the permeability of CNS drug candidates, potentially improving the reliability of in vitro BBB models in drug development.
Collapse
Affiliation(s)
- Junhyeong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.K.); (S.-A.S.); (C.S.L.)
- Anti-Aging Bio Cell factory Regional Leading Research Center (ABC-RLRC), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Seong-Ah Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.K.); (S.-A.S.); (C.S.L.)
| | - Chang Sup Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.K.); (S.-A.S.); (C.S.L.)
| | - Hye Jin Chung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (J.K.); (S.-A.S.); (C.S.L.)
| |
Collapse
|
41
|
Akash SR, Arnob MAJB, Uddin MJ. FDA Modernization Act 2.0: An insight from nondeveloping country. Drug Dev Res 2023; 84:1572-1577. [PMID: 37587871 DOI: 10.1002/ddr.22108] [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: 05/19/2023] [Revised: 07/18/2023] [Accepted: 08/07/2023] [Indexed: 08/18/2023]
Abstract
Animal testing is required in drug development research and is crucial for assessing the efficacy and safety of medications before they are commercialized. However, the newly furnished Food and Drug Administration Modernization Act 2.0 has given new insight into drug development. It opens a new door by offering an alternative testing method for developing a new drug without using animals. This newly proposed system may potentially significantly impact nondeveloped countries worldwide. In this study, we explore the alternative testing options such as in silico modeling, human tissue-on-chip engineering, animal-free recombinant antibodies, tissue engineering, and artificial intelligence presented by this act and discuss its implications for nondeveloped countries.
Collapse
Affiliation(s)
- Sajidur Rahman Akash
- Department of Pharmacy, Bangladesh University, Dhaka, Bangladesh
- ABEx Bio-Research Center, East Azampur, Dhaka, Bangladesh
| | - M A Jobayer Billah Arnob
- Department of Biomedical Engineering, Chittagong University of Engineering & Technology, Chittagong, Bangladesh
| | - Md Jamal Uddin
- ABEx Bio-Research Center, East Azampur, Dhaka, Bangladesh
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Korea
| |
Collapse
|
42
|
Moon HR, Surianarayanan N, Singh T, Han B. Microphysiological systems as reliable drug discovery and evaluation tools: Evolution from innovation to maturity. BIOMICROFLUIDICS 2023; 17:061504. [PMID: 38162229 PMCID: PMC10756708 DOI: 10.1063/5.0179444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Microphysiological systems (MPSs), also known as organ-on-chip or disease-on-chip, have recently emerged to reconstitute the in vivo cellular microenvironment of various organs and diseases on in vitro platforms. These microfluidics-based platforms are developed to provide reliable drug discovery and regulatory evaluation testbeds. Despite recent emergences and advances of various MPS platforms, their adoption of drug discovery and evaluation processes still lags. This delay is mainly due to a lack of rigorous standards with reproducibility and reliability, and practical difficulties to be adopted in pharmaceutical research and industry settings. This review discusses the current and potential use of MPS platforms in drug discovery processes while considering the context of several key steps during drug discovery processes, including target identification and validation, preclinical evaluation, and clinical trials. Opportunities and challenges are also discussed for the broader dissemination and adoption of MPSs in various drug discovery and regulatory evaluation steps. Addressing these challenges will transform long and expensive drug discovery and evaluation processes into more efficient discovery, screening, and approval of innovative drugs.
Collapse
Affiliation(s)
- Hye-Ran Moon
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | | | - Tarun Singh
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Bumsoo Han
- Author to whom correspondence should be addressed:. Tel: +1-765-494-5626
| |
Collapse
|
43
|
Steinberg E, Friedman R, Goldstein Y, Friedman N, Beharier O, Demma JA, Zamir G, Hubert A, Benny O. A fully 3D-printed versatile tumor-on-a-chip allows multi-drug screening and correlation with clinical outcomes for personalized medicine. Commun Biol 2023; 6:1157. [PMID: 37957280 PMCID: PMC10643569 DOI: 10.1038/s42003-023-05531-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Optimal clinical outcomes in cancer treatments could be achieved through the development of reliable, precise ex vivo tumor models that function as drug screening platforms for patient-targeted therapies. Microfluidic tumor-on-chip technology is emerging as a preferred tool since it enables the complex set-ups and recapitulation of the physiologically relevant physical microenvironment of tumors. In order to overcome the common hindrances encountered while using this technology, a fully 3D-printed device was developed that sustains patient-derived multicellular spheroids long enough to conduct multiple drug screening tests. This tool is both cost effective and possesses four necessary characteristics of effective microfluidic devices: transparency, biocompatibility, versatility, and sample accessibility. Compelling correlations which demonstrate a clinical proof of concept were found after testing and comparing different chemotherapies on tumor spheroids, derived from ten patients, to their clinical outcomes. This platform offers a potential solution for personalized medicine by functioning as a predictive drug-performance tool.
Collapse
Affiliation(s)
- Eliana Steinberg
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Roy Friedman
- School of Computer Science and Engineering, Center for Interdisciplinary Data Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yoel Goldstein
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nethanel Friedman
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ofer Beharier
- Hadassah Medical Center and The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jonathan Abraham Demma
- Department of General Surgery, Hadassah Medical Center and Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Gideon Zamir
- Department of General Surgery, Hadassah Medical Center and Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ayala Hubert
- Oncology Department, Hadassah Medical Center, Jerusalem, Israel
| | - Ofra Benny
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
44
|
Mironiuk-Puchalska E, Karatsai O, Żuchowska A, Wróblewski W, Borys F, Lehka L, Rędowicz MJ, Koszytkowska-Stawińska M. Development of 5-fluorouracil-dichloroacetate mutual prodrugs as anticancer agents. Bioorg Chem 2023; 140:106784. [PMID: 37639758 DOI: 10.1016/j.bioorg.2023.106784] [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: 06/02/2023] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 08/31/2023]
Abstract
5-Fluorouracil (5-FU) is one of the most widely applied chemotherapeutic agents with a broad spectrum of activity. However, despite this versatile activity, its use poses many limitations. Herein, novel derivatives of 5-FU and dichloroacetic acid have been designed and synthesized as a new type of codrugs, also known as mutual prodrugs, to overcome the drawbacks of 5-FU and enhance its therapeutic efficiency. The stability of the obtained compounds has been tested at various pH values using different analytical techniques, namely HPLC and potentiometry. The antiproliferative activity of the new 5-FU derivatives was assessed in vitro on SK-MEL-28 and WM793 human melanoma cell lines in 2D culture as well as on A549 human lung carcinoma, MDA-MB-231 breast adenocarcinoma, LL24 normal lung tissue, and HMF normal breast tissue as a multicellular 3D spheroid model cultured in standard (static) conditions and with the use of microfluidic systems, which to a great extent resembles the in vivo environment. In all cases, new mutual prodrugs showed a higher cytotoxic activity toward cancer models and lower to normal cell models than the parent 5-FU itself.
Collapse
Affiliation(s)
- Ewa Mironiuk-Puchalska
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland.
| | - Olena Karatsai
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | - Agnieszka Żuchowska
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Wojciech Wróblewski
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Filip Borys
- Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego St., 00-664 Warsaw, Poland
| | - Lilya Lehka
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology Polish Academy of Science, 3 Pasteur St., 02-093-Warsaw, Poland
| | | |
Collapse
|
45
|
Teixeira SG, Houeto P, Gattacceca F, Petitcollot N, Debruyne D, Guerbet M, Guillemain J, Fabre I, Louin G, Salomon V. National reflection on organs-on-chip for drug development: New regulatory challenges. Toxicol Lett 2023; 388:1-12. [PMID: 37776962 DOI: 10.1016/j.toxlet.2023.09.011] [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: 07/19/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023]
Abstract
Organs-on-chip (OoC) are innovative and promising in vitro models, particularly in the process of developing new drugs, to improve predictivity of preclinical studies in humans. However, a lack of regulatory consensus on acceptance criteria and standards around these technologies currently hinders their adoption and implementation by end-users. A reflection has been conducted at the National Agency for Medicines and Health products safety (ANSM) in order to address this issue, which has gained momentum at the international level in recent years. If the subject of OoC is of international interest, France is also in the process of structuring an OoC network, in order to best support the emergence of this new technological innovation. Focusing on liver-on-a-chip, the authors drafted a first list of regulatory requirements to help standardize these devices and their use. Technological and biological relevance of liver-on-a-chip was also evaluated, in comparison with current in vitro and in vivo models, based on the available literature. The authors offer an analysis of the current scientific and regulatory situation, highlighting the key regulatory issues for the future.
Collapse
Affiliation(s)
- Sonia Gomes Teixeira
- French National Agency for Medicines and Health Products Safety (ANSM), 143/147 Boulevard Anatole France, 93285 Saint-Denis, France
| | - Paul Houeto
- French National Agency for Medicines and Health Products Safety (ANSM), 143/147 Boulevard Anatole France, 93285 Saint-Denis, France.
| | - Florence Gattacceca
- External Experts of Permanent Scientific Committee (PSC) of French National Agency for Medicines and Health Products Safety (ANSM), 143/147 Boulevard Anatole France, 93285 Saint-Denis, France
| | - Nicole Petitcollot
- External Experts of Permanent Scientific Committee (PSC) of French National Agency for Medicines and Health Products Safety (ANSM), 143/147 Boulevard Anatole France, 93285 Saint-Denis, France
| | - Danièle Debruyne
- External Experts of Permanent Scientific Committee (PSC) of French National Agency for Medicines and Health Products Safety (ANSM), 143/147 Boulevard Anatole France, 93285 Saint-Denis, France
| | - Michel Guerbet
- External Experts of Permanent Scientific Committee (PSC) of French National Agency for Medicines and Health Products Safety (ANSM), 143/147 Boulevard Anatole France, 93285 Saint-Denis, France
| | - Joël Guillemain
- External Experts of Permanent Scientific Committee (PSC) of French National Agency for Medicines and Health Products Safety (ANSM), 143/147 Boulevard Anatole France, 93285 Saint-Denis, France
| | - Isabelle Fabre
- French National Agency for Medicines and Health Products Safety (ANSM), 143/147 Boulevard Anatole France, 93285 Saint-Denis, France
| | - Gaelle Louin
- French National Agency for Medicines and Health Products Safety (ANSM), 143/147 Boulevard Anatole France, 93285 Saint-Denis, France
| | - Valérie Salomon
- French National Agency for Medicines and Health Products Safety (ANSM), 143/147 Boulevard Anatole France, 93285 Saint-Denis, France
| |
Collapse
|
46
|
Shang Y, Liu R, Gan J, Yang Y, Sun L. Construction of cardiac fibrosis for biomedical research. SMART MEDICINE 2023; 2:e20230020. [PMID: 39188350 PMCID: PMC11235890 DOI: 10.1002/smmd.20230020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/22/2023] [Indexed: 08/28/2024]
Abstract
Cardiac remodeling is critical for effective tissue recuperation, nevertheless, excessive formation and deposition of extracellular matrix components can result in the onset of cardiac fibrosis. Despite the emergence of novel therapies, there are still no lifelong therapeutic solutions for this issue. Understanding the detrimental cardiac remodeling may aid in the development of innovative treatment strategies to prevent or reverse fibrotic alterations in the heart. Further combining the latest understanding of disease pathogenesis with cardiac tissue engineering has provided the conversion of basic laboratory studies into the therapy of cardiac fibrosis patients as an increasingly viable prospect. This review presents the current main mechanisms and the potential tissue engineering of cardiac fibrosis. Approaches using biomedical materials-based cardiac constructions are reviewed to consider key issues for simulating in vitro cardiac fibrosis, outlining a future perspective for preclinical applications.
Collapse
Affiliation(s)
- Yixuan Shang
- Department of Medical Supplies SupportNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Rui Liu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Jingjing Gan
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Yuzhi Yang
- Department of Medical Supplies SupportNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Lingyun Sun
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| |
Collapse
|
47
|
Chitrangi S, Vaity P, Jamdar A, Bhatt S. Patient-derived organoids for precision oncology: a platform to facilitate clinical decision making. BMC Cancer 2023; 23:689. [PMID: 37479967 PMCID: PMC10362580 DOI: 10.1186/s12885-023-11078-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/16/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND Despite recent advances in research, there are still critical lacunae in our basic understanding of the cause, pathogenesis, and natural history of many cancers, especially heterogeneity in patient response to drugs and mediators in the transition from malignant to invasive phenotypes. The explication of the pathogenesis of cancer has been constrained by limited access to patient samples, tumor heterogeneity and lack of reliable biological models. Amelioration in cancer treatment depends on further understanding of the etiologic, genetic, biological, and clinical heterogeneity of tumor microenvironment. Patient-derived organoids recapitulate the basic features of primary tumors, including histological complexity and genetic heterogeneity, which is instrumental in predicting patient response to drugs. METHODS Human iPSCs from healthy donors, breast and ovarian cancer patients were successfully differentiated towards isogenic hepatic, cardiac, neural and endothelial lineages. Multicellular organoids were established using Primary cells isolated from tumor tissues, histologically normal tissues adjacent to the tumors (NATs) and adipose tissues (source of Mesenchymal Stem Cells) from ovarian and breast cancer patients. Further these organoids were propagated and used for drug resistance/sensitivity studies. RESULTS Ovarian and breast cancer patients' organoids showed heterogeneity in drug resistance and sensitivity. iPSCs-derived cardiomyocytes, hepatocytes and neurons showed donor-to-donor variability of chemotherapeutic drug sensitivity in ovarian cancer patients, breast cancer patients and healthy donors. CONCLUSION We report development of a novel integrated platform to facilitate clinical decision-making using the patient's primary cells, iPSCs and derivatives, to clinically relevant models for oncology research.
Collapse
Affiliation(s)
- Swati Chitrangi
- Department of Integrated Drug Discovery and Development, Yashraj Biotechnology Limited, C-232 and C-113, TTC Industrial Area, MIDC, Pawane, Maharashtra, 400705, India
| | - Pooja Vaity
- Department of Integrated Drug Discovery and Development, Yashraj Biotechnology Limited, C-232 and C-113, TTC Industrial Area, MIDC, Pawane, Maharashtra, 400705, India
| | - Aishwarya Jamdar
- Department of Integrated Drug Discovery and Development, Yashraj Biotechnology Limited, C-232 and C-113, TTC Industrial Area, MIDC, Pawane, Maharashtra, 400705, India
| | - Shweta Bhatt
- Department of Integrated Drug Discovery and Development, Yashraj Biotechnology Limited, C-232 and C-113, TTC Industrial Area, MIDC, Pawane, Maharashtra, 400705, India.
- Yashraj Biotechnology GmbH, Uhlandstraße 20-25, 10623, Berlin, Germany.
- Yashraj Biotechnology Limited, 8, The Green STE A, Dover, Delaware State, 19901, USA.
| |
Collapse
|
48
|
Portugal-Cohen M, Cohen D, Kohen R, Oron M. Exploitation of alternative skin models from academia to industry: proposed functional categories to answer needs and regulation demands. Front Physiol 2023; 14:1215266. [PMID: 37334052 PMCID: PMC10272927 DOI: 10.3389/fphys.2023.1215266] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Affiliation(s)
| | - Dror Cohen
- DermAb.io, Haifa, Israel
- The Myers Skin Research Laboratory, Faculty of Medicine, School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ron Kohen
- The Myers Skin Research Laboratory, Faculty of Medicine, School of Pharmacy, Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | |
Collapse
|
49
|
Zhang W, Liu QY, Haqqani AS, Liu Z, Sodja C, Leclerc S, Baumann E, Delaney CE, Brunette E, Stanimirovic DB. Differential Expression of ABC Transporter Genes in Brain Vessels vs. Peripheral Tissues and Vessels from Human, Mouse and Rat. Pharmaceutics 2023; 15:pharmaceutics15051563. [PMID: 37242805 DOI: 10.3390/pharmaceutics15051563] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/13/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND ATP-binding cassette (ABC) transporters comprise a superfamily of genes encoding membrane proteins with nucleotide-binding domains (NBD). These transporters, including drug efflux across the blood-brain barrier (BBB), carry a variety of substrates through plasma membranes against substrate gradients, fueled by hydrolyzing ATP. The expression patterns/enrichment of ABC transporter genes in brain microvessels compared to peripheral vessels and tissues are largely uncharacterized. METHODS In this study, the expression patterns of ABC transporter genes in brain microvessels, peripheral tissues (lung, liver and spleen) and lung vessels were investigated using RNA-seq and WesTM analyses in three species: human, mouse and rat. RESULTS The study demonstrated that ABC drug efflux transporter genes (including ABCB1, ABCG2, ABCC4 and ABCC5) were highly expressed in isolated brain microvessels in all three species studied; the expression of ABCB1, ABCG2, ABCC1, ABCC4 and ABCC5 was generally higher in rodent brain microvessels compared to those of humans. In contrast, ABCC2 and ABCC3 expression was low in brain microvessels, but high in rodent liver and lung vessels. Overall, most ABC transporters (with the exception of drug efflux transporters) were enriched in peripheral tissues compared to brain microvessels in humans, while in rodent species, additional ABC transporters were found to be enriched in brain microvessels. CONCLUSIONS This study furthers the understanding of species similarities and differences in the expression patterns of ABC transporter genes; this is important for translational studies in drug development. In particular, CNS drug delivery and toxicity may vary among species depending on their unique profiles of ABC transporter expression in brain microvessels and BBB.
Collapse
Affiliation(s)
- Wandong Zhang
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON K1A 0R6, Canada
| | - Qing Yan Liu
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON K1A 0R6, Canada
| | - Arsalan S Haqqani
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON K1A 0R6, Canada
| | - Ziying Liu
- Scientific Data Mining/Digital Technology Research Centre, National Research Council of Canada, Ottawa, ON K1A 0R6, Canada
| | - Caroline Sodja
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON K1A 0R6, Canada
| | - Sonia Leclerc
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON K1A 0R6, Canada
| | - Ewa Baumann
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON K1A 0R6, Canada
| | - Christie E Delaney
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON K1A 0R6, Canada
| | - Eric Brunette
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON K1A 0R6, Canada
| | - Danica B Stanimirovic
- Human Health Therapeutics Research Centre, National Research Council of Canada, Ottawa, ON K1A 0R6, Canada
| |
Collapse
|
50
|
Tsai HF, Podder S, Chen PY. Microsystem Advances through Integration with Artificial Intelligence. MICROMACHINES 2023; 14:826. [PMID: 37421059 PMCID: PMC10141994 DOI: 10.3390/mi14040826] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 07/09/2023]
Abstract
Microfluidics is a rapidly growing discipline that involves studying and manipulating fluids at reduced length scale and volume, typically on the scale of micro- or nanoliters. Under the reduced length scale and larger surface-to-volume ratio, advantages of low reagent consumption, faster reaction kinetics, and more compact systems are evident in microfluidics. However, miniaturization of microfluidic chips and systems introduces challenges of stricter tolerances in designing and controlling them for interdisciplinary applications. Recent advances in artificial intelligence (AI) have brought innovation to microfluidics from design, simulation, automation, and optimization to bioanalysis and data analytics. In microfluidics, the Navier-Stokes equations, which are partial differential equations describing viscous fluid motion that in complete form are known to not have a general analytical solution, can be simplified and have fair performance through numerical approximation due to low inertia and laminar flow. Approximation using neural networks trained by rules of physical knowledge introduces a new possibility to predict the physicochemical nature. The combination of microfluidics and automation can produce large amounts of data, where features and patterns that are difficult to discern by a human can be extracted by machine learning. Therefore, integration with AI introduces the potential to revolutionize the microfluidic workflow by enabling the precision control and automation of data analysis. Deployment of smart microfluidics may be tremendously beneficial in various applications in the future, including high-throughput drug discovery, rapid point-of-care-testing (POCT), and personalized medicine. In this review, we summarize key microfluidic advances integrated with AI and discuss the outlook and possibilities of combining AI and microfluidics.
Collapse
Affiliation(s)
- Hsieh-Fu Tsai
- Department of Biomedical Engineering, Chang Gung University, Taoyuan City 333, Taiwan;
- Department of Neurosurgery, Chang Gung Memorial Hospital, Keelung, Keelung City 204, Taiwan
- Center for Biomedical Engineering, Chang Gung University, Taoyuan City 333, Taiwan
| | - Soumyajit Podder
- Department of Biomedical Engineering, Chang Gung University, Taoyuan City 333, Taiwan;
| | - Pin-Yuan Chen
- Department of Biomedical Engineering, Chang Gung University, Taoyuan City 333, Taiwan;
- Department of Neurosurgery, Chang Gung Memorial Hospital, Keelung, Keelung City 204, Taiwan
| |
Collapse
|