1
|
Liu X, Zhou Z, Zhang Y, Zhong H, Cai X, Guan R. Recent progress on the organoids: Techniques, advantages and applications. Biomed Pharmacother 2025; 185:117942. [PMID: 40043462 DOI: 10.1016/j.biopha.2025.117942] [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: 12/11/2024] [Revised: 01/30/2025] [Accepted: 02/24/2025] [Indexed: 03/23/2025] Open
Abstract
Organoids are a cutting-edge technology in the life sciences field, with applications in precision medicine, bionic organs, and toxicological evaluations of chemicals. Their 3D structure closely resembles that of real organs, allowing more accurate functional mimicry. The 3D organoid culture system can simulate the growth state of cells in vivo and establish a suspension culture system for organoid 3D culture by using scaffold-less or scaffold technology to avoid direct contact between cells and plastic culture vessels. Furthermore, organoids can simulate the pathophysiological state of tissues and organs in vitro. This paper primarily discusses the construction methodologies, as well as the advantages and disadvantages of 3D culture systems for both scaffold-free organoids and scaffolded organoids. This review also summarizes the application of organoid models in chemical toxicology evaluation, drug screening and functional evaluation, establishment of in vitro disease models, and research on disease occurrence and potential mechanisms. The aim is to provide a reference for the research and practical applications of organoid-related scientific fields.
Collapse
Affiliation(s)
- Xiaofeng Liu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Zhiyuan Zhou
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yao Zhang
- Zhejiang Provincial Key Lab for Chem and Bio Processing Technology of Farm Produces, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, China
| | - Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Xiulei Cai
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Rongfa Guan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China; Moganshan Institute ZJUT, Kangqian District, Deqing 313200, China.
| |
Collapse
|
2
|
Ashander LM, Lidgerwood GE, Lumsden AL, Furtado JM, Pébay A, Smith JR. Human Retinal Organoid Model of Ocular Toxoplasmosis. Pathogens 2025; 14:286. [PMID: 40137771 PMCID: PMC11945118 DOI: 10.3390/pathogens14030286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
The health burden of ocular toxoplasmosis is substantial, and there is an unmet need for safe and curative anti-microbial drugs. One major barrier to research on new therapeutics is the lack of in vitro human-based models beyond two-dimensional cultured cells and tissue explants. We aimed to address this research gap by establishing a human retinal organoid model of ocular toxoplasmosis. Retinal organoids, generated from human induced pluripotent stem cells and grown to two stages of organization, were incubated with a suspension of live or heat-killed GT-1 strain T. gondii tachyzoites, or medium without tachyzoites. Both developing (1 month post-isolation) and matured (6 months post-isolation) organoids were susceptible to infection. Spread of live parasites from the margin to the entire organoid over 1 week was indicated by immunolabelling for T. gondii surface antigen 1. This progression was accompanied by changes in the levels of selected tachyzoite transcripts-SAG1, GRA6, and ROP16-and human cytokine transcripts-CCL2, CXCL8, CXCL10, and IL6-in infected versus control conditions. Our human retinal organoid model of ocular toxoplasmosis offers the opportunity for many future lines of study, including tachyzoite interactions with retinal cell populations and leukocyte subsets, parasite stage progression, and disease processes of different T. gondii strains, as well as drug testing.
Collapse
Affiliation(s)
- Liam M. Ashander
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia; (L.M.A.); (A.L.L.)
| | - Grace E. Lidgerwood
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia; (G.E.L.); (A.P.)
| | - Amanda L. Lumsden
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia; (L.M.A.); (A.L.L.)
| | - João M. Furtado
- Division of Ophthalmology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, São Paulo, Brazil;
| | - Alice Pébay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia; (G.E.L.); (A.P.)
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Justine R. Smith
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia; (L.M.A.); (A.L.L.)
| |
Collapse
|
3
|
Ahidjo N, Seke Etet PF, Ngarka L, Maidawa Yaya F, Ndianteng EW, Eyenga Nna AL, Meka’a Zang LY, Kemmo C, Nwasike CNC, Yonkeu Tatchou FG, Njamnshi WY, Nfor LN, Tsouh Fokou PV, Djiogue S, Fekam Boyom F, Ngadjui BT, Njamnshi AK. Effects of diet and ovariectomy on Toxoplasma gondii brain infection: functional alterations and neuronal loss in rats. Brain Commun 2024; 7:fcae441. [PMID: 39741781 PMCID: PMC11686407 DOI: 10.1093/braincomms/fcae441] [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: 12/29/2023] [Revised: 10/14/2024] [Accepted: 12/26/2024] [Indexed: 01/03/2025] Open
Abstract
Epidemiological evidence associates Toxoplasma gondii latent infection with the development of neuropsychiatric disorders, and various immunological and environmental factors play key pathophysiological roles through host immune response alterations. We investigated the cognitive and motor alterations occurring in the terminal stage of T. gondii infection in rats, and whether a low-protein diet, a high-fat diet or ovariectomy may accelerate their development, given the role of malnutrition and menopause on immunity and resistance to infection. In two sets of experiments, 2-month-old (157.5 ± 4.3 g, n = 42) male (n = 18) and female (n = 24) Wistar rats were infected with T. gondii (ATCC 40050). Open-field and elevated plus maze tests were performed in the terminal stage of infection first and then in the early stage in low-protein diet-fed, high-fat diet-fed and ovariectomized infected rats. Late-stage (90 days) infected and early-stage (17 days) low-protein diet-fed groups showed significant decreases in body weight (42.42%↓, P = 0.016 and 57.14%↓, P < 0.001 versus non-infected, respectively), increases in body temperature (P = 0.001 and P < 0.001, respectively), decreases in blood glucose levels (P = 0.006 and P = 0.020, respectively), signs of cognitive and motor impairment and lower neuron counts. The alterations observed in high-fat diet-fed and ovariectomized infected animals were milder. Low-protein diet feeding to T. gondii-infected rats accelerated the occurrence of the infection terminal stage. Thus, a diet low in proteins could transform a slow early-stage T. gondii infection into an active neurotoxoplasmosis with neuropsychiatric manifestations and possible neurodegeneration in rats.
Collapse
Affiliation(s)
- Nene Ahidjo
- Brain Research Africa Initiative (BRAIN), P.O. Box 25625, Yaoundé, Cameroon
| | - Paul F Seke Etet
- Brain Research Africa Initiative (BRAIN), P.O. Box 25625, Yaoundé, Cameroon
- Neuroscience Laboratory, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 25625, Yaoundé, Cameroon
- Basic and Translational Research Unit, Center for Sustainable Health and Development, Garoua, Cameroon
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Garoua, P.O. Box 346 Garoua, Garoua, Cameroon
| | - Leonard Ngarka
- Brain Research Africa Initiative (BRAIN), P.O. Box 25625, Yaoundé, Cameroon
- Neuroscience Laboratory, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 25625, Yaoundé, Cameroon
| | - Frederic Maidawa Yaya
- Neuroscience Laboratory, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 25625, Yaoundé, Cameroon
- Basic and Translational Research Unit, Center for Sustainable Health and Development, Garoua, Cameroon
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Garoua, P.O. Box 346 Garoua, Garoua, Cameroon
| | - Ethel W Ndianteng
- Neuroscience Laboratory, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 25625, Yaoundé, Cameroon
| | - Aude L Eyenga Nna
- Neuroscience Laboratory, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 25625, Yaoundé, Cameroon
| | - Luc Yvan Meka’a Zang
- Neuroscience Laboratory, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 25625, Yaoundé, Cameroon
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| | - Christelle Kemmo
- Neuroscience Laboratory, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 25625, Yaoundé, Cameroon
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| | - Caroline N C Nwasike
- Neuroscience Laboratory, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 25625, Yaoundé, Cameroon
| | - Floriane G Yonkeu Tatchou
- Neuroscience Laboratory, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 25625, Yaoundé, Cameroon
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| | - Wepnyu Y Njamnshi
- Brain Research Africa Initiative (BRAIN), P.O. Box 25625, Yaoundé, Cameroon
- Neuroscience Laboratory, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 25625, Yaoundé, Cameroon
- Division of Health Operations Research, Ministry of Public Health, P. O. Box 1937, Yaoundé, Cameroon
| | - Leonard N Nfor
- Brain Research Africa Initiative (BRAIN), P.O. Box 25625, Yaoundé, Cameroon
- Neuroscience Laboratory, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 25625, Yaoundé, Cameroon
| | - Patrick V Tsouh Fokou
- Antimicrobial & Biocontrol Agents Unit, Laboratory for Phytobiochemistry and Medicinal Plants Studies (LPMPS), The University of Yaoundé I, P. O. Box 812, Yaoundé, Cameroon
- Department of Biochemistry, Faculty of Science, University of Bamenda, P.O. Box 39, Bamenda, Cameroon
- Advanced Research and Health Innovation Hub, P.O. Box 20133, Yaoundé, Cameroon
| | - Sefirin Djiogue
- Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| | - Fabrice Fekam Boyom
- Antimicrobial & Biocontrol Agents Unit, Laboratory for Phytobiochemistry and Medicinal Plants Studies (LPMPS), The University of Yaoundé I, P. O. Box 812, Yaoundé, Cameroon
- Advanced Research and Health Innovation Hub, P.O. Box 20133, Yaoundé, Cameroon
| | - Bonaventure T Ngadjui
- Department of Organic Chemistry, The University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon
| | - Alfred K Njamnshi
- Brain Research Africa Initiative (BRAIN), P.O. Box 25625, Yaoundé, Cameroon
- Neuroscience Laboratory, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 25625, Yaoundé, Cameroon
| |
Collapse
|
4
|
Smirnova L, Hartung T. The Promise and Potential of Brain Organoids. Adv Healthc Mater 2024; 13:e2302745. [PMID: 38252094 DOI: 10.1002/adhm.202302745] [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: 08/20/2023] [Revised: 12/22/2023] [Indexed: 01/23/2024]
Abstract
Brain organoids are 3D in vitro culture systems derived from human pluripotent stem cells that self-organize to model features of the (developing) human brain. This review examines the techniques behind organoid generation, their current and potential applications, and future directions for the field. Brain organoids possess complex architecture containing various neural cell types, synapses, and myelination. They have been utilized for toxicology testing, disease modeling, infection studies, personalized medicine, and gene-environment interaction studies. An emerging concept termed Organoid Intelligence (OI) combines organoids with artificial intelligence systems to generate learning and memory, with the goals of modeling cognition and enabling biological computing applications. Brain organoids allow neuroscience studies not previously achievable with traditional techniques, and have the potential to transform disease modeling, drug development, and the understanding of human brain development and disorders. The aspirational vision of OI parallels the origins of artificial intelligence, and efforts are underway to map a roadmap toward its realization. In summary, brain organoids constitute a disruptive technology that is rapidly advancing and gaining traction across multiple disciplines.
Collapse
Affiliation(s)
- Lena Smirnova
- Center for Alternatives to Animal Testing (CAAT), Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, 615 N Wolfe St, Baltimore, MD, 21205, USA
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, 615 N Wolfe St, Baltimore, MD, 21205, USA
- CAAT-Europe, University of Konstanz, Universitätsstr. 10, 78464, Konstanz, BW, Germany
| |
Collapse
|
5
|
Yan Y, Cho AN. Human Brain In Vitro Model for Pathogen Infection-Related Neurodegeneration Study. Int J Mol Sci 2024; 25:6522. [PMID: 38928228 PMCID: PMC11204318 DOI: 10.3390/ijms25126522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/21/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Recent advancements in stem cell biology and tissue engineering have revolutionized the field of neurodegeneration research by enabling the development of sophisticated in vitro human brain models. These models, including 2D monolayer cultures, 3D organoids, organ-on-chips, and bioengineered 3D tissue models, aim to recapitulate the cellular diversity, structural organization, and functional properties of the native human brain. This review highlights how these in vitro brain models have been used to investigate the effects of various pathogens, including viruses, bacteria, fungi, and parasites infection, particularly in the human brain cand their subsequent impacts on neurodegenerative diseases. Traditional studies have demonstrated the susceptibility of different 2D brain cell types to infection, elucidated the mechanisms underlying pathogen-induced neuroinflammation, and identified potential therapeutic targets. Therefore, current methodological improvement brought the technology of 3D models to overcome the challenges of 2D cells, such as the limited cellular diversity, incomplete microenvironment, and lack of morphological structures by highlighting the need for further technological advancements. This review underscored the significance of in vitro human brain cell from 2D monolayer to bioengineered 3D tissue model for elucidating the intricate dynamics for pathogen infection modeling. These in vitro human brain cell enabled researchers to unravel human specific mechanisms underlying various pathogen infections such as SARS-CoV-2 to alter blood-brain-barrier function and Toxoplasma gondii impacting neural cell morphology and its function. Ultimately, these in vitro human brain models hold promise as personalized platforms for development of drug compound, gene therapy, and vaccine. Overall, we discussed the recent progress in in vitro human brain models, their applications in studying pathogen infection-related neurodegeneration, and future directions.
Collapse
Affiliation(s)
- Yuwei Yan
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW 2008, Australia;
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2050, Australia
| | - Ann-Na Cho
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW 2008, Australia;
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2050, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
6
|
Yin D, Jiang N, Cheng C, Sang X, Feng Y, Chen R, Chen Q. Protein Lactylation and Metabolic Regulation of the Zoonotic Parasite Toxoplasma gondii. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:1163-1181. [PMID: 36216028 PMCID: PMC11082259 DOI: 10.1016/j.gpb.2022.09.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 09/06/2022] [Accepted: 09/29/2022] [Indexed: 06/16/2023]
Abstract
The biology of Toxoplasma gondii, the causative pathogen of one of the most widespread parasitic diseases (toxoplasmosis), remains poorly understood. Lactate, which is derived from glucose metabolism, is not only an energy source in a variety of organisms, including T. gondii, but also a regulatory molecule that participates in gene activation and protein function. Lysine lactylation (Kla) is a type of post-translational modifications (PTMs) that has been recently associated with chromatin remodeling; however, Kla of histone and non-histone proteins has not yet been studied in T. gondii. To examine the prevalence and function of lactylation in T. gondii parasites, we mapped the lactylome of proliferating tachyzoite cells and identified 1964 Kla sites on 955 proteins in the T. gondii RH strain. Lactylated proteins were distributed in multiple subcellular compartments and were closely related to a wide variety of biological processes, including mRNA splicing, glycolysis, aminoacyl-tRNA biosynthesis, RNA transport, and many signaling pathways. We also performed a chromatin immunoprecipitation sequencing (ChIP-seq) analysis using a lactylation-specific antibody and found that the histones H4K12la and H3K14la were enriched in the promoter and exon regions of T. gondii associated with microtubule-based movement and cell invasion. We further confirmed the delactylase activity of histone deacetylases TgHDAC2-4, and found that treatment with anti-histone acetyltransferase (TgMYST-A) antibodies profoundly reduced protein lactylation in T. gondii. This study offers the first dataset of the global lactylation proteome and provides a basis for further dissecting the functional biology of T. gondii.
Collapse
Affiliation(s)
- Deqi Yin
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China
| | - Chang Cheng
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China
| | - Xiaoyu Sang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China
| | - Ying Feng
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China
| | - Ran Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110166, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang 110866, China.
| |
Collapse
|
7
|
Michalski C, Wen Z. Leveraging iPSC technology to assess neuro-immune interactions in neurological and psychiatric disorders. Front Psychiatry 2023; 14:1291115. [PMID: 38025464 PMCID: PMC10672983 DOI: 10.3389/fpsyt.2023.1291115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Communication between the immune and the nervous system is essential for human brain development and homeostasis. Disruption of this intricately regulated crosstalk can lead to neurodevelopmental, psychiatric, or neurodegenerative disorders. While animal models have been essential in characterizing the role of neuroimmunity in development and disease, they come with inherent limitations due to species specific differences, particularly with regard to microglia, the major subset of brain resident immune cells. The advent of induced pluripotent stem cell (iPSC) technology now allows the development of clinically relevant models of the central nervous system that adequately reflect human genetic architecture. This article will review recent publications that have leveraged iPSC technology to assess neuro-immune interactions. First, we will discuss the role of environmental stressors such as neurotropic viruses or pro-inflammatory cytokines on neuronal and glial function. Next, we will review how iPSC models can be used to study genetic risk factors in neurological and psychiatric disorders. Lastly, we will evaluate current challenges and future potential for iPSC models in the field of neuroimmunity.
Collapse
Affiliation(s)
- Christina Michalski
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
8
|
Feix AS, Cruz-Bustos T, Ruttkowski B, Joachim A. In vitro cultivation methods for coccidian parasite research. Int J Parasitol 2023; 53:477-489. [PMID: 36400306 DOI: 10.1016/j.ijpara.2022.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/29/2022] [Accepted: 10/09/2022] [Indexed: 11/17/2022]
Abstract
The subclass Coccidia comprises a large group of protozoan parasites, including important pathogens of humans and animals such as Toxoplasma gondii, Neospora caninum, Eimeria spp., and Cystoisospora spp. Their life cycle includes a switch from asexual to sexual stages and is often restricted to a single host species. Current research on coccidian parasites focuses on cell biology and the underlying mechanisms of protein expression and trafficking in different life stages, host cell invasion and host-parasite interactions. Furthermore, novel anticoccidial drug targets are evaluated. Given the variety of research questions and the requirement to reduce and replace animal experimentation, in vitro cultivation of Coccidia needs to be further developed and refined to meet these requirements. For these purposes, established culture systems are constantly improved. In addition, new in vitro culture systems lately gained considerable importance in research on Coccidia. Well established and optimized in vitro cultures of monolayer cells can support the viability and development of parasite stages and even allow completion of the life cycle in vitro, as shown for Cystoisospora suis and Eimeria tenella. Furthermore, new three-dimensional cell culture models are used for propagation of Cryptosporidium spp. (close relatives of the coccidians), and the infection of three-dimensional organoids with T. gondii also gained popularity as the interaction between the parasite and host tissue can be studied in more detail. The latest advances in three-dimensional culture systems are organ-on-a-chip models, that to date have only been tested for T. gondii but promise to accelerate research in other coccidians. Lastly, the completion of the life cycle of C. suis and Cryptosporidium parvum was reported to continue in a host cell-free environment following the first occurrence of asexual stages. Such axenic cultures are becoming increasingly available and open new avenues for research on parasite life cycle stages and novel intervention strategies.
Collapse
Affiliation(s)
- Anna Sophia Feix
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, A-1210 Vienna, Austria.
| | - Teresa Cruz-Bustos
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, A-1210 Vienna, Austria
| | - Bärbel Ruttkowski
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, A-1210 Vienna, Austria
| | - Anja Joachim
- Institute of Parasitology, Department of Pathobiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, A-1210 Vienna, Austria
| |
Collapse
|
9
|
de Oliveira LF, Filho DM, Marques BL, Maciel GF, Parreira RC, do Carmo Neto JR, Da Silva PEF, Guerra RO, da Silva MV, Santiago HDC, Birbrair A, Kihara AH, Dias da Silva VJ, Glaser T, Resende RR, Ulrich H. Organoids as a novel tool in modelling infectious diseases. Semin Cell Dev Biol 2023; 144:87-96. [PMID: 36182613 DOI: 10.1016/j.semcdb.2022.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/04/2022] [Indexed: 11/23/2022]
Abstract
Infectious diseases worldwide affect human health and have important societal impacts. A better understanding of infectious diseases is urgently needed. In vitro and in vivo infection models have brought notable contributions to the current knowledge of these diseases. Organoids are multicellular culture systems resembling tissue architecture and function, recapitulating many characteristics of human disease and elucidating mechanisms of host-infectious agent interactions in the respiratory and gastrointestinal systems, the central nervous system and the skin. Here, we discuss the applicability of the organoid technology for modeling pathogenesis, host response and features, which can be explored for the development of preventive and therapeutic treatments.
Collapse
Affiliation(s)
- Lucas Felipe de Oliveira
- Departamento de Fisiologia, Instituto de Ciências Biológicas e Naturais, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil; Instituto Nacional de Ciência e Tecnologia de Medicina Regenerativa, Rio de Janeiro, RJ, Brazil
| | - Daniel Mendes Filho
- Departamento de Fisiologia, Escola Médica de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Bruno Lemes Marques
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal deGoiás, Goiânia, GO, Brazil
| | | | | | - José Rodrigues do Carmo Neto
- Departamento de Biociência e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | | | - Rhanoica Oliveira Guerra
- Departamento de Microbiologia, Imunologia eParasitologia, Instituto de Ciências Naturais e Biológicas, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Marcos Vinicius da Silva
- Departamento de Microbiologia, Imunologia eParasitologia, Instituto de Ciências Naturais e Biológicas, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Helton da Costa Santiago
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Belo Horizonte, MG, Brazil
| | - Alexander Birbrair
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Radiology, Columbia University Medical Center, New York, NY, USA; Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Belo Horizonte, MG, Brazil
| | - Alexandre H Kihara
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Valdo José Dias da Silva
- Departamento de Fisiologia, Instituto de Ciências Biológicas e Naturais, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil; Instituto Nacional de Ciência e Tecnologia de Medicina Regenerativa, Rio de Janeiro, RJ, Brazil
| | - Talita Glaser
- Departmento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Belo Horizonte, MG, Brazil
| | - Henning Ulrich
- Instituto Nacional de Ciência e Tecnologia de Medicina Regenerativa, Rio de Janeiro, RJ, Brazil; Departmento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil.
| |
Collapse
|
10
|
Chandrasegaran P, Nabilla Lestari A, Sinton MC, Gopalakrishnan J, Quintana JF. Modelling host- Trypanosoma brucei gambiense interactions in vitro using human induced pluripotent stem cell-derived cortical brain organoids. F1000Res 2023; 12:437. [PMID: 37588058 PMCID: PMC10425695 DOI: 10.12688/f1000research.131507.2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/25/2023] [Indexed: 08/18/2023] Open
Abstract
Background: Sleeping sickness is caused by the extracellular parasite Trypanosoma brucei and is associated with neuroinflammation and neuropsychiatric disorders, including disruption of sleep/wake patterns, and is now recognised as a circadian disorder. Sleeping sickness is traditionally studied using murine models of infection due to the lack of alternative in vitro systems that fully recapitulate the cellular diversity and functionality of the human brain. The aim of this study is to develop a much-needed in vitro system that reduces and replaces live animals for the study of infections in the central nervous system, using sleeping sickness as a model infection. Methods: We developed a co-culture system using induced pluripotent stem cell (iPSC)-derived cortical human brain organoids and the human pathogen T. b. gambiense to model host-pathogen interactions in vitro. Upon co-culture, we analysed the transcriptional responses of the brain organoids to T. b. gambiense over two time points. Results: We detected broad transcriptional changes in brain organoids exposed to T. b. gambiense, mainly associated with innate immune responses, chemotaxis, and blood vessel differentiation compared to untreated organoids. Conclusions: Our co-culture system provides novel, more ethical avenues to study host-pathogen interactions in the brain as alternative models to experimental infections in mice. Although our data support the use of brain organoids to model host-pathogen interactions during T. brucei infection as an alternative to in vivo models, future work is required to increase the complexity of the organoids ( e.g., addition of microglia and vasculature). We envision that the adoption of organoid systems is beneficial to researchers studying mechanisms of brain infection by protozoan parasites. Furthermore, organoid systems have the potential to be used to study other parasites that affect the brain significantly reducing the number of animals undergoing moderate and/or severe protocols associated with the study of neuroinflammation and brain infections.
Collapse
Affiliation(s)
- Praveena Chandrasegaran
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), University of Glasgow, Glasgow, Scotland, G61 1QH, UK
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, Scotland, G61 1QH, UK
| | - Agatha Nabilla Lestari
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), University of Glasgow, Glasgow, Scotland, G61 1QH, UK
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, Scotland, G61 1QH, UK
| | - Matthew C. Sinton
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), University of Glasgow, Glasgow, Scotland, G61 1QH, UK
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, Scotland, G61 1QH, UK
| | - Jay Gopalakrishnan
- Laboratory of Centrosome and Cytoskeleton Biology, Laboratory for Centrosome and Cytoskeleton Biology, Institute für Humangenetik, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität, Düsseldorf, 40225, Germany
| | - Juan F. Quintana
- School of Biodiversity, One Health, and Veterinary Medicine (SBOHVM), University of Glasgow, Glasgow, Scotland, G61 1QH, UK
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, Scotland, G61 1QH, UK
| |
Collapse
|
11
|
Kilpatrick S, Irwin C, Singh KK. Human pluripotent stem cell (hPSC) and organoid models of autism: opportunities and limitations. Transl Psychiatry 2023; 13:217. [PMID: 37344450 PMCID: PMC10284884 DOI: 10.1038/s41398-023-02510-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/09/2023] [Accepted: 06/05/2023] [Indexed: 06/23/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder caused by genetic or environmental perturbations during early development. Diagnoses are dependent on the identification of behavioral abnormalities that likely emerge well after the disorder is established, leaving critical developmental windows uncharacterized. This is further complicated by the incredible clinical and genetic heterogeneity of the disorder that is not captured in most mammalian models. In recent years, advancements in stem cell technology have created the opportunity to model ASD in a human context through the use of pluripotent stem cells (hPSCs), which can be used to generate 2D cellular models as well as 3D unguided- and region-specific neural organoids. These models produce profoundly intricate systems, capable of modeling the developing brain spatiotemporally to reproduce key developmental milestones throughout early development. When complemented with multi-omics, genome editing, and electrophysiology analysis, they can be used as a powerful tool to profile the neurobiological mechanisms underlying this complex disorder. In this review, we will explore the recent advancements in hPSC-based modeling, discuss present and future applications of the model to ASD research, and finally consider the limitations and future directions within the field to make this system more robust and broadly applicable.
Collapse
Affiliation(s)
- Savannah Kilpatrick
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Biochemistry and Biomedical Science, McMaster University, Hamilton, ON, Canada
| | - Courtney Irwin
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Karun K Singh
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada.
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
12
|
Warschkau D, Seeber F. Advances towards the complete in vitro life cycle of Toxoplasma gondii. Fac Rev 2023; 12:1. [PMID: 36846606 PMCID: PMC9944905 DOI: 10.12703/r/12-1] [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] [Indexed: 02/16/2023] Open
Abstract
The full life cycle of Toxoplasma gondii cannot be recapitulated in vitro, and access to certain stages, such as mature tissue cysts (bradyzoites) and oocysts (sporozoites), traditionally requires animal experiments. This has greatly hindered the study of the biology of these morphologically and metabolically distinct stages, which are essential for the infection of humans and animals. However, several breakthrough advances have been made in recent years towards obtaining these life stages in vitro, such as the discovery of several molecular factors that induce differentiation and commitment to the sexual cycle, and different culture methods that use, for example, myotubes and intestinal organoids to obtain mature bradyzoites and different sexual stages of the parasite. We review these novel tools and approaches, highlight their limitations and challenges, and discuss what research questions can already be answered with these models. We finally identify future routes for recapitulating the entire sexual cycle in vitro.
Collapse
Affiliation(s)
- David Warschkau
- FG16: Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institut, Berlin, Germany
| | - Frank Seeber
- FG16: Mycotic and Parasitic Agents and Mycobacteria, Robert Koch-Institut, Berlin, Germany
| |
Collapse
|
13
|
Abstract
Toxoplasma gondii infection in the central nervous system commonly occurs among immunodeficient patients. Its prevalence is high in countries with a high burden of HIV and low coverage of antiretroviral drugs. The brain is one of the predilections for T. gondii infection due to its low inflammatory reaction, and cerebral toxoplasmosis occurs solely due to the reactivation of a latent infection rather than a new infection. Several immune elements have recently been recognized to have an essential role in the immunopathogenesis of cerebral toxoplasmosis. Although real-time isothermal amplification, next-generation sequencing, and enzyme-linked aptamer assays from blood samples have been the recommended diagnostic tools in some in-vivo studies, a combination of clinical symptoms, serology examination, and neuroimaging are still the daily standard for the presumptive diagnosis of cerebral toxoplasmosis and early anti-toxoplasma administration. Clinical trials are needed to find a new therapy that is less likely to affect folate synthesis, have neuroprotective properties, or cure the latent phase of infection. The development of a vaccine is being extensively tested in animals, but its efficacy and safety for humans are still not proven.
Collapse
Affiliation(s)
- Sofiati Dian
- Department of Neurology, Faculty of Medicine, Universitas Padjdjaran/Hasan Sadikin Hospital, Bandung, Indonesia
- Health Research Unit, Faculty of Medicine, Padjadjaran University/Hasan Sadikin Hospital, Bandung, Indonesia
| | - Ahmad Rizal Ganiem
- Department of Neurology, Faculty of Medicine, Universitas Padjdjaran/Hasan Sadikin Hospital, Bandung, Indonesia
- Health Research Unit, Faculty of Medicine, Padjadjaran University/Hasan Sadikin Hospital, Bandung, Indonesia
| | - Savira Ekawardhani
- Parasitology Division, Department of Biomedical Sciences, Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| |
Collapse
|
14
|
dos Santos PV, de Toledo DNM, de Souza DMS, Menezes TP, Perucci LO, Silva ZM, Teixeira DC, Vieira EWR, de Andrade-Neto VF, Guimarães NS, Talvani A. The imbalance in the relationship between inflammatory and regulatory cytokines during gestational toxoplasmosis can be harmful to fetuses: A systematic review. Front Immunol 2023; 14:1074760. [PMID: 36742306 PMCID: PMC9889920 DOI: 10.3389/fimmu.2023.1074760] [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: 10/19/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
Objective To evaluate the available information on inflammatory and regulatory plasma mediators in pregnant women (PW) diagnosed with toxoplasmosis. Source: The PubMed, Embase, Scopus, and Lilacs databases were evaluated until October 2022. Study eligibility criteria: This review was carried out following the PRISMA and registered on the PROSPERO platform (CRD42020203951). Studies that reported inflammatory mediators in PW with toxoplasmosis were considered. Evaluation methods After excluding duplicate articles, two authors independently carried out the process of title and abstract exclusion, and a third resolved disagreements when necessary. The full text was evaluated to detect related articles. The extraction table was built from the following data: Author, year of publication, journal name and impact factors, country, study design, number of gestations and maternal age (years), gestational period, diagnosis of toxoplasmosis, levels of inflammatory markers, laboratory tests, and clinical significance. Methodological quality was assessed using Joanna Briggs Institute tools. Results Of the 1,024 studies reported, only eight were included. Of the 868 PW included in this review, 20.2% were IgM+/IgG- and 50.8% were IgM-/IgG+ to T. gondii, and 29.0% uninfected. Infected PW presented higher plasma levels ofIL-5, IL-6, IL-8, IL-17, CCL5, and IL-10. Regarding the methodological quality, four studies obtained high quality. Data from this review pointed out the maintenance of the inflammatory pattern during pregnancy with a closely related to the parasite. Conclusion Immune status in PW defined the course of the T. gondii infection, where the equilibrium between inflammatory and regulatory cytokines mitigated the harmful placenta and fetus effects. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD420203951.
Collapse
Affiliation(s)
- Priscilla Vilela dos Santos
- Laboratory of the Immunobiology of Inflammation, Department of Biological Sciences/Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Graduate Program in Health and Nutrition, School of Nutrition, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Débora Nonato Miranda de Toledo
- Laboratory of the Immunobiology of Inflammation, Department of Biological Sciences/Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Graduate Program in Health and Nutrition, School of Nutrition, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Débora Maria Soares de Souza
- Laboratory of the Immunobiology of Inflammation, Department of Biological Sciences/Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Graduate Program in Health and Nutrition, School of Nutrition, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Tatiana Prata Menezes
- Laboratory of the Immunobiology of Inflammation, Department of Biological Sciences/Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Graduate Program in Health and Nutrition, School of Nutrition, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Luiza Oliveira Perucci
- Laboratory of the Immunobiology of Inflammation, Department of Biological Sciences/Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - Zolder Marinho Silva
- Laboratory of the Immunobiology of Inflammation, Department of Biological Sciences/Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Graduate Program in Health and Nutrition, School of Nutrition, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | | | - Ed Wilson Rodrigues Vieira
- Department of Maternal and Child Nursing and Public Health, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Valter Ferreira de Andrade-Neto
- Laboratory of Malaria and Toxoplasmosis Biology, Department of Microbiology and Parasitology, Federal University of the Rio Grande do Norte, Natal, RN, Brazil
| | - Nathalia Sernizon Guimarães
- Graduate Program in Health and Nutrition, School of Nutrition, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
| | - André Talvani
- Laboratory of the Immunobiology of Inflammation, Department of Biological Sciences/Institute of Exact and Biological Sciences (ICEB), Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Graduate Program in Health and Nutrition, School of Nutrition, Federal University of Ouro Preto, Ouro Preto, MG, Brazil
- Graduate Program of Health Science, Infectiology and Tropical Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
15
|
Halonen SK. Use of in vitro derived human neuronal models to study host-parasite interactions of Toxoplasma gondii in neurons and neuropathogenesis of chronic toxoplasmosis. Front Cell Infect Microbiol 2023; 13:1129451. [PMID: 36968101 PMCID: PMC10031036 DOI: 10.3389/fcimb.2023.1129451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/14/2023] [Indexed: 03/29/2023] Open
Abstract
Toxoplasma gondii infects approximately one-third of the world's population resulting in a chronic infection with the parasite located in cysts in neurons in the brain. In most immunocompetent hosts the chronic infection is asymptomatic, but several studies have found correlations between Toxoplasma seropositivity and neuropsychiatric disorders, including Schizophrenia, and some other neurological disorders. Host-parasite interactions of bradyzoites in cysts in neurons is not well understood due in part to the lack of suitable in vitro human neuronal models. The advent of stem cell technologies in which human neurons can be derived in vitro from human induced pluripotent stem cells (hiPSCs) or direct conversion of somatic cells generating induced neurons (iNs), affords the opportunity to develop in vitro human neuronal culture systems to advance the understanding of T. gondii in human neurons. Human neurons derived from hiPSCs or iNs, generate pure human neuron monolayers that express differentiated neuronal characteristics. hiPSCs also generate 3D neuronal models that better recapitulate the cytoarchitecture of the human brain. In this review, an overview of iPSC-derived neurons and iN protocols leading to 2D human neuron cultures and hiPSC-derived 3D cerebral organoids will be given. The potential applications of these 2D and 3D human neuronal models to address questions about host-parasite interactions of T. gondii in neurons and the parasite in the CNS, will be discussed. These human neuronal in vitro models hold the promise to advance the understanding of T. gondii in human neurons and to improve the understanding of neuropathogenesis of chronic toxoplasmosis.
Collapse
|
16
|
Lachman HM. Use of cerebral organoids to model environmental and gene x environment interactions in the developing fetus and neurodegenerative disorders. PHENOTYPING OF HUMAN IPSC-DERIVED NEURONS 2023:173-200. [DOI: 10.1016/b978-0-12-822277-5.00006-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
17
|
Priyathilaka TT, Laaker CJ, Herbath M, Fabry Z, Sandor M. Modeling infectious diseases of the central nervous system with human brain organoids. Transl Res 2022; 250:18-35. [PMID: 35811019 PMCID: PMC11185418 DOI: 10.1016/j.trsl.2022.06.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 10/17/2022]
Abstract
Bacteria, fungi, viruses, and protozoa are known to infect and induce diseases in the human central nervous system (CNS). Modeling the mechanisms of interaction between pathogens and the CNS microenvironment is essential to understand their pathophysiology and develop new treatments. Recent advancements in stem cell technologies have allowed for the creation of human brain organoids, which more closely resembles the human CNS microenvironment when compared to classical 2-dimensional (2D) cultures. Now researchers can utilize these systems to investigate and reinvestigate questions related to CNS infection in a human-derived brain organoid system. Here in this review, we highlight several infectious diseases which have been tested in human brain organoids and compare similarities in response to these pathogens across different investigations. We also provide a brief overview of some recent advancements which can further enrich this model to develop new and better therapies to treat brain infections.
Collapse
Affiliation(s)
- Thanthrige Thiunuwan Priyathilaka
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin
| | - Collin James Laaker
- Neuroscience Training Program, University of Wisconsin Madison, Madison, Wisconsin
| | - Melinda Herbath
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin.
| |
Collapse
|
18
|
Pan M, Ge CC, Fan YM, Jin QW, Shen B, Huang SY. The determinants regulating Toxoplasma gondii bradyzoite development. Front Microbiol 2022; 13:1027073. [PMID: 36439853 PMCID: PMC9691885 DOI: 10.3389/fmicb.2022.1027073] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/24/2022] [Indexed: 11/04/2023] Open
Abstract
Toxoplasma gondii is an obligate intracellular zoonotic pathogen capable of infecting almost all cells of warm-blooded vertebrates. In intermediate hosts, this parasite reproduces asexually in two forms, the tachyzoite form during acute infection that proliferates rapidly and the bradyzoite form during chronic infection that grows slowly. Depending on the growth condition, the two forms can interconvert. The conversion of tachyzoites to bradyzoites is critical for T. gondii transmission, and the reactivation of persistent bradyzoites in intermediate hosts may lead to symptomatic toxoplasmosis. However, the mechanisms that control bradyzoite differentiation have not been well studied. Here, we review recent advances in the study of bradyzoite biology and stage conversion, aiming to highlight the determinants associated with bradyzoite development and provide insights to design better strategies for controlling toxoplasmosis.
Collapse
Affiliation(s)
- Ming Pan
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Ceng-Ceng Ge
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yi-Min Fan
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Qi-Wang Jin
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Bang Shen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Si-Yang Huang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
19
|
Tucker MS, Khan A, Jenkins MC, Dubey JP, Rosenthal BM. Hastening Progress in Cyclospora Requires Studying Eimeria Surrogates. Microorganisms 2022; 10:1977. [PMID: 36296256 PMCID: PMC9608778 DOI: 10.3390/microorganisms10101977] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/16/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
Cyclospora cayetanensis is an enigmatic human parasite that sickens thousands of people worldwide. The scarcity of research material and lack of any animal model or cell culture system slows research, denying the produce industry, epidemiologists, and regulatory agencies of tools that might aid diagnosis, risk assessment, and risk abatement. Fortunately, related species offer a strong foundation when used as surrogates to study parasites of this type. Species of Eimeria lend themselves especially well as surrogates for C. cayetanensis. Those Eimeria that infect poultry can be produced in abundance, share many biological features with Cyclospora, pose no risk to the health of researchers, and can be studied in their natural hosts. Here, we overview the actual and potential uses of such surrogates to advance understanding of C. cayetanensis biology, diagnostics, control, and genomics, focusing on opportunities to improve prevention, surveillance, risk assessment, and risk reduction. Studying Eimeria surrogates accelerates progress, closing important research gaps and refining promising tools for producers and food safety regulators to monitor and ameliorate the food safety risks imposed by this emerging, enigmatic parasite.
Collapse
Affiliation(s)
| | | | | | | | - Benjamin M. Rosenthal
- Animal Parasitic Disease Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, BARC-East, Beltsville, MD 20705, USA
| |
Collapse
|
20
|
Kim H, Hong SH, Jeong HE, Han S, Ahn J, Kim JA, Yang JH, Oh HJ, Chung S, Lee SE. Microfluidic model for in vitro acute Toxoplasma gondii infection and transendothelial migration. Sci Rep 2022; 12:11449. [PMID: 35794197 PMCID: PMC9259589 DOI: 10.1038/s41598-022-15305-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/22/2022] [Indexed: 11/10/2022] Open
Abstract
The protozoan parasite Toxoplasma gondii (T. gondii) causes one of the most common human zoonotic diseases and infects approximately one-third of the global population. T. gondii infects nearly every cell type and causes severe symptoms in susceptible populations. In previous laboratory animal studies, T. gondii movement and transmission were not analyzed in real time. In a three-dimensional (3D) microfluidic assay, we successfully supported the complex lytic cycle of T. gondii in situ by generating a stable microvasculature. The physiology of the T. gondii-infected microvasculature was monitored in order to investigate the growth, paracellular and transcellular migration, and transmission of T. gondii, as well as the efficacy of T. gondii drugs.
Collapse
Affiliation(s)
- Hyunho Kim
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea.,Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, USA
| | - Sung-Hee Hong
- Division of Vectors and Parasitic Diseases, Korea Diseases Control and Prevention Agency, Cheongju, Republic of Korea
| | - Hyo Eun Jeong
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
| | | | - Jinchul Ahn
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
| | - Jin-A Kim
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
| | | | - Hyun Jeong Oh
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea.
| | - Seok Chung
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea. .,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.
| | - Sang-Eun Lee
- Division of Vectors and Parasitic Diseases, Korea Diseases Control and Prevention Agency, Cheongju, Republic of Korea.
| |
Collapse
|
21
|
Kim MB, Hwangbo S, Jang S, Jo YK. Bioengineered Co-culture of organoids to recapitulate host-microbe interactions. Mater Today Bio 2022; 16:100345. [PMID: 35847376 PMCID: PMC9283667 DOI: 10.1016/j.mtbio.2022.100345] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 11/05/2022] Open
Abstract
The recent spike in the instances of complex physiological host-microbe interactions has raised the demand for developing in vitro models that recapitulate the microbial microenvironment in the human body. Organoids are steadily emerging as an in vitro culture system that closely mimics the structural, functional, and genetic features of complex human organs, particularly for better understanding host-microbe interactions. Recent advances in organoid culture technology have become new avenues for assessing the pathogenesis of symbiotic interactions, pathogen-induced infectious diseases, and various other diseases. The co-cultures of organoids with microbes have shown great promise in simulating host-microbe interactions with a high level of complexity for further advancement in related fields. In this review, we provide an overview of bioengineering approaches for microbe-co-cultured organoids. Latest developments in the applications of microbe-co-cultured organoids to study human physiology and pathophysiology are also highlighted. Further, an outlook on future research on bioengineered organoid co-cultures for various applications is presented.
Collapse
|
22
|
Adams Y, Jensen AR. Cerebral malaria - modelling interactions at the blood-brain barrier in vitro. Dis Model Mech 2022; 15:275963. [PMID: 35815443 PMCID: PMC9302004 DOI: 10.1242/dmm.049410] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The blood–brain barrier (BBB) is a continuous endothelial barrier that is supported by pericytes and astrocytes and regulates the passage of solutes between the bloodstream and the brain. This structure is called the neurovascular unit and serves to protect the brain from blood-borne disease-causing agents and other risk factors. In the past decade, great strides have been made to investigate the neurovascular unit for delivery of chemotherapeutics and for understanding how pathogens can circumvent the barrier, leading to severe and, at times, fatal complications. One such complication is cerebral malaria, in which Plasmodium falciparum-infected red blood cells disrupt the barrier function of the BBB, causing severe brain swelling. Multiple in vitro models of the BBB are available to investigate the mechanisms underlying the pathogenesis of cerebral malaria and other diseases. These range from single-cell monolayer cultures to multicellular BBB organoids and highly complex cerebral organoids. Here, we review the technologies available in malaria research to investigate the interaction between P. falciparum-infected red blood cells and the BBB, and discuss the advantages and disadvantages of each model. Summary: This Review discusses the available in vitro models to investigate the impact of adhesion of Plasmodium falciparum-infected red blood cells on the blood–brain barrier, a process associated with cerebral malaria.
Collapse
Affiliation(s)
- Yvonne Adams
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Anja Ramstedt Jensen
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| |
Collapse
|
23
|
Ramírez-Flores CJ, Tibabuzo Perdomo AM, Gallego-López GM, Knoll LJ. Transcending Dimensions in Apicomplexan Research: from Two-Dimensional to Three-Dimensional In Vitro Cultures. Microbiol Mol Biol Rev 2022; 86:e0002522. [PMID: 35412359 PMCID: PMC9199416 DOI: 10.1128/mmbr.00025-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Parasites belonging to the Apicomplexa phylum are among the most successful pathogens known in nature. They can infect a wide range of hosts, often remain undetected by the immune system, and cause acute and chronic illness. In this phylum, we can find parasites of human and veterinary health relevance, such as Toxoplasma, Plasmodium, Cryptosporidium, and Eimeria. There are still many unknowns about the biology of these pathogens due to the ethical and practical issues of performing research in their natural hosts. Animal models are often difficult or nonexistent, and as a result, there are apicomplexan life cycle stages that have not been studied. One recent alternative has been the use of three-dimensional (3D) systems such as organoids, 3D scaffolds with different matrices, microfluidic devices, organs-on-a-chip, and other tissue culture models. These 3D systems have facilitated and expanded the research of apicomplexans, allowing us to explore life stages that were previously out of reach and experimental procedures that were practically impossible to perform in animal models. Human- and animal-derived 3D systems can be obtained from different organs, allowing us to model host-pathogen interactions for diagnostic methods and vaccine development, drug testing, exploratory biology, and other applications. In this review, we summarize the most recent advances in the use of 3D systems applied to apicomplexans. We show the wide array of strategies that have been successfully used so far and apply them to explore other organisms that have been less studied.
Collapse
Affiliation(s)
- Carlos J. Ramírez-Flores
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Andrés M. Tibabuzo Perdomo
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Gina M. Gallego-López
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Laura J. Knoll
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
| |
Collapse
|
24
|
Humayun M, Ayuso JM, Park KY, Martorelli Di Genova B, Skala MC, Kerr SC, Knoll LJ, Beebe DJ. Innate immune cell response to host-parasite interaction in a human intestinal tissue microphysiological system. SCIENCE ADVANCES 2022; 8:eabm8012. [PMID: 35544643 PMCID: PMC9075809 DOI: 10.1126/sciadv.abm8012] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/23/2022] [Indexed: 05/03/2023]
Abstract
Protozoan parasites that infect humans are widespread and lead to varied clinical manifestations, including life-threatening illnesses in immunocompromised individuals. Animal models have provided insight into innate immunity against parasitic infections; however, species-specific differences and complexity of innate immune responses make translation to humans challenging. Thus, there is a need for in vitro systems that can elucidate mechanisms of immune control and parasite dissemination. We have developed a human microphysiological system of intestinal tissue to evaluate parasite-immune-specific interactions during infection, which integrates primary intestinal epithelial cells and immune cells to investigate the role of innate immune cells during epithelial infection by the protozoan parasite, Toxoplasma gondii, which affects billions of people worldwide. Our data indicate that epithelial infection by parasites stimulates a broad range of effector functions in neutrophils and natural killer cell-mediated cytokine production that play immunomodulatory roles, demonstrating the potential of our system for advancing the study of human-parasite interactions.
Collapse
Affiliation(s)
- Mouhita Humayun
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Jose M. Ayuso
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI, USA
- Department of Dermatology, University of Wisconsin-Madison, Madison, WI, USA
| | - Keon Young Park
- Department of Surgery, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Melissa C. Skala
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Sheena C. Kerr
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Laura J. Knoll
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - David J. Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
25
|
Bhattacharya A, Choi WWY, Muffat J, Li Y. Modeling Developmental Brain Diseases Using Human Pluripotent Stem Cells-Derived Brain Organoids - Progress and Perspective. J Mol Biol 2021; 434:167386. [PMID: 34883115 DOI: 10.1016/j.jmb.2021.167386] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023]
Abstract
Developmental brain diseases encompass a group of conditions resulting from genetic or environmental perturbations during early development. Despite the increased research attention in recent years following recognition of the prevalence of these diseases, there is still a significant lack of knowledge of their etiology and treatment options. The genetic and clinical heterogeneity of these diseases, in addition to the limitations of experimental animal models, contribute to this difficulty. In this regard, the advent of brain organoid technology has provided a new means to study the cause and progression of developmental brain diseases in vitro. Derived from human pluripotent stem cells, brain organoids have been shown to recapitulate key developmental milestones of the early human brain. Combined with technological advancements in genome editing, tissue engineering, electrophysiology, and multi-omics analysis, brain organoids have expanded the frontiers of human neurobiology, providing valuable insight into the cellular and molecular mechanisms of normal and pathological brain development. This review will summarize the current progress of applying brain organoids to model human developmental brain diseases and discuss the challenges that need to be overcome to further advance their utility.
Collapse
Affiliation(s)
- Afrin Bhattacharya
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Wendy W Y Choi
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Julien Muffat
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Program in Neurosciences and Mental Health, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Yun Li
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
26
|
Long RKM, Piatti L, Korbmacher F, Bernabeu M. Understanding parasite-brain microvascular interactions with engineered 3D blood-brain barrier models. Mol Microbiol 2021; 117:693-704. [PMID: 34837419 DOI: 10.1111/mmi.14852] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 11/23/2021] [Indexed: 01/25/2023]
Abstract
Microbial interactions with the blood-brain barrier (BBB) can be highly pathogenic and are still not well understood. Among these, parasites present complex interactions with the brain microvasculature that are difficult to decipher using experimental animal models or reductionist 2D in vitro cultures. Novel 3D engineered blood-brain barrier models hold great promise to overcome limitations in traditional research approaches. These models better mimic the intricate 3D architecture of the brain microvasculature and recapitulate several aspects of BBB properties, physiology, and function. Moreover, they provide improved control over biophysical and biochemical experimental parameters and are compatible with advanced imaging and molecular biology techniques. Here, we review design considerations and methodologies utilized to successfully engineer BBB microvessels. Finally, we highlight the advantages and limitations of existing engineered models and propose applications to study parasite interactions with the BBB, including mechanisms of barrier disruption.
Collapse
Affiliation(s)
- Rory K M Long
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
| | - Livia Piatti
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
| | | | - Maria Bernabeu
- European Molecular Biology Laboratory (EMBL) Barcelona, Barcelona, Spain
| |
Collapse
|
27
|
Affiliation(s)
| | - Laura J. Knoll
- University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
28
|
Organoids in modelling infectious diseases. Drug Discov Today 2021; 27:223-233. [PMID: 34418577 DOI: 10.1016/j.drudis.2021.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 05/14/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022]
Abstract
Approaches based on animal and two-dimensional (2D) cell culture models cannot ensure reliable results in modeling novel pathogens or in drug testing in the short term; therefore, there is rising interest in platforms such as organoids. To develop a toolbox that can be used successfully to overcome current issues in modeling various infections, it is essential to provide a framework of recent achievements in applying organoids. Organoids have been used to study viruses, bacteria, and protists that cause, for example, respiratory, gastrointestinal, and liver diseases. Their future as models of infection will be associated with improvements in system complexity, including abilities to model tissue structure, a dynamic microenvironment, and coinfection. Teaser. Organoids are a flexible tool for modelling viral, bacterial and protist infections. They can provide fast and reliable information on the biology of pathogens and in drug screening, and thus have become essential in combatting emerging infectious diseases.
Collapse
|
29
|
Kulkeaw K, Pengsart W. Progress and Challenges in the Use of a Liver-on-a-Chip for Hepatotropic Infectious Diseases. MICROMACHINES 2021; 12:mi12070842. [PMID: 34357252 PMCID: PMC8306537 DOI: 10.3390/mi12070842] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/13/2021] [Accepted: 07/17/2021] [Indexed: 12/16/2022]
Abstract
The liver is a target organ of life-threatening pathogens and prominently contributes to the variation in drug responses and drug-induced liver injury among patients. Currently available drugs significantly decrease the morbidity and mortality of liver-dwelling pathogens worldwide; however, emerging clinical evidence reveals the importance of host factors in the design of safe and effective therapies for individuals, known as personalized medicine. Given the primary adherence of cells in conventional two-dimensional culture, the use of these one-size-fit-to-all models in preclinical drug development can lead to substantial failures in assessing therapeutic safety and efficacy. Advances in stem cell biology, bioengineering and material sciences allow us to develop a more physiologically relevant model that is capable of recapitulating the human liver. This report reviews the current use of liver-on-a-chip models of hepatotropic infectious diseases in the context of precision medicine including hepatitis virus and malaria parasites, assesses patient-specific responses to antiviral drugs, and designs personalized therapeutic treatments to address the need for a personalized liver-like model. Second, most organs-on-chips lack a monitoring system for cell functions in real time; thus, the review discusses recent advances and challenges in combining liver-on-a-chip technology with biosensors for assessing hepatocyte viability and functions. Prospectively, the biosensor-integrated liver-on-a-chip device would provide novel biological insights that could accelerate the development of novel therapeutic compounds.
Collapse
Affiliation(s)
- Kasem Kulkeaw
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Correspondence: ; Tel.: +66-2-419-6468 (ext. 96484)
| | - Worakamol Pengsart
- Faculty of Graduate Studies, Mahidol University, Nakhon Pathom 73170, Thailand;
| |
Collapse
|
30
|
Advances in modelling the human microbiome-gut-brain axis in vitro. Biochem Soc Trans 2021; 49:187-201. [PMID: 33544117 PMCID: PMC7924999 DOI: 10.1042/bst20200338] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/23/2020] [Accepted: 01/15/2021] [Indexed: 02/06/2023]
Abstract
The human gut microbiome has emerged as a key player in the bidirectional communication of the gut–brain axis, affecting various aspects of homeostasis and pathophysiology. Until recently, the majority of studies that seek to explore the mechanisms underlying the microbiome–gut–brain axis cross-talk, relied almost exclusively on animal models, and particularly gnotobiotic mice. Despite the great progress made with these models, various limitations, including ethical considerations and interspecies differences that limit the translatability of data to human systems, pushed researchers to seek for alternatives. Over the past decades, the field of in vitro modelling of tissues has experienced tremendous growth, thanks to advances in 3D cell biology, materials, science and bioengineering, pushing further the borders of our ability to more faithfully emulate the in vivo situation. The discovery of stem cells has offered a new source of cells, while their use in generating gastrointestinal and brain organoids, among other tissues, has enabled the development of novel 3D tissues that better mimic the native tissue structure and function, compared with traditional assays. In parallel, organs-on-chips technology and bioengineered tissues have emerged as highly promising alternatives to animal models for a wide range of applications. Here, we discuss how recent advances and trends in this area can be applied in host–microbe and host–pathogen interaction studies. In addition, we highlight paradigm shifts in engineering more robust human microbiome-gut-brain axis models and their potential to expand our understanding of this complex system and hence explore novel, microbiome-based therapeutic approaches.
Collapse
|
31
|
Pance A. The Stem Cell Revolution Revealing Protozoan Parasites' Secrets and Paving the Way towards Vaccine Development. Vaccines (Basel) 2021; 9:105. [PMID: 33572549 PMCID: PMC7911700 DOI: 10.3390/vaccines9020105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
Protozoan infections are leading causes of morbidity and mortality in humans and some of the most important neglected diseases in the world. Despite relentless efforts devoted to vaccine and drug development, adequate tools to treat and prevent most of these diseases are still lacking. One of the greatest hurdles is the lack of understanding of host-parasite interactions. This gap in our knowledge comes from the fact that these parasites have complex life cycles, during which they infect a variety of specific cell types that are difficult to access or model in vitro. Even in those cases when host cells are readily available, these are generally terminally differentiated and difficult or impossible to manipulate genetically, which prevents assessing the role of human factors in these diseases. The advent of stem cell technology has opened exciting new possibilities to advance our knowledge in this field. The capacity to culture Embryonic Stem Cells, derive Induced Pluripotent Stem Cells from people and the development of protocols for differentiation into an ever-increasing variety of cell types and organoids, together with advances in genome editing, represent a huge resource to finally crack the mysteries protozoan parasites hold and unveil novel targets for prevention and treatment.
Collapse
Affiliation(s)
- Alena Pance
- The Wellcome Sanger Institute, Genome Campus, Hinxton Cambridgeshire CB10 1SA, UK
| |
Collapse
|
32
|
Willner MJ, Xiao Y, Kim HS, Chen X, Xu B, Leong KW. Modeling SARS-CoV-2 infection in individuals with opioid use disorder with brain organoids. J Tissue Eng 2021; 12:2041731420985299. [PMID: 33738089 PMCID: PMC7934045 DOI: 10.1177/2041731420985299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/13/2020] [Indexed: 01/10/2023] Open
Abstract
The COVID-19 pandemic has aggravated a preexisting epidemic: the opioid crisis. Much literature has shown that the circumstances imposed by COVID-19, such as social distancing regulations, medical and financial instability, and increased mental health issues, have been detrimental to those with opioid use disorder (OUD). In addition, unexpected neurological sequelae in COVID-19 patients suggest that COVID-19 compromises neuroimmunity, induces hypoxia, and causes respiratory depression, provoking similar effects as those caused by opioid exposure. Combined conditions of COVID-19 and OUD could lead to exacerbated complications. With limited human in vivo options to study these complications, we suggest that iPSC-derived brain organoid models may serve as a useful platform to investigate the physiological connection between COVID-19 and OUD. This mini-review highlights the advances of brain organoids in other neuropsychiatric and infectious diseases and suggests their potential utility for investigating OUD and COVID-19, respectively.
Collapse
Affiliation(s)
- Moshe J Willner
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Hye Sung Kim
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, Republic of Korea
| | - Xuejing Chen
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Physics, Tsinghua University, Beijing, China
| | - Bin Xu
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|