1
|
Israeli Dangoor S, Khoury R, Salomon K, Pozzi S, Shahar S, Miari A, Leichtmann-Bardoogo Y, Bar-Hai N, Frommer N, Yeini E, Winkler T, Balint Lahat N, Kamer I, Hadad O, Laue K, Brem H, Hyde TM, Bar J, Barshack I, Ben-David U, Ishay-Ronen D, Maoz BM, Satchi-Fainaro R. CCL2 blockade combined with PD-1/P-selectin immunomodulators impedes breast cancer brain metastasis. Brain 2025; 148:1740-1756. [PMID: 39450648 PMCID: PMC12073999 DOI: 10.1093/brain/awae347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 09/11/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Over the last two decades, the diagnosis and treatment of breast cancer patients have improved considerably. However, brain metastases remain a major clinical challenge and a leading cause of mortality. Thus, a better understanding of the pathways involved in the metastatic cascade is essential. To this end, we have investigated the reciprocal effects of astrocytes and breast cancer cells, employing traditional 2D cell culture and our unique 3D multicellular tumouroid models. Our findings revealed that astrocytes enhance the proliferation, migration and invasion of breast cancer cells, suggesting a supportive role for astrocytes in breast cancer outgrowth to the brain. Elucidating the key players in astrocyte-breast cancer cells crosstalk, we found that CCL2 is highly expressed in breast cancer brain metastases tissue sections from both patients and mice. Our in vitro and in vivo models further confirmed that CCL2 has a functional role in brain metastasis. Given their aggressive nature, we sought additional immune checkpoints for rationale combination therapy. Among the promising candidates were the adhesion molecule P-selectin, which we have recently shown to play a key role in the crosstalk with microglia cells and the co-inhibitory receptor PD-1, the main target of currently approved immunotherapies. Finally, combining CCL2 inhibition with immunomodulators targeting either PD-1/PD-L1 or P-selectin/P-Selectin Ligand-1 axes in our human 3D tumouroid models and in vivo presented more favourable outcomes than each monotherapy. Taken together, we propose that CCL2-CCR2/CCR4 is a key pathway promoting breast cancer brain metastases and a promising target for an immunotherapeutic combination approach.
Collapse
Affiliation(s)
- Sahar Israeli Dangoor
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Rami Khoury
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Koren Salomon
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shir Shahar
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Adan Miari
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | | | - Neta Bar-Hai
- Cancer Research Center, Oncology Institute, Sheba Medical Center, Tel-Hashomer 5262000, Israel
- Affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Neta Frommer
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Eilam Yeini
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tom Winkler
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Nora Balint Lahat
- Department of Pathology, Sheba Medical Center, Tel Hashomer 5262000, Israel
| | - Iris Kamer
- Cancer Research Center, Oncology Institute, Sheba Medical Center, Tel-Hashomer 5262000, Israel
| | - Ori Hadad
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Kathrin Laue
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21218, USA
- Department of Psychiatry & Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Jair Bar
- Cancer Research Center, Oncology Institute, Sheba Medical Center, Tel-Hashomer 5262000, Israel
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Uri Ben-David
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dana Ishay-Ronen
- Cancer Research Center, Oncology Institute, Sheba Medical Center, Tel-Hashomer 5262000, Israel
- Affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ben M Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol Center for Regenerative Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
2
|
Oh JM, Park Y, Lee J, Shen K. Microfabricated Organ-Specific Models of Tumor Microenvironments. Annu Rev Biomed Eng 2025; 27:307-333. [PMID: 40310890 DOI: 10.1146/annurev-bioeng-110222-103522] [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: 05/03/2025]
Abstract
Despite the advances in detection, diagnosis, and treatments, cancer remains a lethal disease, claiming the lives of more than 600,000 people in the United States alone in 2024. To accelerate the development of new therapeutic strategies with improved responses, significant efforts have been made to develop microfabricated in vitro models of tumor microenvironments (TMEs) that address the limitations of animal-based cancer models. These models incorporate several advanced tissue engineering techniques to better reflect the organ- and patient-specific TMEs. Additionally, microfabricated models integrated with next-generation single-cell omics technologies provide unprecedented insights into patient's cellular and molecular heterogeneity and complexity. This review provides an overview of the recent understanding of cancer development and outlines the key TME elements that can be captured in microfabricated models to enhance their physiological relevance. We highlight the recent advances in microfabricated cancer models that reflect the unique characteristics of their organs of origin or sites of dissemination.
Collapse
Affiliation(s)
- Jeong Min Oh
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
| | - Yongkuk Park
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - Keyue Shen
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
3
|
Liu L, He Y, Du H, Tang M, Wang T, Tan J, Zha L, Yang L, Ashrafizadeh M, Tian Y, Zhou H. Biological profile of breast cancer brain metastasis. Acta Neuropathol Commun 2025; 13:78. [PMID: 40253355 PMCID: PMC12008903 DOI: 10.1186/s40478-025-01983-4] [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: 07/29/2024] [Accepted: 03/08/2025] [Indexed: 04/21/2025] Open
Abstract
Breast cancer is one of the leading causes of death worldwide. The aggressive behaviour of breast tumor results from their metastasis. Notably, the brain tissue is one of the common regions of metastasis, thereby reducing the overall survival of patients. Moreover, the metastatic tumors demonstrate poor response or resistance to therapies. In addition, breast cancer brain metastasis provides the poor prognosis of patients. Therefore, it is of importance to understand the mechanisms in breast cancer brain metastasis. Both cell lines and animal models have been developed for the evaluation of breast cancer brain metastasis. Moreover, different tumor microenvironment components and other factors such as lymphocytes and astrocytes can affect brain metastasis. The breast cancer cells can disrupt the blood-brain barrier (BBB) during their metastasis into brain, developing blood-tumor barrier to enhance carcinogenesis. The breast cancer brain metastasis can be increased by the dysregulation of chemokines, STAT3, Wnt, Notch and PI3K/Akt. On the other hand, the effective therapeutics have been developed for the brain metastasis such as introduction of nanoparticles. Moreover, the disruption of BBB by ultrasound can increase the entrance of bioactive compounds to the brain tissue. In order to improve specificity and selectivity, the nanoparticles for the delivery of therapeutics and crossing over BBB have been developed to suppress breast cancer brain metastasis.
Collapse
Affiliation(s)
- Li Liu
- Department of Oncology, Suining Central Hospital, Suning, 629000, China
| | - Yuan He
- Department of Oncology, Yunyang County People's Hospital, Chongqing, 404500, China
| | - Hongyu Du
- Department of General Medicine, The Seventh People's Hospital of Chongqing, The Central Hospital Affiliated to Chongging University of Technology, Chongqing, 400054, China
| | - Min Tang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401120, China
| | - Tingting Wang
- Department of Gynecology and Obstetrics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jieren Tan
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, PR China
| | - Lisha Zha
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, PR China
| | - Li Yang
- Department of Nephrology, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, Guangdong Province, 510515, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250000, China.
| | - Yu Tian
- School of Public Health, Benedictine University, No.5700 College Road, Lisle, IL, 60532, USA.
- Research Center, the Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, Guangdong, China.
| | - Hui Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19 Nonglinxia Road, Guangzhou, 510080, China.
| |
Collapse
|
4
|
Saliba J, Saliba J, El-Sabban M, Mhanna R. A Biomimetic Human Multi-Cellular In Vitro Model of the Blood-Brain Barrier. Int J Mol Sci 2025; 26:3592. [PMID: 40332140 PMCID: PMC12027270 DOI: 10.3390/ijms26083592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/06/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Current in vitro models fail to recapitulate specific physiological properties of the human blood-brain barrier (BBB); hence the need for a reliable platform to study central nervous system diseases and drug permeability. To mimic the normally tight blood-brain interface, primary human endothelial cells (HAECs) and primary human astrocytes (A) were grown in a confined space of the physical scaffold created by gelatin methacrylate (GelMA) hydrogel to allow optimal astrocyte-endothelial cell direct/indirect interaction. Evidence for a physiologically relevant BBB was established by assessing the expression of tight junction markers conferring the barrier function, and by measuring biophysical attributes using the trans-endothelial electrical resistance (TEER) and the Evans blue albumin (EBA) permeability assay. An HAEC+A three-dimensional (3D) co-culture was associated with 12-fold higher claudin-5 (CLDN5) and cadherin-1 (CDH1 or Epithelial [E]-cadherin) transcriptional levels than two-dimensional (2D) models. This model conferred the highest TEER (45 Ω·cm2) in 3D HAEC+A, which value was 30 Ω·cm2 in 2D (p < 0.01) and 25 Ω·cm2 in 3D HAEC cultures (p < 0.001). Functionally, in 3D HAEC+A co-cultures, higher TEER resulted in 10-fold and 7-fold lower EBA permeability at 120 min, in HAECs alone or in to 2D co-cultures (p < 0.01). The established human primary cell model has acquired features mimicking the human BBB in vitro, and is now poised to be tested for the permeability of the BBB to pharmacological agents, parasites, cells (such as brain-tropic cancer cell metastasis) and any mechanisms that might involve traversing the BBB.
Collapse
Affiliation(s)
- John Saliba
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon;
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| | - Jessica Saliba
- Department of Public Health, Faculty of Health Sciences, University of Balamand, Beirut 1100, Lebanon;
- Department of Biology, Faculty of Sciences, Lebanese University, Beirut 1533, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon;
| | - Rami Mhanna
- Biomedical Engineering Program, Maroun Semaan Faculty of Engineering and Architecture, American University of Beirut, Beirut 1107 2020, Lebanon
| |
Collapse
|
5
|
Romero-Ben E, Goswami U, Soto-Cruz J, Mansoori-Kermani A, Mishra D, Martin-Saldaña S, Muñoz-Ugartemendia J, Sosnik A, Calderón M, Beloqui A, Larrañaga A. Polymer-based nanocarriers to transport therapeutic biomacromolecules across the blood-brain barrier. Acta Biomater 2025; 196:17-49. [PMID: 40032217 DOI: 10.1016/j.actbio.2025.02.065] [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: 09/16/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Therapeutic biomacromolecules such as genetic material, antibodies, growth factors and enzymes represent a novel therapeutic alternative for neurological diseases and disorders. In comparison to traditional therapeutics, which are mainly based on small molecular weight drugs that address the symptoms of these disorders, therapeutic biomacromolecules can reduce undesired side effects and target specific pathological pathways, thus paving the way towards personalized medicine. However, these biomacromolecules undergo degradation/denaturation processes in the physiological environment and show poor capacity to cross the blood-brain barrier (BBB). Consequently, they rarely reach the central nervous system (CNS) in their active form. Herein, we critically overview several polymeric nanocarriers that can protect and deliver therapeutic biomacromolecules across the BBB. Polymeric nanocarriers are first categorized based on their architecture (biodegradable solid nanoparticles, nanogels, dendrimers, self-assembled nanoparticles) that ultimately determines their physico-chemical properties and function. The available polymeric formulations are then thoroughly analyzed, placing particular attention on those strategies that ensure the stability of the biomacromolecules during their encapsulation process and promote their passage across the BBB by controlling their physical (e.g., mechanical properties, size, surface charge) and chemical (e.g., surface functional groups, targeting motifs) properties. Accordingly, this review gives a unique perspective on polymeric nanocarriers for the delivery of therapeutic biomacromolecules across the BBB, representing a concise, complete and easy-to-follow guide, which will be of high interest for chemists, material scientists, pharmacologists, and biologists. Besides, it also provides a critical perspective about the limited clinical translation of these systems. STATEMENT OF SIGNIFICANCE: The increasing incidence of central nervous system disorders is a major health concern. The use of therapeutic biomacromolecules has been placed in the spotlight of many investigations. However, reaching therapeutic concentration levels of biomacromolecules in the central nervous system is restricted by the blood-brain barrier and, thus, this represents the main clinical challenge when developing efficient therapies. Herein, we provide a critical discussion about the use of polymeric nanocarriers to deliver therapeutic biomacromolecules into the central nervous system, highlighting potential future directions to overcome the current challenges.
Collapse
Affiliation(s)
- Elena Romero-Ben
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain
| | - Upashi Goswami
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain; Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, Bilbao 48013, Spain
| | - Jackeline Soto-Cruz
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain
| | - Amirreza Mansoori-Kermani
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, Bilbao 48013, Spain; Istituto Italiano di Tecnologia, Smart Bio-Interfaces, Viale Rinaldo Piaggio 34, Pontedera 56025, Italy; Scuola Superiore Sant'Anna, The Biorobotics Institute, Viale Rinaldo PIaggio 34, Pontedera 56025, Italy
| | - Dhiraj Mishra
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain; Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Sergio Martin-Saldaña
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain
| | - Jone Muñoz-Ugartemendia
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, Bilbao 48013, Spain
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Marcelo Calderón
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain; IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, Bilbao 48009, Spain
| | - Ana Beloqui
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, Donostia-San Sebastián 20018, Spain; IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, Bilbao 48009, Spain
| | - Aitor Larrañaga
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, Bilbao 48013, Spain.
| |
Collapse
|
6
|
Soliman Y, Al-Khodor J, Yildirim Köken G, Mustafaoglu N. A guide for blood-brain barrier models. FEBS Lett 2025; 599:599-644. [PMID: 39533665 DOI: 10.1002/1873-3468.15053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Understanding the intricate mechanisms underlying brain-related diseases hinges on unraveling the pivotal role of the blood-brain barrier (BBB), an essential dynamic interface crucial for maintaining brain equilibrium. This review offers a comprehensive analysis of BBB physiology, delving into its cellular and molecular components while exploring a wide range of in vivo and in vitro BBB models. Notably, recent advancements in 3D cell culture techniques are explicitly discussed, as they have significantly improved the fidelity of BBB modeling by enabling the replication of physiologically relevant environments under flow conditions. Special attention is given to the cellular aspects of in vitro BBB models, alongside discussions on advances in stem cell technologies, providing valuable insights into generating robust cellular systems for BBB modeling. The diverse array of cell types used in BBB modeling, depending on their sources, is meticulously examined in this comprehensive review, scrutinizing their respective derivation protocols and implications. By synthesizing diverse approaches, this review sheds light on the improvements of BBB models to capture physiological conditions, aiding in understanding BBB interactions in health and disease conditions to foster clinical developments.
Collapse
Affiliation(s)
- Yomna Soliman
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Faculty of Pharmacy, Mansoura University, Egypt
| | - Jana Al-Khodor
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
| | | | - Nur Mustafaoglu
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul, Turkey
- Sabancı University Nanotechnology Research and Application Center, Istanbul, Turkey
| |
Collapse
|
7
|
Wang K, Chen X. Protective effect of flavonoids on oxidative stress injury in Alzheimer's disease. Nat Prod Res 2025; 39:1272-1299. [PMID: 38910339 DOI: 10.1080/14786419.2024.2345760] [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: 01/13/2024] [Revised: 03/28/2024] [Accepted: 04/14/2024] [Indexed: 06/25/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, which is mainly caused by the damage of the structure and function of the central nervous system. At present, there are many adverse reactions in market-available drugs, which can't significantly inhibit the occurrence of AD. Therefore, the current focus of research is to find safe and effective therapeutic drugs to improve the clinical treatment of AD. Oxidative stress bridges different mechanism hypotheses of AD and plays a key role in AD. Numerous studies have shown that natural flavonoids have good antioxidant effects. They can directly or indirectly resist -oxidative stress, inhibit Aβ aggregation and Tau protein hyperphosphorylation by activating Nrf2 and other oxidation-antioxidation-related signals, regulating synaptic function-related pathways, promoting mitochondrial autophagy, etc., and play a neuroprotective role in AD. In this review, we summarised the mechanism of flavonoids inhibiting oxidative stress injury in AD in recent years. Moreover, because of the shortcomings of poor biofilm permeability and low bioavailability of flavonoids, the advantages and recent research progress of nano-drug delivery systems such as liposomes and solid lipid nanoparticles were highlighted. We hope this review provides a useful way to explore safe and effective AD treatments.
Collapse
Affiliation(s)
- Kaixuan Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xinmei Chen
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
8
|
Mokarram N, Case A, Hossainy NN, Lyon JG, MacDonald TJ, Bellamkonda R. Device-assisted strategies for drug delivery across the blood-brain barrier to treat glioblastoma. COMMUNICATIONS MATERIALS 2025; 6:5. [PMID: 39790893 PMCID: PMC11706785 DOI: 10.1038/s43246-024-00721-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025]
Abstract
The blood-brain barrier, essential for protecting the central nervous system, also restricts drug delivery to this region. Thus, delivering drugs across the blood-brain barrier is an active research area in immunology, oncology, and neurology; moreover, novel methods are urgently needed to expand therapeutic options for central nervous system pathologies. While previous strategies have focused on small molecules that modulate blood-brain barrier permeability or penetrate the barrier, there is an increased focus on biomedical devices-external or implanted-for improving drug delivery. Here, we review device-assisted drug delivery across the blood-brain barrier, emphasizing its application in glioblastoma, an aggressively malignant primary brain cancer in which the blood-brain barrier plays a central role. We examine the blood-brain barrier and its features in glioblastoma, emerging models for studying the blood-brain barrier, and device-assisted methods for crossing the blood-brain barrier. We conclude by presenting methods to monitor the blood-brain barrier and paradigms for combined cross-BBB drug delivery.
Collapse
Affiliation(s)
- Nassir Mokarram
- Department of Neurosurgery, Emory University, Atlanta, GA USA
| | - Ayden Case
- Trinity College of Arts and Sciences, Duke University, Durham, NC USA
| | | | - Johnathan G. Lyon
- Department of Biomedical Engineering, Duke University, Durham, NC USA
| | - Tobey J. MacDonald
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA USA
| | | |
Collapse
|
9
|
Chong JK, Jain P, Prasad S, Dubey NK, Saxena S, Lo WC. Optimizing Glioblastoma, IDH-wildtype Treatment Outcomes : A Radiomics and Support Vector Machine-Based Approach to Overall Survival Estimation. J Korean Neurosurg Soc 2025; 68:7-18. [PMID: 39099033 PMCID: PMC11725457 DOI: 10.3340/jkns.2024.0100] [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: 05/18/2024] [Revised: 06/20/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024] Open
Abstract
OBJECTIVE Glioblastoma multiforme (GBM), particularly the isocitrate dehydrogenase (IDH)-wildtype type, represents a significant clinical challenge due to its aggressive nature and poor prognosis. Despite advancements in medical imaging and its modalities, survival rates have not improved significantly, demanding innovative treatment planning and outcome prediction approaches. METHODS This study utilizes a support vector machine (SVM) classifier using radiomics features to predict the overall survival (OS) of GBM, IDH-wildtype patients to short (<12 months) and long (≥12 months) survivors. A dataset comprising multi-parametric magnetic resonance imaging scans from 574 patients was analyzed. Radiomic features were extracted from T1, T2, fluid-attenuated inversion recovery, and T1 with gadolinium (T1GD) sequences. Low variance features were removed, and recursive feature elimination was used to select the most informative features. The SVM model was trained using a k-fold cross-validation approach. Furthermore, clinical parameters such as age, gender, and MGMT (O6-methylguanine-DNA methyltransferase) promoter methylation status were integrated to enhance prediction accuracy. RESULTS The model showed reasonable results in terms of cross-validated area under the curve of 0.84 (95% confidence interval, 0.80-0.90) with (p<0.001) effectively categorizing patients into short and long survivors. Log-rank test (chi-square statistics) analysis for the developed model was 0.00029 along with the 1.20 Cohen's d effect size. Most importantly, clinical data integration further refined the survival estimates, providing a more fitted prediction that considers individual patient characteristics by Kaplan-Meier curve with p-value <0.0001. CONCLUSION The proposed method significantly enhances the predictive accuracy of OS outcomes in GBM, IDH-wildtype patients. By integrating detailed imaging features with key clinical indicators, this model offers a robust tool for personalized treatment planning, potentially improving OS.
Collapse
Affiliation(s)
- Jiunn-Kai Chong
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Priyanka Jain
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, India
| | - Shivani Prasad
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, India
| | - Navneet Kumar Dubey
- Executive Programme in Healthcare Management, Indian Institute of Management, Lucknow, India
| | - Sanjay Saxena
- Department of Computer Science & Engineering, International Institute of Information Technology, Bhubaneswar, India
| | - Wen-Cheng Lo
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
- Division of Neurosurgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Neuroscience Institute, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
10
|
Walia A, Kaur A, Singh R, Rani N, Swami R. Unveiling the Mysteries of the Blood-brain Barrier: The Problem of the Brain/spinal Pharmacotherapy. Cent Nerv Syst Agents Med Chem 2025; 25:91-108. [PMID: 39206486 DOI: 10.2174/0118715249297247240813104929] [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: 01/11/2024] [Revised: 06/03/2024] [Accepted: 06/20/2024] [Indexed: 09/04/2024]
Abstract
The most critical issue impeding the development of innovative cerebrospinal medications is the blood-brain barrier (BBB). The BBB limits the ability of most medications to penetrate the brain to the CNS. The BBB structure and functions are summarized, with the physical barrier generated by endothelial tight junctions and the transport barrier formed by transporters within the membrane and vesicular processes. The functions of connected cells, particularly the end feet of astrocytic glial cells, microglia, and pericytes, are described. The drugs that cross the blood brain barrier are explained below along with their mechanisms. Some of the associated conditions and problems are given.
Collapse
Affiliation(s)
- Aditya Walia
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Amandeep Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Randhir Singh
- Central University of Punjab, Bathinda, Punjab, India
| | - Nidhi Rani
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Rajan Swami
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
11
|
Zimina TM, Sitkov NO, Gareev KG, Mikhailova NV, Combs SE, Shevtsov MA. Hybrid-integrated devices for mimicking malignant brain tumors ("tumor-on-a-chip") for in vitro development of targeted drug delivery and personalized therapy approaches. Front Med (Lausanne) 2024; 11:1452298. [PMID: 39629230 PMCID: PMC11611596 DOI: 10.3389/fmed.2024.1452298] [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: 06/20/2024] [Accepted: 10/14/2024] [Indexed: 12/07/2024] Open
Abstract
Acute and requiring attention problem of oncotheranostics is a necessity for the urgent development of operative and precise diagnostics methods, followed by efficient therapy, to significantly reduce disability and mortality of citizens. A perspective way to achieve efficient personalized treatment is to use methods for operative evaluation of the individual drug load, properties of specific tumors and the effectiveness of selected therapy, and other actual features of pathology. Among the vast diversity of tumor types-brain tumors are the most invasive and malignant in humans with poor survival after diagnosis. Among brain tumors glioblastoma shows exceptionally high mortality. More studies are urgently needed to understand the risk factors and improve therapy approaches. One of the actively developing approaches is the tumor-on-a-chip (ToC) concept. This review examines the achievements of recent years in the field of ToC system developments. The basics of microfluidic chips technologies are considered in the context of their applications in solving oncological problems. Then the basic principles of tumors cultivation are considered to evaluate the main challengers in implementation of microfluidic devices, for growing cell cultures and possibilities of their treatment and observation. The main achievements in the culture types diversity approaches and their advantages are being analyzed. The modeling of angiogenesis and blood-brain barrier (BBB) on a chip, being a principally important elements of the life system, were considered in detail. The most interesting examples and achievements in the field of tumor-on-a-chip developments have been presented.
Collapse
Affiliation(s)
- Tatiana M. Zimina
- Department of Micro and Nanoelectronics, St. Petersburg Electrotechnical University “LETI” (ETU), Saint Petersburg, Russia
| | - Nikita O. Sitkov
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Kamil G. Gareev
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Natalia V. Mikhailova
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Stephanie E. Combs
- Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Maxim A. Shevtsov
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg, Russia
- Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
12
|
Stangler LA, Chang SY, Kim I, Choi J, Kouzani AZ, Bennet KE, Burns TC, Van Gompel JJ, Worrell GA, Howe CL. Defining the Spatial Resolution of Analyte Recovery during Microperfusion-Based Sampling of Brain Parenchyma. ACS Chem Neurosci 2024; 15:3220-3227. [PMID: 39155540 PMCID: PMC11378288 DOI: 10.1021/acschemneuro.4c00410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024] Open
Abstract
The unique architecture of the brain and the blood-brain barrier imposes challenges for the measurement of parenchyma-derived biomarkers that prevent sufficient understanding of transient neuropathogenic processes. One solution to this challenge is direct sampling of brain interstitial fluid via implanted microperfusion probes. Seeking to understand spatial limitations to microperfusion in the brain, we employed computational fluid dynamics modeling and empirical recovery of fluorescently labeled dextrans in an animal model. We found that dextrans were successfully recovered via microperfusion over a 6 h sampling period, especially at probes implanted 2 mm from the dextran infusion point relative to probes implanted 5 mm from the injection site. Experimental recovery was consistently around 1% of simulated, suggesting that this parameter can be used to set practical limits on the maximal tissue concentration of proteins measured in microperfusates and on the spatial domain sampled by our multimodal microperfusion probe.
Collapse
Affiliation(s)
- Luke A Stangler
- School
of Engineering, Deakin University, Geelong, Victoria 3216, Australia
- Division
of Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Su-Youne Chang
- Department
of Neurosurgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Inyong Kim
- Department
of Neurology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Jonghoon Choi
- Department
of Neurology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Abbas Z Kouzani
- School
of Engineering, Deakin University, Geelong, Victoria 3216, Australia
| | - Kevin E. Bennet
- Division
of Engineering, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Terry C Burns
- Department
of Neurosurgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Jamie J Van Gompel
- Department
of Neurosurgery, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Gregory A Worrell
- Department
of Neurology, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Charles L Howe
- Department
of Neurology, Mayo Clinic, Rochester, Minnesota 55905, United States
- Division
of Experimental Neurology, Mayo Clinic, Rochester, Minnesota 55905, United States
| |
Collapse
|
13
|
Banik M, Ledray AP, Wu Y, Lu Y. Delivering DNA Aptamers Across the Blood-Brain Barrier Reveals Heterogeneous Decreased ATP in Different Brain Regions of Alzheimer's Disease Mouse Models. ACS CENTRAL SCIENCE 2024; 10:1585-1593. [PMID: 39220690 PMCID: PMC11363336 DOI: 10.1021/acscentsci.4c00563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024]
Abstract
DNA aptamers have been developed as sensors to detect metabolites with high sensitivity, selectivity, and biocompatibility. While they are effective in sensing important targets in the brain, the lack of methods for their efficient delivery across the blood-brain barrier (BBB) has significantly hindered their applications in brain research. To address this issue, we herein report the development of brain cell-derived exosomes as endogenous BBB delivery vehicles to deliver an ATP-responsive aptamer across the BBB of live mice for noninvasive live brain imaging. We found that the system uses endosome recycling to transfer the sensors between the delivered exosomes and native recycling endosomes, resulting in high delivery efficiencies. Using this system, we observed unique signal distributions for ATP across different brain regions, with significant accumulation in the subiculum and cortex in healthy mice. In an Alzheimer's disease transgenic mouse model, ATP levels decreased in the subiculum and cortex, demonstrating this method's capability to determine metabolite location and relative abundance with high spatial resolution in vivo. Since DNA aptamers have been obtained for many other targets, the method developed in this work can be applied to deliver sensors across the BBB to image a wide range of other brain-related metabolites.
Collapse
Affiliation(s)
- Mandira Banik
- University
of Texas at Austin, Department of Chemistry, Austin, Texas 78712, United States
| | - Aaron P. Ledray
- University
of Texas at Austin, Department of Chemistry, Austin, Texas 78712, United States
| | - Yuting Wu
- University
of Texas at Austin, Department of Chemistry, Austin, Texas 78712, United States
| | - Yi Lu
- University
of Texas at Austin, Department of Chemistry, Austin, Texas 78712, United States
| |
Collapse
|
14
|
Royse MK, Fowler M, Mai AK, He Y, Durante MR, Buist N, Procopio A, Xu J, Veiseh O. Development of a 3D printed perfusable in vitro blood-brain barrier model for use as a scalable screening tool. Biomater Sci 2024; 12:4363-4375. [PMID: 39023223 DOI: 10.1039/d4bm00663a] [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: 07/20/2024]
Abstract
Despite recent technological advances in drug discovery, the success rate for neurotherapeutics remains alarmingly low compared to treatments for other areas of the body. One of the biggest challenges for delivering therapeutics to the central nervous system (CNS) is the presence of the blood-brain barrier (BBB). In vitro blood-brain barrier models with high predictability are essential to aid in designing parameters for new therapeutics, assess their ability to cross the BBB, and investigate therapeutic strategies that can be employed to enhance transport. Here, we demonstrate the development of a 3D printable hydrogel blood-brain barrier model that mimics the cellular composition and structure of the blood-brain barrier with human brain endothelial cells lining the surface, pericytes in direct contact with the endothelial cells on the abluminal side of the endothelium, and astrocytes in the surrounding printed bulk matrix. We introduce a simple, static printed hemi-cylinder model to determine design parameters such as media selection, co-culture ratios, and cell incorporation timing in a resource-conservative and high-throughput manner. Presence of cellular adhesion junction, VE-Cadherin, efflux transporters, P-glycoprotein (P-gp) and Breast cancer resistance protein (BCRP), and receptor-mediated transporters, Transferrin receptor (TfR) and low-density lipoprotein receptor-related protein 1 (LRP1) were confirmed via immunostaining demonstrating the ability of this model for screening in therapeutic strategies that rely on these transport systems. Design parameters determined in the hemi-cylinder model were translated to a more complex, perfusable vessel model to demonstrate its utility for determining barrier function and assessing permeability to model therapeutic compounds. This 3D-printed blood-brain barrier model represents one of the first uses of projection stereolithography to fabricate a perfusable blood-brain barrier model, enabling the patterning of complex vessel geometries and precise arrangement of cell populations. This model demonstrates potential as a new platform to investigate the delivery of neurotherapeutic compounds and drug delivery strategies through the blood-brain barrier, providing a useful in vitro screening tool in central nervous system drug discovery and development.
Collapse
Affiliation(s)
- Madison K Royse
- Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, USA.
| | - Martha Fowler
- Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, USA.
| | - A Kristen Mai
- Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, USA.
| | - Yufang He
- Division of Technology, Infrastructure, Operations & Experience, Merck & Co., Inc. Rahway, NJ 07065, USA
| | - Marc R Durante
- Division of Technology, Infrastructure, Operations & Experience, Merck & Co., Inc. Rahway, NJ 07065, USA
| | - Nicole Buist
- Department of Pharmaceutical Sciences & Clinical Supply, Merck & Co., Inc. Rahway, NJ 07065, USA.
| | - Adam Procopio
- Department of Pharmaceutical Sciences & Clinical Supply, Merck & Co., Inc. Rahway, NJ 07065, USA.
| | - Jun Xu
- Department of Pharmaceutical Sciences & Clinical Supply, Merck & Co., Inc. Rahway, NJ 07065, USA.
| | - Omid Veiseh
- Department of Bioengineering, Rice University, 6100 Main St., Houston, TX 77005, USA.
| |
Collapse
|
15
|
Dancy C, Heintzelman KE, Katt ME. The Glycocalyx: The Importance of Sugar Coating the Blood-Brain Barrier. Int J Mol Sci 2024; 25:8404. [PMID: 39125975 PMCID: PMC11312458 DOI: 10.3390/ijms25158404] [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: 06/28/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
The endothelial glycocalyx (GCX), located on the luminal surface of vascular endothelial cells, is composed of glycoproteins, proteoglycans, and glycosaminoglycans. It plays a pivotal role in maintaining blood-brain barrier (BBB) integrity and vascular health within the central nervous system (CNS), influencing critical processes such as blood flow regulation, inflammation modulation, and vascular permeability. While the GCX is ubiquitously expressed on the surface of every cell in the body, the GCX at the BBB is highly specialized, with a distinct composition of glycans, physical structure, and surface charge when compared to GCX elsewhere in the body. There is evidence that the GCX at the BBB is disrupted and partially shed in many diseases that impact the CNS. Despite this, the GCX has yet to be a major focus of therapeutic targeting for CNS diseases. This review examines diverse model systems used in cerebrovascular GCX-related research, emphasizing the importance of selecting appropriate models to ensure clinical relevance and translational potential. This review aims to highlight the importance of the GCX in disease and how targeting the GCX at the BBB specifically may be an effective approach for brain specific targeting for therapeutics.
Collapse
Affiliation(s)
- Candis Dancy
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA; (C.D.); (K.E.H.)
| | - Kaitlyn E. Heintzelman
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA; (C.D.); (K.E.H.)
- School of Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Moriah E. Katt
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA; (C.D.); (K.E.H.)
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA
| |
Collapse
|
16
|
Nahon DM, Moerkens R, Aydogmus H, Lendemeijer B, Martínez-Silgado A, Stein JM, Dostanić M, Frimat JP, Gontan C, de Graaf MNS, Hu M, Kasi DG, Koch LS, Le KTT, Lim S, Middelkamp HHT, Mooiweer J, Motreuil-Ragot P, Niggl E, Pleguezuelos-Manzano C, Puschhof J, Revyn N, Rivera-Arbelaez JM, Slager J, Windt LM, Zakharova M, van Meer BJ, Orlova VV, de Vrij FMS, Withoff S, Mastrangeli M, van der Meer AD, Mummery CL. Standardizing designed and emergent quantitative features in microphysiological systems. Nat Biomed Eng 2024; 8:941-962. [PMID: 39187664 DOI: 10.1038/s41551-024-01236-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 04/06/2024] [Indexed: 08/28/2024]
Abstract
Microphysiological systems (MPSs) are cellular models that replicate aspects of organ and tissue functions in vitro. In contrast with conventional cell cultures, MPSs often provide physiological mechanical cues to cells, include fluid flow and can be interlinked (hence, they are often referred to as microfluidic tissue chips or organs-on-chips). Here, by means of examples of MPSs of the vascular system, intestine, brain and heart, we advocate for the development of standards that allow for comparisons of quantitative physiological features in MPSs and humans. Such standards should ensure that the in vivo relevance and predictive value of MPSs can be properly assessed as fit-for-purpose in specific applications, such as the assessment of drug toxicity, the identification of therapeutics or the understanding of human physiology or disease. Specifically, we distinguish designed features, which can be controlled via the design of the MPS, from emergent features, which describe cellular function, and propose methods for improving MPSs with readouts and sensors for the quantitative monitoring of complex physiology towards enabling wider end-user adoption and regulatory acceptance.
Collapse
Affiliation(s)
- Dennis M Nahon
- Leiden University Medical Center, Leiden, the Netherlands
| | - Renée Moerkens
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Bas Lendemeijer
- Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Adriana Martínez-Silgado
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, the Netherlands
| | - Jeroen M Stein
- Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Cristina Gontan
- Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Michel Hu
- Leiden University Medical Center, Leiden, the Netherlands
| | - Dhanesh G Kasi
- Leiden University Medical Center, Leiden, the Netherlands
| | - Lena S Koch
- University of Twente, Enschede, the Netherlands
| | - Kieu T T Le
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sangho Lim
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, the Netherlands
| | | | - Joram Mooiweer
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Eva Niggl
- Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Jens Puschhof
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and UMC Utrecht, Utrecht, the Netherlands
| | - Nele Revyn
- Delft University of Technology, Delft, the Netherlands
| | | | - Jelle Slager
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Laura M Windt
- Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | - Sebo Withoff
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | | | | |
Collapse
|
17
|
Nahon DM, Vila Cuenca M, van den Hil FE, Hu M, de Korte T, Frimat JP, van den Maagdenberg AMJM, Mummery CL, Orlova VV. Self-assembling 3D vessel-on-chip model with hiPSC-derived astrocytes. Stem Cell Reports 2024; 19:946-956. [PMID: 38876110 PMCID: PMC11252484 DOI: 10.1016/j.stemcr.2024.05.006] [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: 01/16/2023] [Revised: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 06/16/2024] Open
Abstract
Functionality of the blood-brain barrier (BBB) relies on the interaction between endothelial cells (ECs), pericytes, and astrocytes to regulate molecule transport within the central nervous system. Most experimental models for the BBB rely on freshly isolated primary brain cells. Here, we explored human induced pluripotent stem cells (hiPSCs) as a cellular source for astrocytes in a 3D vessel-on-chip (VoC) model. Self-organized microvascular networks were formed by combining hiPSC-derived ECs, human brain vascular pericytes, and hiPSC-derived astrocytes within a fibrin hydrogel. The hiPSC-ECs and pericytes showed close interactions, but, somewhat unexpectedly, addition of astrocytes disrupted microvascular network formation. However, continuous fluid perfusion or activation of cyclic AMP (cAMP) signaling rescued the vascular organization and decreased vascular permeability. Nevertheless, astrocytes did not affect the expression of proteins related to junction formation, transport, or extracellular matrix, indicating that, despite other claims, hiPSC-derived ECs do not entirely acquire a BBB-like identity in the 3D VoC model.
Collapse
Affiliation(s)
- Dennis M Nahon
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZA Leiden, the Netherlands
| | - Marc Vila Cuenca
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZA Leiden, the Netherlands; Department of Clinical Genetics, Leiden University Medical Centre, 2333ZA Leiden, the Netherlands
| | - Francijna E van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZA Leiden, the Netherlands
| | - Michel Hu
- Department of Human Genetics, Leiden University Medical Centre, 2333ZA Leiden, the Netherlands; Department of Neurology, Leiden University Medical Centre, 2333ZA Leiden, the Netherlands
| | - Tessa de Korte
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZA Leiden, the Netherlands
| | - Jean-Philippe Frimat
- Department of Human Genetics, Leiden University Medical Centre, 2333ZA Leiden, the Netherlands; Department of Neurology, Leiden University Medical Centre, 2333ZA Leiden, the Netherlands
| | - Arn M J M van den Maagdenberg
- Department of Human Genetics, Leiden University Medical Centre, 2333ZA Leiden, the Netherlands; Department of Neurology, Leiden University Medical Centre, 2333ZA Leiden, the Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZA Leiden, the Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333ZA Leiden, the Netherlands.
| |
Collapse
|
18
|
Sun J, Song S. Advances in modeling permeability and selectivity of the blood-brain barrier using microfluidics. MICROFLUIDICS AND NANOFLUIDICS 2024; 28:44. [PMID: 39781566 PMCID: PMC11709447 DOI: 10.1007/s10404-024-02741-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/15/2024] [Indexed: 01/12/2025]
Abstract
The blood-brain barrier (BBB) protects the brain by actively allowing the entry of ions and nutrients while limiting the passage of from toxins and pathogens. A healthy BBB has low permeability and high selectivity to maintain normal brain functions. Increased BBB permeability can result from neurological diseases and traumatic injuries. Modern engineering technologies such as microfluidics and fabrication techniques have advanced the development of BBB models to simulate the basic functions of BBB. However, the intrinsic BBB properties are difficult to replicate. Existing in vitro BBB models demonstrate inconsistent BBB permeability and selectivity due to variations in microfluidic design, cell types and arrangement, expression of tight junction (TJ) proteins, and use of shear stress. Specifically, microfluidic designs have flow channels of different sizes, complexity, topology, and modular structure. Different cell types are selected to mimic various physiological conditions. These factors make it challenging to compare results obtained using different experimental setups. This paper highlights key factors that play important roles in influencing microfluidic models and discusses how these factors contribute to permeability and selectivity of the BBB models.
Collapse
Affiliation(s)
- Jindi Sun
- Department of Biomedical Engineering, The University of Arizona, 1200 E University Blvd, Tucson 85721, Arizona, USA
| | - Shang Song
- Department of Biomedical Engineering, The University of Arizona, 1200 E University Blvd, Tucson 85721, Arizona, USA
- Departments of Neuroscience GIDP, Materials Science and Engineering, and BIO5 Institute, The University of Arizona, 1200 E University Blvd, Tucson 85721, Arizona, USA
| |
Collapse
|
19
|
Iqbal I, Saqib F, Mubarak Z, Latif MF, Wahid M, Nasir B, Shahzad H, Sharifi-Rad J, Mubarak MS. Alzheimer's disease and drug delivery across the blood-brain barrier: approaches and challenges. Eur J Med Res 2024; 29:313. [PMID: 38849950 PMCID: PMC11161981 DOI: 10.1186/s40001-024-01915-3] [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: 02/07/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024] Open
Abstract
Alzheimer's disease (AD) is a diverse disease with a complex pathophysiology. The presence of extracellular β-amyloid deposition as neuritic plaques and intracellular accumulation of hyper-phosphorylated tau as neurofibrillary tangles remain the core neuropathologic criteria for diagnosing Alzheimer's disease. Nonetheless, several recent basic discoveries have revealed significant pathogenic roles for other essential cellular and molecular processes. Previously, there were not so many disease-modifying medications (DMT) available as drug distribution through the blood-brain barrier (BBB) is difficult due to its nature, especially drugs of polypeptides nature and proteins. Recently FDA has approved lecanemab as DMT for its proven efficacy. It is also complicated to deliver drugs for diseases like epilepsy or any brain tumor due to the limitations of the BBB. After the advancements in the drug delivery system, different techniques are used to transport the medication across the BBB. Other methods are used, like enhancement of brain blood vessel fluidity by liposomes, infusion of hyperosmotic solutions, and local intracerebral implants, but these are invasive approaches. Non-invasive approaches include the formulation of nanoparticles and their coating with polymers. This review article emphasizes all the above-mentioned techniques, procedures, and challenges to transporting medicines across the BBB. It summarizes the most recent literature dealing with drug delivery across the BBB.
Collapse
Affiliation(s)
- Iram Iqbal
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
- Primary and Secondary Healthcare Department, Govt of the Punjab, Lahore, Pakistan
| | - Fatima Saqib
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan.
| | - Zobia Mubarak
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan
- Primary and Secondary Healthcare Department, Govt of the Punjab, Lahore, Pakistan
| | - Muhammad Farhaj Latif
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Muqeet Wahid
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Bushra Nasir
- Department of Pharmaceutics, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Hamna Shahzad
- Department of Biochemistry, Bahauddin Zakariya University Multan, Multan, Pakistan
| | - Javad Sharifi-Rad
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea.
| | - Mohammad S Mubarak
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
20
|
Dao L, You Z, Lu L, Xu T, Sarkar AK, Zhu H, Liu M, Calandrelli R, Yoshida G, Lin P, Miao Y, Mierke S, Kalva S, Zhu H, Gu M, Vadivelu S, Zhong S, Huang LF, Guo Z. Modeling blood-brain barrier formation and cerebral cavernous malformations in human PSC-derived organoids. Cell Stem Cell 2024; 31:818-833.e11. [PMID: 38754427 PMCID: PMC11162335 DOI: 10.1016/j.stem.2024.04.019] [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: 06/30/2023] [Revised: 02/24/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024]
Abstract
The human blood-brain barrier (hBBB) is a highly specialized structure that regulates passage across blood and central nervous system (CNS) compartments. Despite its critical physiological role, there are no reliable in vitro models that can mimic hBBB development and function. Here, we constructed hBBB assembloids from brain and blood vessel organoids derived from human pluripotent stem cells. We validated the acquisition of blood-brain barrier (BBB)-specific molecular, cellular, transcriptomic, and functional characteristics and uncovered an extensive neuro-vascular crosstalk with a spatial pattern within hBBB assembloids. When we used patient-derived hBBB assembloids to model cerebral cavernous malformations (CCMs), we found that these assembloids recapitulated the cavernoma anatomy and BBB breakdown observed in patients. Upon comparison of phenotypes and transcriptome between patient-derived hBBB assembloids and primary human cavernoma tissues, we uncovered CCM-related molecular and cellular alterations. Taken together, we report hBBB assembloids that mimic the core properties of the hBBB and identify a potentially underlying cause of CCMs.
Collapse
Affiliation(s)
- Lan Dao
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Zhen You
- Department of Pediatric and Adolescent Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Lu Lu
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Tianyang Xu
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Avijite Kumer Sarkar
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Hui Zhu
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Miao Liu
- Department of Pediatric and Adolescent Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Riccardo Calandrelli
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - George Yoshida
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Pei Lin
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yifei Miao
- Center for Stem Cell and Organoid Medicine, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sarah Mierke
- Divisions of Pediatric Neurosurgery and Interventional Neuroradiology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | - Srijan Kalva
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Haining Zhu
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sudhakar Vadivelu
- Divisions of Pediatric Neurosurgery and Interventional Neuroradiology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | - Sheng Zhong
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| | - L Frank Huang
- Department of Pediatric and Adolescent Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA.
| | - Ziyuan Guo
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
21
|
Kumar V, Banerjee A, Roy K. Breaking the Barriers: Machine-Learning-Based c-RASAR Approach for Accurate Blood-Brain Barrier Permeability Prediction. J Chem Inf Model 2024; 64:4298-4309. [PMID: 38700741 DOI: 10.1021/acs.jcim.4c00433] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2024]
Abstract
The intricate nature of the blood-brain barrier (BBB) poses a significant challenge in predicting drug permeability, which is crucial for assessing central nervous system (CNS) drug efficacy and safety. This research utilizes an innovative approach, the classification read-across structure-activity relationship (c-RASAR) framework, that leverages machine learning (ML) to enhance the accuracy of BBB permeability predictions. The c-RASAR framework seamlessly integrates principles from both read-across and QSAR methodologies, underscoring the need to consider similarity-related aspects during the development of the c-RASAR model. It is crucial to note that the primary goal of this research is not to introduce yet another model for predicting BBB permeability but rather to showcase the refinement in predicting the BBB permeability of organic compounds through the introduction of a c-RASAR approach. This groundbreaking methodology aims to elevate the accuracy of assessing neuropharmacological implications and streamline the process of drug development. In this study, an ML-based c-RASAR linear discriminant analysis (LDA) model was developed using a dataset of 7807 compounds, encompassing both BBB-permeable and -nonpermeable substances sourced from the B3DB database (freely accessible from https://github.com/theochem/B3DB), for predicting BBB permeability in lead discovery for CNS drugs. The model's predictive capability was then validated using three external sets: one containing 276,518 natural products (NPs) from the LOTUS database (accessible from https://lotus.naturalproducts.net/download) for data gap filling, another comprising 13,002 drug-like/drug compounds from the DrugBank database (available from https://go.drugbank.com/), and a third set of 56 FDA-approved drugs to assess the model's reliability. Further diversifying the predictive arsenal, various other ML-based c-RASAR models were also developed for comparison purposes. The proposed c-RASAR framework emerged as a powerful tool for predicting BBB permeability. This research not only advances the understanding of molecular determinants influencing CNS drug permeability but also provides a versatile computational platform for the rapid assessment of diverse compounds, facilitating informed decision-making in drug development and design.
Collapse
Affiliation(s)
- Vinay Kumar
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Arkaprava Banerjee
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Kunal Roy
- Drug Theoretics and Cheminformatics Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| |
Collapse
|
22
|
Xu Z, You Y, Bai S, Wang L, Liu C. Microliquid/Liquid Interfacial Sensors: Biomimetic Investigation of Transmembrane Mechanisms and Real-Time Determinations of Clemastine, Cyproheptadine, Epinastine, Cetirizine, and Desloratadine. Anal Chem 2024; 96:6599-6608. [PMID: 38640514 DOI: 10.1021/acs.analchem.3c05640] [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: 04/21/2024]
Abstract
Antihistamines relieve allergic symptoms by inhibiting the action of histamine. Further understanding of antihistamine transmembrane mechanisms and optimizing the selectivity and real-time monitoring capabilities of drug sensors is necessary. In this study, a micrometer liquid/liquid (L/L) interfacial sensor has served as a biomimetic membrane to investigate the mechanism of interfacial transfer of five antihistamines, i.e., clemastine (CLE), cyproheptadine (CYP), epinastine (EPI), desloratadine (DSL), and cetirizine (CET), and realize the real-time determinations. Cyclic voltammetry (CV) and differential pulse voltammetry (DPV) techniques have been used to uncover the electrochemical transfer behavior of the five antihistamines at the L/L interface. Additionally, finite element simulations (FEMs) have been employed to reveal the thermodynamics and kinetics of the process. Visualization of antihistamine partitioning in two phases at different pH values can be realized by ion partition diagrams (IPDs). The IPDs also reveal the transfer mechanism at the L/L interface and provide effective lipophilicity at different pH values. Real-time determinations of these antihistamines have been achieved through potentiostatic chronoamperometry (I-t), exhibiting good selectivity with the addition of nine common organic or inorganic compounds in living organisms and revealing the potential for in vivo pharmacokinetics. Besides providing a satisfactory surrogate for studying the transmembrane mechanism of antihistamines, this work also sheds light on micro- and nano L/L interfacial sensors for in vivo analysis of pharmacokinetics at a single-cell or single-organelle level.
Collapse
Affiliation(s)
- Zhidan Xu
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510641, China
| | - Yongtao You
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510641, China
| | - Silan Bai
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510641, China
| | - Lishi Wang
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510641, China
| | - Cheng Liu
- Center for Advanced Analytical Science, Guangzhou Key Laboratory of Sensing Materials & Devices, Guangdong Key Laboratory for Photoelectric Sensing Materials and Devices, School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou 510006, China
| |
Collapse
|
23
|
Boyetey MJB, Choi Y, Lee HY, Choi J. Nanotechnology-based delivery of therapeutics through the intranasal pathway and the blood-brain barrier for Alzheimer's disease treatment. Biomater Sci 2024; 12:2007-2018. [PMID: 38456516 DOI: 10.1039/d3bm02003g] [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: 03/09/2024]
Abstract
Background: drugs for Alzheimer's disease (AD) fail to exhibit efficacy in clinical trials for a number of reasons, a major one being blood-brain barrier (BBB) permeability. Meanwhile, the increasing incidence of this disease emphasizes the need for effective therapeutics. Herein, we discuss novel nanoplatform technologies developed for the effective delivery of AD drugs by traversing the BBB. Main text: the interfacial and surface chemistry of nanomaterials is utilized in several industries, including pharmaceutical, and has drawn considerable attention in the field of nanotechnology. Various reports have suggested the potential of nanotechnology for AD treatment, describing unique drug carriers that improve drug stability and solubility while maintaining therapeutic dosages. These nanotechnologies are harnessed for the transport of drugs across the BBB, with or without surface modifications. We also discuss the transfer of drugs via the nose-to-brain pathway, as intranasal delivery enables direct drug distribution in the brain. In addition, nanomaterial modifications that prolong drug delivery and improve safety following intranasal administration are addressed. Conclusion: although several studies have yielded promising results, limited efforts have been undertaken to translate research findings into clinical contexts. Nevertheless, nanomaterials hold considerable potential for the development of novel effective therapeutic solutions against AD.
Collapse
Affiliation(s)
| | - Yonghyun Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea.
- Feynman Institute of Technology, Nanomedicine Corporation, Seoul 06974, Republic of Korea
| | - Hee-Young Lee
- Department of Chemical Engineering, Kumoh National Institute of Technology, 61, Daehak-ro, Gumi-si, Gyeongsangbuk-do 39177, Republic of Korea.
| | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea.
- Feynman Institute of Technology, Nanomedicine Corporation, Seoul 06974, Republic of Korea
| |
Collapse
|
24
|
Zapata-Acevedo JF, Losada-Barragán M, Osma JF, Cruz JC, Reiber A, Petry KG, Caillard A, Sauldubois A, Llamosa Pérez D, Morillo Zárate AJ, Muñoz SB, Daza Moreno A, Silva RV, Infante-Duarte C, Chamorro-Coral W, González-Reyes RE, Vargas-Sánchez K. Specific nanoprobe design for MRI: Targeting laminin in the blood-brain barrier to follow alteration due to neuroinflammation. PLoS One 2024; 19:e0302031. [PMID: 38603692 PMCID: PMC11008835 DOI: 10.1371/journal.pone.0302031] [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: 01/16/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Chronic neuroinflammation is characterized by increased blood-brain barrier (BBB) permeability, leading to molecular changes in the central nervous system that can be explored with biomarkers of active neuroinflammatory processes. Magnetic resonance imaging (MRI) has contributed to detecting lesions and permeability of the BBB. Ultra-small superparamagnetic particles of iron oxide (USPIO) are used as contrast agents to improve MRI observations. Therefore, we validate the interaction of peptide-88 with laminin, vectorized on USPIO, to explore BBB molecular alterations occurring during neuroinflammation as a potential tool for use in MRI. The specific labeling of NPS-P88 was verified in endothelial cells (hCMEC/D3) and astrocytes (T98G) under inflammation induced by interleukin 1β (IL-1β) for 3 and 24 hours. IL-1β for 3 hours in hCMEC/D3 cells increased their co-localization with NPS-P88, compared with controls. At 24 hours, no significant differences were observed between groups. In T98G cells, NPS-P88 showed similar nonspecific labeling among treatments. These results indicate that NPS-P88 has a higher affinity towards brain endothelial cells than astrocytes under inflammation. This affinity decreases over time with reduced laminin expression. In vivo results suggest that following a 30-minute post-injection, there is an increased presence of NPS-P88 in the blood and brain, diminishing over time. Lastly, EAE animals displayed a significant accumulation of NPS-P88 in MRI, primarily in the cortex, attributed to inflammation and disruption of the BBB. Altogether, these results revealed NPS-P88 as a biomarker to evaluate changes in the BBB due to neuroinflammation by MRI in biological models targeting laminin.
Collapse
Affiliation(s)
- Juan F. Zapata-Acevedo
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Mónica Losada-Barragán
- Grupo de Biología Celular y Funcional e Ingeniería de Biomoleculas, Departamento de Biología, Universidad Antonio Nariño, Bogotá, Colombia
| | - Johann F. Osma
- Department of Electrical and Electronic Engineering, Universidad de los Andes, Bogotá, Colombia
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá, Colombia
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá, Colombia
| | - Andreas Reiber
- Chemistry Department, Grupo La Quimica en la interfase inorgánica-orgánica QUINORG, Universidad de los Andes, Bogotá, Colombia
| | - Klaus G. Petry
- CNRS UMR 5536 Centre de Resonance Magnétique des Systemes Biologiques and INSERM U1049 Neuroinflammation, University of Bordeaux, Bordeaux, France
| | | | | | - Daniel Llamosa Pérez
- Facultad de Ciencias, Grupo Investigación fundamental y aplicada en Materiales, Universidad Antonio Nariño, Bogotá, Colombia
| | | | | | - Agustín Daza Moreno
- Oficial de Protección Radiológica, Fundación Santa Fé de Bogotá, Bogotá, Colombia
| | - Rafaela V. Silva
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Carmen Infante-Duarte
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - William Chamorro-Coral
- Laboratorio de Neurofisiología Celular, Grupo de Neurociencia Traslacional, Facultad de Medicina, Universidad de los Andes, Bogotá, Colombia
| | - Rodrigo E. González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Karina Vargas-Sánchez
- Laboratorio de Neurofisiología Celular, Grupo de Neurociencia Traslacional, Facultad de Medicina, Universidad de los Andes, Bogotá, Colombia
| |
Collapse
|
25
|
Aazmi A, Zhang D, Mazzaglia C, Yu M, Wang Z, Yang H, Huang YYS, Ma L. Biofabrication methods for reconstructing extracellular matrix mimetics. Bioact Mater 2024; 31:475-496. [PMID: 37719085 PMCID: PMC10500422 DOI: 10.1016/j.bioactmat.2023.08.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/19/2023] Open
Abstract
In the human body, almost all cells interact with extracellular matrices (ECMs), which have tissue and organ-specific compositions and architectures. These ECMs not only function as cellular scaffolds, providing structural support, but also play a crucial role in dynamically regulating various cellular functions. This comprehensive review delves into the examination of biofabrication strategies used to develop bioactive materials that accurately mimic one or more biophysical and biochemical properties of ECMs. We discuss the potential integration of these ECM-mimics into a range of physiological and pathological in vitro models, enhancing our understanding of cellular behavior and tissue organization. Lastly, we propose future research directions for ECM-mimics in the context of tissue engineering and organ-on-a-chip applications, offering potential advancements in therapeutic approaches and improved patient outcomes.
Collapse
Affiliation(s)
- Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Duo Zhang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 51817, China
| | - Corrado Mazzaglia
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhen Wang
- Center for Laboratory Medicine, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Huayong Yang
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yan Yan Shery Huang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
26
|
Rajagopalan NR, Vista WR, Fujimori M, Vroomen LGPH, Jiménez JM, Khadka N, Bikson M, Srimathveeravalli G. Cytoskeletal Remodeling and Gap Junction Translocation Mediates Blood-Brain Barrier Disruption by Non-invasive Low-Voltage Pulsed Electric Fields. Ann Biomed Eng 2024; 52:89-102. [PMID: 37115366 DOI: 10.1007/s10439-023-03211-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 04/14/2023] [Indexed: 04/29/2023]
Abstract
High-voltage pulsed electric fields (HV-PEF) delivered with invasive needle electrodes for electroporation applications is known to induce off-target blood-brain barrier (BBB) disruption. In this study, we sought to determine the feasibility of minimally invasive PEF application to produce BBB disruption in rat brain and identify the putative mechanisms mediating the effect. We observed dose-dependent presence of Evans Blue (EB) dye in rat brain when PEF were delivered with a skull mounted electrode used for neurostimulation application. Maximum region of dye uptake was observed while using 1500 V, 100 pulses, 100 µs and 10 Hz. Results of computational models suggested that the region of BBB disruption was occurring at thresholds of 63 V/cm or higher; well below intensity levels for electroporation. In vitro experiments recapitulating this effect with human umbilical vein endothelial cells (HUVEC) demonstrated cellular alterations that underlie BBB manifests at low-voltage high-pulse conditions without affecting cell viability or proliferation. Morphological changes in HUVECs due to PEF were accompanied by disruption of actin cytoskeleton, loss of tight junction protein-ZO-1 and VE-Cadherin at cell junctions and partial translocation into the cytoplasm. Uptake of propidium iodide (PI) in PEF treated conditions is less than 1% and 2.5% of total number of cells in high voltage (HV) and low-voltage (LV) groups, respectively, implying that BBB disruption to be independent of electroporation under these conditions. 3-D microfabricated blood vessel permeability was found to increase significantly following PEF treatment and confirmed with correlative cytoskeletal changes and loss of tight junction proteins. Finally, we show that the rat brain model can be scaled to human brains with a similar effect on BBB disruption characterized by electric field strength (EFS) threshold and using a combination of two bilateral HD electrode configurations.
Collapse
Affiliation(s)
| | - William-Ray Vista
- Department of Radiology, Interventional Radiology Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Masashi Fujimori
- Department of Radiology, Interventional Radiology Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Department of Radiology, Mie University, Tsu, Mie, Japan
| | | | - Juan M Jiménez
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, USA
| | - Niranjan Khadka
- Division of Neuromodulation, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Synchron Inc, Brooklyn, NY, USA
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Marom Bikson
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Govindarajan Srimathveeravalli
- Department of Mechanical and Industrial Engineering, University of Massachusetts Amherst, Amherst, MA, USA.
- Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA, USA.
| |
Collapse
|
27
|
Liabeuf S, Pešić V, Spasovski G, Maciulaitis R, Bobot M, Farinha A, Wagner CA, Unwin RJ, Capasso G, Bumblyte IA, Hafez G. Drugs with a negative impact on cognitive function (Part 1): chronic kidney disease as a risk factor. Clin Kidney J 2023; 16:2365-2377. [PMID: 38045996 PMCID: PMC10689135 DOI: 10.1093/ckj/sfad241] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Indexed: 12/05/2023] Open
Abstract
People living with chronic kidney disease (CKD) frequently suffer from mild cognitive impairment and/or other neurocognitive disorders. This review in two parts will focus on adverse drug reactions resulting in cognitive impairment as a potentially modifiable risk factor in CKD patients. Many patients with CKD have a substantial burden of comorbidities leading to polypharmacy. A recent study found that patients seen by nephrologists were the most complex to treat because of their high number of comorbidities and medications. Due to polypharmacy, these patients may experience a wide range of adverse drug reactions. Along with CKD progression, the accumulation of uremic toxins may lead to blood-brain barrier (BBB) disruption and pharmacokinetic alterations, increasing the risk of adverse reactions affecting the central nervous system (CNS). In patients on dialysis, the excretion of drugs that depend on kidney function is severely reduced such that adverse and toxic levels of a drug or its metabolites may be reached at relatively low doses, unless dosing is adjusted. This first review will discuss how CKD represents a risk factor for adverse drug reactions affecting the CNS via (i) BBB disruption associated with CKD and (ii) the impact of reduced kidney function and dialysis itself on drug pharmacokinetics.
Collapse
Affiliation(s)
- Sophie Liabeuf
- Pharmacoepidemiology Unit, Department of Clinical Pharmacology, Amiens University Medical Center, Amiens, France
- MP3CV Laboratory, EA7517, Jules Verne University of Picardie, Amiens, France
| | - Vesna Pešić
- Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Goce Spasovski
- Department of Nephrology, Clinical Centre “Mother Theresa”, Saints Cyril and Methodius University, Skopje, North Macedonia
| | - Romaldas Maciulaitis
- Department of Nephrology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Institute of Physiology and Pharmacology, Faculty of Medicines, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Mickaël Bobot
- Aix-Marseille University, Department of Nephrology, AP-HM, La Conception Hospital, Marseille, France; C2VN Laboratory, Inserm 1263, INRAE 1260, Aix-Marseille University, Marseille, France
| | - Ana Farinha
- Department of Nephrology, Hospital de Vila Franca de Xira, Lisbon, Portugal
| | - Carsten A Wagner
- Institute of Physiology, University of Zürich, Zurich, Switzerland
| | - Robert J Unwin
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| | - Giovambattista Capasso
- Department of Translantional Medical Sciences, University of Campania Luigi Vanvitelli , Naples, Italy
- Biogem Research Institute , Ariano Irpino, Italy
| | - Inga Arune Bumblyte
- Department of Nephrology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Gaye Hafez
- Department of Pharmacology, Faculty of Pharmacy, Altinbas University, Istanbul, Turkey
| |
Collapse
|
28
|
Janjua TI, Cao Y, Kleitz F, Linden M, Yu C, Popat A. Silica nanoparticles: A review of their safety and current strategies to overcome biological barriers. Adv Drug Deliv Rev 2023; 203:115115. [PMID: 37844843 DOI: 10.1016/j.addr.2023.115115] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/18/2023]
Abstract
Silica nanoparticles (SNP) have gained tremendous attention in the recent decades. They have been used in many different biomedical fields including diagnosis, biosensing and drug delivery. Medical uses of SNP for anti-cancer, anti-microbial and theranostic applications are especially prominent due to their exceptional performance to deliver many different small molecules and recently biologics (mRNA, siRNA, antigens, antibodies, proteins, and peptides) at targeted sites. The physical and chemical properties of SNP such as large specific surface area, tuneable particle size and porosity, excellent biodegradability and biocompatibility make them an ideal drug delivery and diagnostic platform. Based on the available data and the pre-clinical performance of SNP, recent interest has driven these innovative materials towards clinical application with many of the formulations already in Phase I and Phase II trials. Herein, the progress of SNP in biomedical field is reviewed, and their safety aspects are analysed. Importantly, we critically evaluate the key structural characteristics of SNP to overcome different biological barriers including the blood-brain barrier (BBB), skin, tumour barrier and mucosal barrier. Future directions, potential pathways, and target areas towards rapid clinical translation of SNP are also recommended.
Collapse
Affiliation(s)
- Taskeen Iqbal Janjua
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia.
| | - Yuxue Cao
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Freddy Kleitz
- Department of Functional Materials and Catalysis, Faculty of Chemistry, University of Vienna, Währinger Straße 42, 1090 Vienna, Austria
| | - Mika Linden
- Institute of Inorganic Chemistry II, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Queensland, QLD 4072, Australia.
| | - Amirali Popat
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia; Department of Functional Materials and Catalysis, Faculty of Chemistry, University of Vienna, Währinger Straße 42, 1090 Vienna, Austria.
| |
Collapse
|
29
|
Niazi SK. Non-Invasive Drug Delivery across the Blood-Brain Barrier: A Prospective Analysis. Pharmaceutics 2023; 15:2599. [PMID: 38004577 PMCID: PMC10674293 DOI: 10.3390/pharmaceutics15112599] [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: 10/12/2023] [Revised: 11/01/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Non-invasive drug delivery across the blood-brain barrier (BBB) represents a significant advancement in treating neurological diseases. The BBB is a tightly packed layer of endothelial cells that shields the brain from harmful substances in the blood, allowing necessary nutrients to pass through. It is a highly selective barrier, which poses a challenge to delivering therapeutic agents into the brain. Several non-invasive procedures and devices have been developed or are currently being investigated to enhance drug delivery across the BBB. This paper presents a review and a prospective analysis of the art and science that address pharmacology, technology, delivery systems, regulatory approval, ethical concerns, and future possibilities.
Collapse
Affiliation(s)
- Sarfaraz K Niazi
- College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
30
|
Hang Z, Zhou L, Xing C, Wen Y, Du H. The blood-brain barrier, a key bridge to treat neurodegenerative diseases. Ageing Res Rev 2023; 91:102070. [PMID: 37704051 DOI: 10.1016/j.arr.2023.102070] [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: 06/09/2023] [Revised: 09/06/2023] [Accepted: 09/09/2023] [Indexed: 09/15/2023]
Abstract
As a highly selective and semi-permeable barrier that separates the circulating blood from the brain and central nervous system (CNS), the blood-brain barrier (BBB) plays a critical role in the onset and treatment of neurodegenerative diseases (NDs). To delay or reverse the NDs progression, the dysfunction of BBB should be improved to protect the brain from harmful substances. Simultaneously, a highly efficient drug delivery across the BBB is indispensable. Here, we summarized several methods to improve BBB dysfunction in NDs, including knocking out risk geneAPOE4, regulating circadian rhythms, restoring the gut microenvironment, and activating the Wnt/β-catenin signaling pathway. Then we discussed the advances in BBB penetration techniques, such as transient BBB opening, carrier-mediated drug delivery, and nasal administration, which facilitates drug delivery across the BBB. Furthermore, various in vivo and in vitro BBB models and research methods related to NDs are reviewed. Based on the current research progress, the treatment of NDs in the long term should prioritize the integrity of the BBB. However, a treatment approach that combines precise control of transient BBB permeability and non-invasive targeted BBB drug delivery holds profound significance in improving treatment effectiveness, safety, and clinical feasibility during drug therapy. This review involves the cross application of biology, materials science, imaging, engineering and other disciplines in the field of BBB, aiming to provide multi-dimensional research directions and clinical ideas for the treating NDs.
Collapse
Affiliation(s)
- Zhongci Hang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Liping Zhou
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; Beijing Key Laboratory for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China
| | - Cencan Xing
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Yongqiang Wen
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Beijing Key Laboratory for Bioengineering and Sensing Technology, University of Science and Technology Beijing, Beijing 100083, China.
| | - Hongwu Du
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China.
| |
Collapse
|
31
|
Ali A, Sohail Arshad M, Ahmad Khan M, Chang MW, Ahmad Z. Recent advances towards overcoming the blood-brain barrier. Drug Discov Today 2023; 28:103735. [PMID: 37573965 DOI: 10.1016/j.drudis.2023.103735] [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: 05/19/2023] [Revised: 07/28/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
The blood-brain barrier (BBB) is a protective element of the neurovascular unit (NVU) surrounded by astrocytes, pericytes, extracellular matrix, and the tight junctional complex, which play a fundamental role in brain homeostasis. Due to its impeccable structural architecture, the BBB is referred to as the brain's gatekeeper, a near-impenetrable barrier to therapeutics. This review summarises the significant strides that have been made in the last 5 years towards circumventing the BBB and developing efficient drug delivery systems. Challenges associated with several CNS disorders related to BBB failure and exploitation of this unique NVU component for targeted treatment of brain-related disorders are also discussed.
Collapse
Affiliation(s)
- Amna Ali
- Leicester School of Pharmacy, De Montfort University, Leicester, UK
| | | | - Mahtab Ahmad Khan
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Ming-Wei Chang
- Nanotechnology and Integrated Bioengineering Centre, University of Ulster, Belfast, UK
| | - Zeeshan Ahmad
- Leicester School of Pharmacy, De Montfort University, Leicester, UK.
| |
Collapse
|
32
|
Jakimiuk A, Piechal A, Wiercińska-Drapało A, Nowaczyk A, Mirowska-Guzel D. Central nervous system disorders after use of dolutegravir: evidence from preclinical and clinical studies. Pharmacol Rep 2023; 75:1138-1151. [PMID: 37605102 PMCID: PMC10539422 DOI: 10.1007/s43440-023-00515-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/23/2023]
Abstract
The evaluation of dolutegravir based on available preclinical and clinical studies reveals a risk of central nervous system (CNS) disorders associated with long-term use of the drug. The available literature on the pharmacokinetics of the drug, including its penetration of the blood-brain barrier, was reviewed, as well as clinical trials assessing the incidence of adverse effects in the CNS and the frequency of its discontinuation. This paper also summarizes the impact of factors affecting the occurrence of CNS disorders and indicates the key role of pharmacovigilance in the process of supplementing knowledge on the safety of drugs, especially those that are newly registered.
Collapse
Affiliation(s)
- Alicja Jakimiuk
- Department of Clinical and Experimental Pharmacology, Centre for Preclinical Research and Technology, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Agnieszka Piechal
- Department of Clinical and Experimental Pharmacology, Centre for Preclinical Research and Technology, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Alicja Wiercińska-Drapało
- Department of Hepatology and Infectious and Tropical Diseases, Medical University of Warsaw, Provincial Infectious Diseases Hospital in Warsaw, Wolska 37, 01-201, Warsaw, Poland
| | - Alicja Nowaczyk
- Department of Organic Chemistry, Faculty of Pharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 2 dr. A. Jurasza, 85-094, Bydgoszcz, Poland
| | - Dagmara Mirowska-Guzel
- Department of Clinical and Experimental Pharmacology, Centre for Preclinical Research and Technology, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland.
| |
Collapse
|
33
|
Kawakita S, Li S, Nguyen HT, Maity S, Haghniaz R, Bahari J, Yu N, Mandal K, Bandaru P, Mou L, Ermis M, Khalil E, Khosravi S, Peirsman A, Nasiri R, Adachi A, Nakayama A, Bell R, Zhu Y, Jucaud V, Dokmeci MR, Khademhosseini A. Rapid integration of screen-printed electrodes into thermoplastic organ-on-a-chip devices for real-time monitoring of trans-endothelial electrical resistance. Biomed Microdevices 2023; 25:37. [PMID: 37740819 DOI: 10.1007/s10544-023-00669-9] [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] [Accepted: 07/24/2023] [Indexed: 09/25/2023]
Abstract
Trans-endothelial electrical resistance (TEER) is one of the most widely used indicators to quantify the barrier integrity of endothelial layers. Over the last decade, the integration of TEER sensors into organ-on-a-chip (OOC) platforms has gained increasing interest for its efficient and effective measurement of TEER in OOCs. To date, microfabricated electrodes or direct insertion of wires has been used to integrate TEER sensors into OOCs, with each method having advantages and disadvantages. In this study, we developed a TEER-SPE chip consisting of carbon-based screen-printed electrodes (SPEs) embedded in a poly(methyl methacrylate) (PMMA)-based multi-layered microfluidic device with a porous poly(ethylene terephthalate) membrane in-between. As proof of concept, we demonstrated the successful cultures of hCMEC/D3 cells and the formation of confluent monolayers in the TEER-SPE chip and obtained TEER measurements for 4 days. Additionally, the TEER-SPE chip could detect changes in the barrier integrity due to shear stress or an inflammatory cytokine (i.e., tumor necrosis factor-α). The novel approach enables a low-cost and facile fabrication of carbon-based SPEs on PMMA substrates and the subsequent assembly of PMMA layers for rapid prototyping. Being cost-effective and cleanroom-free, our method lowers the existing logistical and technical barriers presenting itself as another step forward to the broader adoption of OOCs with TEER measurement capability.
Collapse
Affiliation(s)
- Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Shaopei Li
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Jamal Bahari
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Ning Yu
- Department of Chemical and Environmental Engineering, University of California-Riverside, Riverside, California, 92521, USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Praveen Bandaru
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Lei Mou
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Enam Khalil
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- School of Pharmacy, The University of Jordan, Amman, 11942, Jordan
| | - Safoora Khosravi
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC, V6T1Z4, Canada
| | - Arne Peirsman
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Department of Plastic, Aesthetic & Reconstructive Surgery and Laboratory of Experimental Cancer Research, Ghent University, 9000, Ghent, Belgium
| | - Rohollah Nasiri
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, 17165, Solna, Sweden
| | - Annie Adachi
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Cardiovascular Research Institute, University of California, San Francisco, California, 94158, USA
| | - Aya Nakayama
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, 91125, USA
| | - Remy Bell
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC, 29208, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA.
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, USA.
| |
Collapse
|
34
|
Yan L, Dwiggins CW, Gupta U, Stroka KM. A Rapid-Patterning 3D Vessel-on-Chip for Imaging and Quantitatively Analyzing Cell-Cell Junction Phenotypes. Bioengineering (Basel) 2023; 10:1080. [PMID: 37760182 PMCID: PMC10525190 DOI: 10.3390/bioengineering10091080] [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: 05/12/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The blood-brain barrier (BBB) is a dynamic interface that regulates the molecular exchanges between the brain and peripheral blood. The permeability of the BBB is primarily regulated by the junction proteins on the brain endothelial cells. In vitro BBB models have shown great potential for the investigation of the mechanisms of physiological function, pathologies, and drug delivery in the brain. However, few studies have demonstrated the ability to monitor and evaluate the barrier integrity by quantitatively analyzing the junction presentation in 3D microvessels. This study aimed to fabricate a simple vessel-on-chip, which allows for a rigorous quantitative investigation of junction presentation in 3D microvessels. To this end, we developed a rapid protocol that creates 3D microvessels with polydimethylsiloxane and microneedles. We established a simple vessel-on-chip model lined with human iPSC-derived brain microvascular endothelial-like cells (iBMEC-like cells). The 3D image of the vessel structure can then be "unwrapped" and converted to 2D images for quantitative analysis of cell-cell junction phenotypes. Our findings revealed that 3D cylindrical structures altered the phenotype of tight junction proteins, along with the morphology of cells. Additionally, the cell-cell junction integrity in our 3D models was disrupted by the tumor necrosis factor α. This work presents a "quick and easy" 3D vessel-on-chip model and analysis pipeline, together allowing for the capability of screening and evaluating the cell-cell junction integrity of endothelial cells under various microenvironment conditions and treatments.
Collapse
Affiliation(s)
- Li Yan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.W.D.); (U.G.)
| | - Cole W. Dwiggins
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.W.D.); (U.G.)
| | - Udit Gupta
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.W.D.); (U.G.)
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (C.W.D.); (U.G.)
- Biophysics Program, University of Maryland, College Park, MD 20742, USA
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
35
|
Zhou R, Li J, Wang R, Chen Z, Zhou F. The neurovascular unit in healthy and injured spinal cord. J Cereb Blood Flow Metab 2023; 43:1437-1455. [PMID: 37190756 PMCID: PMC10414016 DOI: 10.1177/0271678x231172008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 02/09/2023] [Accepted: 03/24/2023] [Indexed: 05/17/2023]
Abstract
The neurovascular unit (NVU) reflects the close temporal and spatial link between neurons and blood vessels. However, the understanding of the NVU in the spinal cord is far from clear and largely based on generalized knowledge obtained from the brain. Herein, we review the present knowledge of the NVU and highlight candidate approaches to investigate the NVU, particularly focusing on the spinal cord. Several unique features maintain the highly regulated microenvironment in the NVU. Autoregulation and neurovascular coupling ensure regional blood flow meets the metabolic demand according to the blood supply or local neural activation. The blood-central nervous system barrier partitions the circulating blood from neural parenchyma and facilitates the selective exchange of substances. Furthermore, we discuss spinal cord injury (SCI) as a common injury from the perspective of NVU dysfunction. Hopefully, this review will help expand the understanding of the NVU in the spinal cord and inspire new insights into SCI.
Collapse
Affiliation(s)
- Rubing Zhou
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Junzhao Li
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Ruideng Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Zhengyang Chen
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Fang Zhou
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
36
|
Kaur A, Singh N, Kaur H, Kakoty V, Sharma DS, Khursheed R, Babu MR, Harish V, Gupta G, Gulati M, Kumar P, Dureja H, Alharthi NS, Khan FR, Rehman ZU, Hakami MA, Patel M, Patel R, Zandi M, Vishwas S, Dua K, Singh SK. Neurodegenerative diseases and brain delivery of therapeutics: Bridging the gap using dendrimers. J Drug Deliv Sci Technol 2023; 87:104868. [DOI: 10.1016/j.jddst.2023.104868] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
37
|
Galpayage Dona KNU, Ramirez SH, Andrews AM. A Next-Generation 3D Tissue-Engineered Model of the Human Brain Microvasculature to Study the Blood-Brain Barrier. Bioengineering (Basel) 2023; 10:817. [PMID: 37508844 PMCID: PMC10376721 DOI: 10.3390/bioengineering10070817] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/09/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
More than a billion people are affected by neurological disorders, and few have effective therapeutic options. A key challenge that has prevented promising preclinically proven strategies is the translation gap to the clinic. Humanized tissue engineering models that recreate the brain environment may aid in bridging this translational gap. Here, we showcase the methodology that allows for the practical fabrication of a comprehensive microphysicological system (MPS) of the blood-brain barrier (BBB). Compared to other existing 2D and 3D models of the BBB, this model features relevant cytoarchitecture and multicellular arrangement, with branching and network topologies of the vascular bed. This process utilizes 3D bioprinting with digital light processing to generate a vasculature lumen network surrounded by embedded human astrocytes. The lumens are then cellularized with primary human brain microvascular endothelial cells and pericytes. To initiate mechanotransduction pathways and complete maturation, vascular structures are continuously perfused for 7 days. Constructs are validated for complete endothelialization with viability dyes prior to functional assessments that include barrier integrity (permeability) and immune-endothelial interactions. This MPS has applications for the study of novel therapeutics, toxins, and elucidating mechanisms of pathophysiology.
Collapse
Affiliation(s)
- Kalpani N. Udeni Galpayage Dona
- Department of Pathology & Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (K.N.U.G.D.)
| | - Servio H. Ramirez
- Department of Pathology & Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (K.N.U.G.D.)
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
- The Shriners Hospitals Pediatric Research Center, Philadelphia, PA 19140, USA
| | - Allison M. Andrews
- Department of Pathology & Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA; (K.N.U.G.D.)
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
38
|
Bao Y, Lu W. Targeting cerebral diseases with enhanced delivery of therapeutic proteins across the blood-brain barrier. Expert Opin Drug Deliv 2023; 20:1681-1698. [PMID: 36945117 DOI: 10.1080/17425247.2023.2193390] [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: 01/14/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023]
Abstract
INTRODUCTION Cerebral diseases have been threatening public physical and psychological health in the recent years. With the existence of the blood-brain barrier (BBB), it is particularly hard for therapeutic proteins like peptides, enzymes, antibodies, etc. to enter the central nervous system (CNS) and function in diagnosis and treatment in cerebral diseases. Fortunately, the past decade has witnessed some emerging strategies of delivering macromolecular therapeutic proteins across the BBB. AREAS COVERED Based on the structure, functions, and substances transport mechanisms, various enhanced delivery strategies of therapeutic proteins were reviewed, categorized by molecule-mediated delivery strategies, carrier-mediated delivery strategies, and other delivery strategies. EXPERT OPINION As for molecule-mediated delivery strategies, development of genetic engineering technology, optimization of protein expression and purification techniques, and mature of quality control systems all help to realize large-scale production of recombinant antibodies, making it possible to apply to the clinical practice. In terms of carrier-mediated delivery strategies and others, although nano-carriers/adeno-associated virus (AAV) are also promising candidates for delivering therapeutic proteins or genes across the BBB, some issues still remain to be further investigated, including safety concerns related to applied materials, large-scale production costs, quality control standards, combination therapies with auxiliary delivery strategies like focused ultrasound, etc.
Collapse
Affiliation(s)
- Yanning Bao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai, China
- Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, China
- Department of Research and Development, Shanghai Tayzen PharmLab Co., Ltd. Lingang of Shanghai, China
| |
Collapse
|
39
|
Rauti R, Navok S, Biran D, Tadmor K, Leichtmann-Bardoogo Y, Ron EZ, Maoz BM. Insight on Bacterial Newborn Meningitis Using a Neurovascular-Unit-on-a-Chip. Microbiol Spectr 2023; 11:e0123323. [PMID: 37222614 PMCID: PMC10269748 DOI: 10.1128/spectrum.01233-23] [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] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023] Open
Abstract
Understanding the pathogenesis of bacterial infections is critical for combatting them. For some infections, animal models are inadequate and functional genomic studies are not possible. One example is bacterial meningitis, a life-threatening infection with high mortality and morbidity. Here, we used the newly developed, physiologically relevant, organ-on-a-chip platform integrating the endothelium with neurons, closely mimicking in vivo conditions. Using high-magnification microscopy, permeability measurements, electrophysiological recordings, and immunofluorescence staining, we studied the dynamic by which the pathogens cross the blood-brain barrier and damage the neurons. Our work opens up possibilities for performing large-scale screens with bacterial mutant libraries for identifying the virulence genes involved in meningitis and determining the role of these genes, including various capsule types, in the infection process. These data are essential for understanding and therapy of bacterial meningitis. Moreover, our system offers possibilities for the study of additional infections-bacterial, fungal, and viral. IMPORTANCE The interactions of newborn meningitis (NBM) with the neurovascular unit are very complex and are hard to study. This work presents a new platform to study NBM in a system that enables monitoring of multicellular interactions and identifies processes that were not observed before.
Collapse
Affiliation(s)
- Rossana Rauti
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Sharon Navok
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Dvora Biran
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Keshet Tadmor
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | | | - Eliora Z. Ron
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Ben M. Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
40
|
Soldati S, Bär A, Vladymyrov M, Glavin D, McGrath JL, Gosselet F, Nishihara H, Goelz S, Engelhardt B. High levels of endothelial ICAM-1 prohibit natalizumab mediated abrogation of CD4 + T cell arrest on the inflamed BBB under flow in vitro. J Neuroinflammation 2023; 20:123. [PMID: 37221552 PMCID: PMC10204262 DOI: 10.1186/s12974-023-02797-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/02/2023] [Indexed: 05/25/2023] Open
Abstract
INTRODUCTION The humanized anti-α4 integrin blocking antibody natalizumab (NTZ) is an effective treatment for relapsing-remitting multiple sclerosis (RRMS) that is associated with the risk of progressive multifocal leukoencephalopathy (PML). While extended interval dosing (EID) of NTZ reduces the risk for PML, the minimal dose of NTZ required to maintain its therapeutic efficacy remains unknown. OBJECTIVE Here we aimed to identify the minimal NTZ concentration required to inhibit the arrest of human effector/memory CD4+ T cell subsets or of PBMCs to the blood-brain barrier (BBB) under physiological flow in vitro. RESULTS Making use of three different human in vitro BBB models and in vitro live-cell imaging we observed that NTZ mediated inhibition of α4-integrins failed to abrogate T cell arrest to the inflamed BBB under physiological flow. Complete inhibition of shear resistant T cell arrest required additional inhibition of β2-integrins, which correlated with a strong upregulation of endothelial intercellular adhesion molecule (ICAM)-1 on the respective BBB models investigated. Indeed, NTZ mediated inhibition of shear resistant T cell arrest to combinations of immobilized recombinant vascular cell adhesion molecule (VCAM)-1 and ICAM-1 was abrogated in the presence of tenfold higher molar concentrations of ICAM-1 over VCAM-1. Also, monovalent NTZ was less potent than bivalent NTZ in inhibiting T cell arrest to VCAM-1 under physiological flow. In accordance with our previous observations ICAM-1 but not VCAM-1 mediated T cell crawling against the direction of flow. CONCLUSION Taken together, our in vitro observations show that high levels of endothelial ICAM-1 abrogate NTZ mediated inhibition of T cell interaction with the BBB. EID of NTZ in MS patients may thus require consideration of the inflammatory status of the BBB as high levels of ICAM-1 may provide an alternative molecular cue allowing for pathogenic T cell entry into the CNS in the presence of NTZ.
Collapse
Affiliation(s)
- Sasha Soldati
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012 Bern, Switzerland
| | - Alexander Bär
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012 Bern, Switzerland
| | - Mykhailo Vladymyrov
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012 Bern, Switzerland
| | - Dale Glavin
- Department of Biomedical Engineering, University of Rochester, Rochester, NY USA
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY USA
| | - Fabien Gosselet
- Blood-Brain Barrier Laboratory, University of Artois, Lens, France
| | - Hideaki Nishihara
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012 Bern, Switzerland
- Present Address: Department of Neurotherapeutics, Yamaguchi University, Yamaguchi, Japan
| | | | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012 Bern, Switzerland
| |
Collapse
|
41
|
Salimi L, Seyedaghamiri F, Karimipour M, Mobarak H, Mardi N, Taghavi M, Rahbarghazi R. Physiological and pathological consequences of exosomes at the blood-brain-barrier interface. Cell Commun Signal 2023; 21:118. [PMID: 37208741 DOI: 10.1186/s12964-023-01142-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/22/2023] [Indexed: 05/21/2023] Open
Abstract
Blood-brain barrier (BBB) interface with multicellular structure controls strictly the entry of varied circulating macromolecules from the blood-facing surface into the brain parenchyma. Under several pathological conditions within the central nervous system, the integrity of the BBB interface is disrupted due to the abnormal crosstalk between the cellular constituents and the recruitment of inflammatory cells. Exosomes (Exos) are nano-sized extracellular vesicles with diverse therapeutic outcomes. These particles transfer a plethora of signaling molecules with the potential to modulate target cell behavior in a paracrine manner. Here, in the current review article, the therapeutic properties of Exos and their potential in the alleviation of compromised BBB structure were discussed. Video Abstract.
Collapse
Affiliation(s)
- Leila Salimi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemehsadat Seyedaghamiri
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Karimipour
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Halimeh Mobarak
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Narges Mardi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Taghavi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
42
|
Qiao R, Fu C, Forgham H, Javed I, Huang X, Zhu J, Whittaker AK, Davis TP. Magnetic Iron Oxide Nanoparticles for Brain Imaging and Drug Delivery. Adv Drug Deliv Rev 2023; 197:114822. [PMID: 37086918 DOI: 10.1016/j.addr.2023.114822] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 03/14/2023] [Accepted: 04/09/2023] [Indexed: 04/24/2023]
Abstract
Central nervous system (CNS) disorders affect as many as 1.5 billion people globally. The limited delivery of most imaging and therapeutic agents into the brain is a major challenge for treatment of CNS disorders. With the advent of nanotechnologies, controlled delivery of drugs with nanoparticles holds great promise in CNS disorders for overcoming the blood-brain barrier (BBB) and improving delivery efficacy. In recent years, magnetic iron oxide nanoparticles (MIONPs) have stood out as a promising theranostic nanoplatform for brain imaging and drug delivery as they possess unique physical properties and biodegradable characteristics. In this review, we summarize the recent advances in MIONP-based platforms as imaging and drug delivery agents for brain diseases. We firstly introduce the methods of synthesis and surface functionalization of MIONPs with emphasis on the inclusion of biocompatible polymers that allow for the addition of tailored physicochemical properties. We then discuss the recent advances in in vivo imaging and drug delivery applications using MIONPs. Finally, we present a perspective on the remaining challenges and possible future directions for MIONP-based brain delivery systems.
Collapse
Affiliation(s)
- Ruirui Qiao
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Changkui Fu
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Helen Forgham
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Ibrahim Javed
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Xumin Huang
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jiayuan Zhu
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andrew K Whittaker
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Thomas P Davis
- Australian Institute of Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
43
|
Straehla JP, Reardon DA, Wen PY, Agar NYR. The Blood-Brain Barrier: Implications for Experimental Cancer Therapeutics. ANNUAL REVIEW OF CANCER BIOLOGY 2023; 7:265-289. [PMID: 38323268 PMCID: PMC10846865 DOI: 10.1146/annurev-cancerbio-061421-040433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
The blood-brain barrier is critically important for the treatment of both primary and metastatic cancers of the central nervous system (CNS). Clinical outcomes for patients with primary CNS tumors are poor and have not significantly improved in decades. As treatments for patients with extracranial solid tumors improve, the incidence of CNS metastases is on the rise due to suboptimal CNS exposure of otherwise systemically active agents. Despite state-of-the art surgical care and increasingly precise radiation therapy, clinical progress is limited by the ability to deliver an effective dose of a therapeutic agent to all cancerous cells. Given the tremendous heterogeneity of CNS cancers, both across cancer subtypes and within a single tumor, and the range of diverse therapies under investigation, a nuanced examination of CNS drug exposure is needed. With a shared goal, common vocabulary, and interdisciplinary collaboration, the field is poised for renewed progress in the treatment of CNS cancers.
Collapse
Affiliation(s)
- Joelle P Straehla
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts, USA
| | - David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
44
|
Hernandez L, Ward LJ, Arefin S, Barany P, Wennberg L, Söderberg M, Bruno S, Cantaluppi V, Stenvinkel P, Kublickiene K. Blood–Brain Barrier Biomarkers before and after Kidney Transplantation. Int J Mol Sci 2023; 24:ijms24076628. [PMID: 37047601 PMCID: PMC10095132 DOI: 10.3390/ijms24076628] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Kidney transplantation (KT) may improve the neurological status of chronic kidney disease (CKD) patients, reflected by the altered levels of circulating BBB-specific biomarkers. This study compares the levels of neuron specific enolase (NSE), brain-derived neurotrophic factor (BDNF), neurofilament light chain (NfL), and circulating plasma extracellular vesicles (EVs) in kidney-failure patients before KT and at a two-year follow up. Using ELISA, NSE, BDNF, and NfL levels were measured in the plasma of 74 living-donor KT patients. Plasma EVs were isolated with ultracentrifugation, and characterized for concentration/size and surface protein expression using flow cytometry from a subset of 25 patients. Lower NSE levels, and higher BDNF and NfL were observed at the two-year follow-up compared to the baseline (p < 0.05). Male patients had significantly higher BDNF levels compared to those of females. BBB biomarkers correlated with the baseline lipid profile and with glucose, vitamin D, and inflammation markers after KT. BBB surrogate marker changes in the microcirculation of early vascular aging phenotype patients with calcification and/or fibrosis were observed only in NSE and BDNF. CD31+ microparticles from endothelial cells expressing inflammatory markers such as CD40 and integrins were significantly reduced after KT. KT may, thus, improve the neurological status of CKD patients, as reflected by changes in BBB-specific biomarkers.
Collapse
Affiliation(s)
- Leah Hernandez
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Liam J. Ward
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, 587 58 Linköping, Sweden
| | - Samsul Arefin
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Peter Barany
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Lars Wennberg
- Department of Transplantation Surgery, Karolinska University Hospital, 141 86 Stockholm, Sweden
| | - Magnus Söderberg
- Department of Pathology, Clinical Pharmacology and Safety Sciences, R&D AstraZeneca, 431 83 Gothenburg, Sweden
| | - Stefania Bruno
- Department of Medical Sciences, University of Torino, 10124 Torino, Italy
| | - Vincenzo Cantaluppi
- Nephrology and Kidney Transplant Unit, Department of Translational Medicine (DIMET), University of Piemonte Orientale (UPO), “Maggiore della Carita” University Hospital, 28100 Novara, Italy
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Karolina Kublickiene
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
45
|
Sepehrinezhad A, Stolze Larsen F, Ashayeri Ahmadabad R, Shahbazi A, Sahab Negah S. The Glymphatic System May Play a Vital Role in the Pathogenesis of Hepatic Encephalopathy: A Narrative Review. Cells 2023; 12:cells12070979. [PMID: 37048052 PMCID: PMC10093707 DOI: 10.3390/cells12070979] [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: 10/19/2022] [Revised: 02/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Hepatic encephalopathy (HE) is a neurological complication of liver disease resulting in cognitive, psychiatric, and motor symptoms. Although hyperammonemia is a key factor in the pathogenesis of HE, several other factors have recently been discovered. Among these, the impairment of a highly organized perivascular network known as the glymphatic pathway seems to be involved in the progression of some neurological complications due to the accumulation of misfolded proteins and waste substances in the brain interstitial fluids (ISF). The glymphatic system plays an important role in the clearance of brain metabolic derivatives and prevents aggregation of neurotoxic agents in the brain ISF. Impairment of it will result in aggravated accumulation of neurotoxic agents in the brain ISF. This could also be the case in patients with liver failure complicated by HE. Indeed, accumulation of some metabolic by-products and agents such as ammonia, glutamine, glutamate, and aromatic amino acids has been reported in the human brain ISF using microdialysis technique is attributed to worsening of HE and correlates with brain edema. Furthermore, it has been reported that the glymphatic system is impaired in the olfactory bulb, prefrontal cortex, and hippocampus in an experimental model of HE. In this review, we discuss different factors that may affect the function of the glymphatic pathways and how these changes may be involved in HE.
Collapse
Affiliation(s)
- Ali Sepehrinezhad
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad 9919191778, Iran
| | - Fin Stolze Larsen
- Department of Gastroenterology and Hepatology, Rigshospitalet, Copenhagen University Hospital, 999017 Copenhagen, Denmark
| | | | - Ali Shahbazi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad 9919191778, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1449614535, Iran
| |
Collapse
|
46
|
Gonzales-Aloy E, Ahmed-Cox A, Tsoli M, Ziegler DS, Kavallaris M. From cells to organoids: The evolution of blood-brain barrier technology for modelling drug delivery in brain cancer. Adv Drug Deliv Rev 2023; 196:114777. [PMID: 36931346 DOI: 10.1016/j.addr.2023.114777] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/13/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
Brain cancer remains the deadliest cancer. The blood-brain barrier (BBB) is impenetrable to most drugs and is a complex 3D network of multiple cell types including endothelial cells, astrocytes, and pericytes. In brain cancers, the BBB becomes disrupted during tumor progression and forms the blood-brain tumor barrier (BBTB). To advance therapeutic development, there is a critical need for physiologically relevant BBB in vitro models. 3D cell systems are emerging as valuable preclinical models to accelerate discoveries for diseases. Given the versatility and capability of 3D cell models, their potential for modelling the BBB and BBTB is reviewed. Technological advances of BBB models and challenges of in vitro modelling the BBTB, and application of these models as tools for assessing therapeutics and nano drug delivery, are discussed. Quantitative, in vitro BBB models that are predictive of effective brain cancer therapies will be invaluable for accelerating advancing new treatments to the clinic.
Collapse
Affiliation(s)
- Estrella Gonzales-Aloy
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; Australian Center for NanoMedicine, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia
| | - Aria Ahmed-Cox
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; Australian Center for NanoMedicine, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia; Katharina Gaus Light Microscopy Facility, Mark Wainright Analytical Center, UNSW Sydney, NSW, Australia
| | - Maria Tsoli
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia
| | - David S Ziegler
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia; Kids Cancer Center, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Center, UNSW Sydney, NSW, Australia; Australian Center for NanoMedicine, UNSW Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, NSW, Australia; UNSW RNA Institute, UNSW Sydney, NSW, Australia.
| |
Collapse
|
47
|
Contreras EG, Klämbt C. The Drosophila blood-brain barrier emerges as a model for understanding human brain diseases. Neurobiol Dis 2023; 180:106071. [PMID: 36898613 DOI: 10.1016/j.nbd.2023.106071] [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: 12/29/2022] [Revised: 02/24/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
The accurate regulation of the microenvironment within the nervous system is one of the key features characterizing complex organisms. To this end, neural tissue has to be physically separated from circulation, but at the same time, mechanisms must be in place to allow controlled transport of nutrients and macromolecules into and out of the brain. These roles are executed by cells of the blood-brain barrier (BBB) found at the interface of circulation and neural tissue. BBB dysfunction is observed in several neurological diseases in human. Although this can be considered as a consequence of diseases, strong evidence supports the notion that BBB dysfunction can promote the progression of brain disorders. In this review, we compile the recent evidence describing the contribution of the Drosophila BBB to the further understanding of brain disease features in human patients. We discuss the function of the Drosophila BBB during infection and inflammation, drug clearance and addictions, sleep, chronic neurodegenerative disorders and epilepsy. In summary, this evidence suggests that the fruit fly, Drosophila melanogaster, can be successfully employed as a model to disentangle mechanisms underlying human diseases.
Collapse
Affiliation(s)
- Esteban G Contreras
- University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, Münster, Germany.
| | - Christian Klämbt
- University of Münster, Institute of Neuro- and Behavioral Biology, Badestr. 9, Münster, Germany.
| |
Collapse
|
48
|
Experimental Models of In Vitro Blood-Brain Barrier for CNS Drug Delivery: An Evolutionary Perspective. Int J Mol Sci 2023; 24:ijms24032710. [PMID: 36769032 PMCID: PMC9916529 DOI: 10.3390/ijms24032710] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Central nervous system (CNS) disorders represent one of the leading causes of global health burden. Nonetheless, new therapies approved against these disorders are among the lowest compared to their counterparts. The absence of reliable and efficient in vitro blood-brain barrier (BBB) models resembling in vivo barrier properties stands out as a significant roadblock in developing successful therapy for CNS disorders. Therefore, advancement in the creation of robust and sensitive in vitro BBB models for drug screening might allow us to expedite neurological drug development. This review discusses the major in vitro BBB models developed as of now for exploring the barrier properties of the cerebral vasculature. Our main focus is describing existing in vitro models, including the 2D transwell models covering both single-layer and co-culture models, 3D organoid models, and microfluidic models with their construction, permeability measurement, applications, and limitations. Although microfluidic models are better at recapitulating the in vivo properties of BBB than other models, significant gaps still exist for their use in predicting the performance of neurotherapeutics. However, this comprehensive account of in vitro BBB models can be useful for researchers to create improved models in the future.
Collapse
|
49
|
Spitz S, Ko E, Ertl P, Kamm RD. How Organ-on-a-Chip Technology Can Assist in Studying the Role of the Glymphatic System in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:2171. [PMID: 36768495 PMCID: PMC9916687 DOI: 10.3390/ijms24032171] [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: 12/13/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
The lack of a conventional lymphatic system that permeates throughout the entire human brain has encouraged the identification and study of alternative clearance routes within the cerebrum. In 2012, the concept of the glymphatic system, a perivascular network that fluidically connects the cerebrospinal fluid to the lymphatic vessels within the meninges via the interstitium, emerged. Although its exact mode of action has not yet been fully characterized, the key underlying processes that govern solute transport and waste clearance have been identified. This review briefly describes the perivascular glial-dependent clearance system and elucidates its fundamental role in neurodegenerative diseases. The current knowledge of the glymphatic system is based almost exclusively on animal-based measurements, but these face certain limitations inherent to in vivo experiments. Recent advances in organ-on-a-chip technology are discussed to demonstrate the technology's ability to provide alternative human-based in vitro research models. Herein, the specific focus is on how current microfluidic-based in vitro models of the neurovascular system and neurodegenerative diseases might be employed to (i) gain a deeper understanding of the role and function of the glymphatic system and (ii) to identify new opportunities for pharmacological intervention.
Collapse
Affiliation(s)
- Sarah Spitz
- Faculty of Technical Chemistry, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eunkyung Ko
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peter Ertl
- Faculty of Technical Chemistry, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria
| | - Roger D. Kamm
- Department of Mechanical Engineering and Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
50
|
Gribkoff VK, Kaczmarek LK. The Difficult Path to the Discovery of Novel Treatments in Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2023; 30:255-285. [PMID: 36928854 PMCID: PMC10599454 DOI: 10.1007/978-3-031-21054-9_11] [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] [Indexed: 03/18/2023]
Abstract
CNS diseases, including psychiatric disorders, represent a significant opportunity for the discovery and development of new drugs and therapeutic treatments with the potential to have a significant impact on human health. CNS diseases, however, present particular challenges to therapeutic discovery efforts, and psychiatric diseases/disorders may be among the most difficult. With specific exceptions such as psychostimulants for ADHD, a large number of psychiatric patients are resistant to existing treatments. In addition, clinicians have no way of knowing which psychiatric patients will respond to which drugs. By definition, psychiatric diagnoses are syndromal in nature; determinations of efficacy are often self-reported, and drug discovery is largely model-based. While such models of psychiatric disease are amenable to screening for new drugs, whether cellular or whole-animal based, they have only modest face validity and, more importantly, predictive validity. Multiple academic, pharmaceutical industry, and government agencies are dedicated to the translation of new findings about the neurobiology of major psychiatric disorders into the discovery and advancement of novel therapies. The collaboration of these agencies provide a pathway for developing new therapeutics. These efforts will be greatly helped by recent advances in understanding the genetic bases of psychiatric disorders, the ongoing search for diagnostic and therapy-responsive biomarkers, and the validation of new animal models.
Collapse
Affiliation(s)
- Valentin K Gribkoff
- Department of Internal Medicine, Section on Endocrinology, Yale University School of Medicine, New Haven, CT, USA.
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|