1
|
Sánchez SV, Otavalo GN, Gazeau F, Silva AKA, Morales JO. Intranasal delivery of extracellular vesicles: A promising new approach for treating neurological and respiratory disorders. J Control Release 2025; 379:489-523. [PMID: 39800240 DOI: 10.1016/j.jconrel.2025.01.018] [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: 09/13/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Extracellular vesicles (EVs) are membrane vesicles secreted by all types of cells, including bacteria, animals, and plants. These vesicles contain proteins, nucleic acids, and lipids from their parent cells and can transfer these components between cells. EVs have attracted attention for their potential use in diagnosis and therapy due to their natural properties, such as low immunogenicity, high biocompatibility, and ability to cross the blood-brain barrier. They can also be engineered to carry therapeutic molecules. EVs can be delivered via various routes. The intranasal route is particularly advantageous for delivering them to the central nervous system, making it a promising approach for treating neurological disorders. SCOPE OF REVIEW This review delves into the promising potential of intranasally administered EVs-based therapies for various medical conditions, with a particular focus on those affecting the brain and central nervous system. Additionally, the potential use of these therapies for pulmonary conditions, cancer, and allergies is examined, offering a hopeful outlook for the future of medical treatments. MAJOR CONCLUSIONS The intranasal administration of EVs offers significant advantages over other delivery methods. By directly delivering EVs to the brain, specifically targeting areas that have been injured, this administration proves to be highly efficient and effective, providing reassurance about the progress in medical treatments. Intranasal delivery is not limited to brain-related conditions. It can also benefit other organs like the lungs and stimulate a mucosal immune response against various pathogens due to the highly vascularized nature of the nasal cavity and airways. Moreover, it has the added benefit of minimizing toxicity to non-targeted organs and allows the EVs to remain longer in the body. As a result, there is a growing emphasis on conducting clinical trials for intranasal administration of EVs, particularly in treating respiratory tract pathologies such as coronavirus disease.
Collapse
Affiliation(s)
- Sofía V Sánchez
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile; Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile
| | - Gabriela N Otavalo
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile; Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile
| | - Florence Gazeau
- Université Paris Cité, CNRS UMR8175, INSERM U1334, Laboratory NABI (Nanomédecine, Biologie Extracellulaire, Intégratome et Innovations en santé), Paris, France
| | - Amanda K A Silva
- Université Paris Cité, CNRS UMR8175, INSERM U1334, Laboratory NABI (Nanomédecine, Biologie Extracellulaire, Intégratome et Innovations en santé), Paris, France
| | - Javier O Morales
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile; Center of New Drugs for Hypertension and Heart Failure (CENDHY), Santiago, Chile.
| |
Collapse
|
2
|
Madhu LN, Kodali M, Upadhya R, Rao S, Somayaji Y, Attaluri S, Shuai B, Kirmani M, Gupta S, Maness N, Rao X, Cai JJ, Shetty AK. Extracellular vesicles from human-induced pluripotent stem cell-derived neural stem cells alleviate proinflammatory cascades within disease-associated microglia in Alzheimer's disease. J Extracell Vesicles 2024; 13:e12519. [PMID: 39499013 PMCID: PMC11536387 DOI: 10.1002/jev2.12519] [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: 03/26/2024] [Revised: 08/17/2024] [Accepted: 08/28/2024] [Indexed: 11/07/2024] Open
Abstract
As current treatments for Alzheimer's disease (AD) lack disease-modifying interventions, novel therapies capable of restraining AD progression and maintaining better brain function have great significance. Anti-inflammatory extracellular vesicles (EVs) derived from human induced pluripotent stem cell (hiPSC)-derived neural stem cells (NSCs) hold promise as a disease-modifying biologic for AD. This study directly addressed this issue by examining the effects of intranasal (IN) administrations of hiPSC-NSC-EVs in 3-month-old 5xFAD mice. IN administered hiPSC-NSC-EVs incorporated into microglia, including plaque-associated microglia, and encountered astrocyte soma and processes in the brain. Single-cell RNA sequencing revealed transcriptomic changes indicative of diminished activation of microglia and astrocytes. Multiple genes linked to disease-associated microglia, NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3)-inflammasome and interferon-1 (IFN-1) signalling displayed reduced expression in microglia. Adding hiPSC-NSC-EVs to cultured human microglia challenged with amyloid-beta oligomers resulted in similar effects. Astrocytes also displayed reduced expression of genes linked to IFN-1 and interleukin-6 signalling. Furthermore, the modulatory effects of hiPSC-NSC-EVs on microglia in the hippocampus persisted 2 months post-EV treatment without impacting their phagocytosis function. Such effects were evidenced by reductions in microglial clusters and inflammasome complexes, concentrations of mediators, and end products of NLRP3 inflammasome activation, the expression of genes and/or proteins involved in the activation of p38/mitogen-activated protein kinase and IFN-1 signalling, and unaltered phagocytosis function. The extent of astrocyte hypertrophy, amyloid-beta plaques, and p-tau were also reduced in the hippocampus. Such modulatory effects of hiPSC-NSC-EVs also led to better cognitive and mood function. Thus, early hiPSC-NSC-EV intervention in AD can maintain better brain function by reducing adverse neuroinflammatory signalling cascades, amyloid-beta plaque load, and p-tau. These results reflect the first demonstration of the efficacy of hiPSC-NSC-EVs to restrain neuroinflammatory signalling cascades in an AD model by inducing transcriptomic changes in activated microglia and reactive astrocytes.
Collapse
Affiliation(s)
- Leelavathi N. Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Shama Rao
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Yogish Somayaji
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Maha Kirmani
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Shreyan Gupta
- Department of Veterinary Integrative BiosciencesTexas A&M College of Veterinary Medicine, College StationTexasUSA
| | - Nathaniel Maness
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - Xiaolan Rao
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| | - James J. Cai
- Department of Veterinary Integrative BiosciencesTexas A&M College of Veterinary Medicine, College StationTexasUSA
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, College of MedicineTexas A&M University Health Science Center, College StationTexasUSA
| |
Collapse
|
3
|
Shanmugam I, Radhakrishnan S, Santosh S, Ramnath A, Anil M, Devarajan Y, Maheswaran S, Narayanan V, Pitchaimani A. Emerging role and translational potential of small extracellular vesicles in neuroscience. Life Sci 2024; 355:122987. [PMID: 39151884 DOI: 10.1016/j.lfs.2024.122987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Small extracellular vesicles (sEV) are endogenous lipid-bound membrane vesicles secreted by both prokaryotic and eukaryotic cells into the extracellular environment, performs several biological functions such as cell-cell communication, transfer of proteins, mRNA, and ncRNA to target cells in distant sites. Due to their role in molecular pathogenesis and its potential to deliver biological cargo to target cells, it has become a prominent area of interest in recent research in the field of Neuroscience. However, their role in neurological disorders, like neurodegenerative diseases is more complex and still unaddressed. Thus, this review focuses on the role of sEV in neurodegenerative and neurodevelopmental diseases, including their biogenesis, classification, and pathogenesis, with translational advantages and limitations in the area of neurobiology.
Collapse
Affiliation(s)
- Iswarya Shanmugam
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Sivani Radhakrishnan
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Shradha Santosh
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Akansha Ramnath
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Meghna Anil
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Yogesh Devarajan
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Saravanakumar Maheswaran
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Vaibav Narayanan
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Arunkumar Pitchaimani
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
4
|
Sankarappan K, Shetty AK. Promise of mesenchymal stem cell-derived extracellular vesicles for alleviating subarachnoid hemorrhage-induced brain dysfunction by neuroprotective and antiinflammatory effects. Brain Behav Immun Health 2024; 40:100835. [PMID: 39165307 PMCID: PMC11334735 DOI: 10.1016/j.bbih.2024.100835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 08/22/2024] Open
Abstract
Subarachnoid hemorrhage (SAH), accounting for ∼5% of all strokes, represents a catastrophic subtype of cerebrovascular accident. SAH predominantly results from intracranial aneurysm ruptures and affects ∼30,000 individuals annually in the United States and ∼6 individuals per 100,000 people worldwide. Recent studies have implicated that administering mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) may be beneficial in inducing neuroprotective and antiinflammatory effects following SAH. EVs are nanosized particles bound by a lipid bilayer. MSC-EVs comprise a therapeutic cargo of nucleic acids, lipids, and proteins, having the promise to ease SAH-induced long-term brain impairments. This review evaluated the findings of published studies on the therapeutic efficacy of MSC-EVs in the context of SAH. A growing body of evidence points out the therapeutic potential of MSC-EVs for improving brain function in animal models of SAH. Specifically, studies demonstrated their ability to reduce neuronal apoptosis and neuroinflammation and enhance neurological recovery through neuroprotective and antiinflammatory mechanisms. Such outcomes reported in various studies suggest that MSC-EVs hold great potential as a novel and minimally invasive approach to ameliorate SAH-induced neurological damage and improve patient outcomes. The review also discusses the limitations of EV therapy and the required future research efforts toward harnessing the full potential of MSC-EVs in treating SAH.
Collapse
Affiliation(s)
- Kiran Sankarappan
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, USA
| | - Ashok K. Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, USA
| |
Collapse
|
5
|
Willman J, Kurian AL, Lucke-Wold B. Mechanisms of vascular injury in neurotrauma: A critical review of the literature. World J Meta-Anal 2024; 12:95417. [DOI: 10.13105/wjma.v12.i3.95417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/20/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
One in every two individuals will experience a traumatic brain injury in their lifetime with significant impacts on the global economy and healthcare system each year. Neurovascular injury is a key aspect of neurotrauma to both the brain and the spinal cord and an important avenue of current and future research seeking innovative therapies. In this paper, we discuss primary and secondary neurotrauma, mechanisms of injury, the glymphatic system, repair and recovery. Each of these topics are directly connected to the vasculature of the central nervous system, affecting severity of injury and recovery. Consequently, neurovascular injury in trauma represents a promising target for future therapeutics and innovation.
Collapse
Affiliation(s)
- Jonathan Willman
- College of Medicine, University of Florida, Gainesville, FL 32610, United States
| | - Annu Lisa Kurian
- College of Medicine, Florida State University, Tallahassee, FL 32304, United States
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, United States
| |
Collapse
|
6
|
Zhang Y, Zheng Z, Sun J, Xu S, Wei Y, Ding X, Ding G. The application of mesenchymal stem cells in the treatment of traumatic brain injury: Mechanisms, results, and problems. Histol Histopathol 2024; 39:1109-1131. [PMID: 38353136 DOI: 10.14670/hh-18-716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells that can be derived from a wide variety of human tissues and organs. They can differentiate into a variety of cell types, including osteoblasts, adipocytes, and chondrocytes, and thus show great potential in regenerative medicine. Traumatic brain injury (TBI) is an organic injury to brain tissue with a high rate of disability and death caused by an external impact or concussive force acting directly or indirectly on the head. The current treatment of TBI mainly includes symptomatic, pharmacological, and rehabilitation treatment. Although some efficacy has been achieved, the definitive recovery effect on neural tissue is still limited. Recent studies have shown that MSC therapies are more effective than traditional treatment strategies due to their strong multi-directional differentiation potential, self-renewal capacity, and low immunogenicity and homing properties, thus MSCs are considered to play an important role and are an ideal cell for the treatment of injurious diseases, including TBI. In this paper, we systematically reviewed the role and mechanisms of MSCs and MSC-derived exosomes in the treatment of TBI, thereby providing new insights into the clinical applications of MSCs and MSC-derived exosomes in the treatment of central nervous system disorders.
Collapse
Affiliation(s)
- Ying Zhang
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Zejun Zheng
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Jinmeng Sun
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Shuangshuang Xu
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yanan Wei
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Xiaoling Ding
- Clinical Competency Training Center, Shandong Second Medical University, Weifang, Shandong Province, China.
| | - Gang Ding
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, China.
| |
Collapse
|
7
|
Putthanbut N, Lee JY, Borlongan CV. Extracellular vesicle therapy in neurological disorders. J Biomed Sci 2024; 31:85. [PMID: 39183263 PMCID: PMC11346291 DOI: 10.1186/s12929-024-01075-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024] Open
Abstract
Extracellular vesicles (EVs) are vital for cell-to-cell communication, transferring proteins, lipids, and nucleic acids in various physiological and pathological processes. They play crucial roles in immune modulation and tissue regeneration but are also involved in pathogenic conditions like inflammation and degenerative disorders. EVs have heterogeneous populations and cargo, with numerous subpopulations currently under investigations. EV therapy shows promise in stimulating tissue repair and serving as a drug delivery vehicle, offering advantages over cell therapy, such as ease of engineering and minimal risk of tumorigenesis. However, challenges remain, including inconsistent nomenclature, complex characterization, and underdeveloped large-scale production protocols. This review highlights the recent advances and significance of EVs heterogeneity, emphasizing the need for a better understanding of their roles in disease pathologies to develop tailored EV therapies for clinical applications in neurological disorders.
Collapse
Affiliation(s)
- Napasiri Putthanbut
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Salaya, Thailand
| | - Jea Young Lee
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA
| | - Cesario V Borlongan
- Department of Neurosurgery, Center of Aging and Brain Repair, University of South Florida, Tampa, USA.
| |
Collapse
|
8
|
Kodali M, Madhu LN, Kolla VSV, Attaluri S, Huard C, Somayaji Y, Shuai B, Jordan C, Rao X, Shetty S, Shetty AK. FDA-approved cannabidiol [Epidiolex ®] alleviates Gulf War Illness-linked cognitive and mood dysfunction, hyperalgesia, neuroinflammatory signaling, and declined neurogenesis. Mil Med Res 2024; 11:61. [PMID: 39169440 PMCID: PMC11340098 DOI: 10.1186/s40779-024-00563-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Chronic Gulf War Illness (GWI) is characterized by cognitive and mood impairments, as well as persistent neuroinflammation and oxidative stress. This study aimed to investigate the efficacy of Epidiolex®, a Food and Drug Administration (FDA)-approved cannabidiol (CBD), in improving brain function in a rat model of chronic GWI. METHODS Six months after exposure to low doses of GWI-related chemicals [pyridostigmine bromide, N,N-diethyl-meta-toluamide (DEET), and permethrin (PER)] along with moderate stress, rats with chronic GWI were administered either vehicle (VEH) or CBD (20 mg/kg, oral) for 16 weeks. Neurobehavioral tests were conducted on 11 weeks after treatment initiation to evaluate the performance of rats in tasks related to associative recognition memory, object location memory, pattern separation, and sucrose preference. The effect of CBD on hyperalgesia was also examined. The brain tissues were processed for immunohistochemical and molecular studies following behavioral tests. RESULTS GWI rats treated with VEH exhibited impairments in all cognitive tasks and anhedonia, whereas CBD-treated GWI rats showed improvements in all cognitive tasks and no anhedonia. Additionally, CBD treatment alleviated hyperalgesia in GWI rats. Analysis of hippocampal tissues from VEH-treated rats revealed astrocyte hypertrophy and increased percentages of activated microglia presenting NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) complexes as well as elevated levels of proteins involved in NLRP3 inflammasome activation and Janus kinase/signal transducers and activators of the transcription (JAK/STAT) signaling. Furthermore, there were increased concentrations of proinflammatory and oxidative stress markers along with decreased neurogenesis. In contrast, the hippocampus from CBD-treated GWI rats displayed reduced levels of proteins mediating the activation of NLRP3 inflammasomes and JAK/STAT signaling, normalized concentrations of proinflammatory cytokines and oxidative stress markers, and improved neurogenesis. Notably, CBD treatment did not alter the concentration of endogenous cannabinoid anandamide in the hippocampus. CONCLUSIONS The use of an FDA-approved CBD (Epidiolex®) has been shown to effectively alleviate cognitive and mood impairments as well as hyperalgesia associated with chronic GWI. Importantly, the improvements observed in rats with chronic GWI in this study were attributed to the ability of CBD to significantly suppress signaling pathways that perpetuate chronic neuroinflammation.
Collapse
Affiliation(s)
- Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Venkata Sai Vashishta Kolla
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Charles Huard
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Yogish Somayaji
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Chase Jordan
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Xiaolan Rao
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Sanath Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University Health Science Center School of Medicine, College Station, TX, 77843, USA.
| |
Collapse
|
9
|
Toomajian V, Tundo A, Ural EE, Greeson EM, Contag CH, Makela AV. Magnetic Particle Imaging Reveals that Iron-Labeled Extracellular Vesicles Accumulate in Brains of Mice with Metastases. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30860-30873. [PMID: 38860682 PMCID: PMC11194773 DOI: 10.1021/acsami.4c04920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/12/2024]
Abstract
The incidence of breast cancer remains high worldwide and is associated with a significant risk of metastasis to the brain that can be fatal; this is due, in part, to the inability of therapeutics to cross the blood-brain barrier (BBB). Extracellular vesicles (EVs) have been found to cross the BBB and further have been used to deliver drugs to tumors. EVs from different cell types appear to have different patterns of accumulation and retention as well as the efficiency of bioactive cargo delivery to recipient cells in the body. Engineering EVs as delivery tools to treat brain metastases, therefore, will require an understanding of the timing of EV accumulation and their localization relative to metastatic sites. Magnetic particle imaging (MPI) is a sensitive and quantitative imaging method that directly detects superparamagnetic iron. Here, we demonstrate MPI as a novel tool to characterize EV biodistribution in metastatic disease after labeling EVs with superparamagnetic iron oxide (SPIO) nanoparticles. Iron-labeled EVs (FeEVs) were collected from iron-labeled parental primary 4T1 tumor cells and brain-seeking 4T1BR5 cells, followed by injection into the mice with orthotopic tumors or brain metastases. MPI quantification revealed that FeEVs were retained for longer in orthotopic mammary carcinomas compared to SPIOs. MPI signal due to iron could only be detected in brains of mice bearing brain metastases after injection of FeEVs, but not SPIOs, or FeEVs when mice did not have brain metastases. These findings indicate the potential use of EVs as a therapeutic delivery tool in primary and metastatic tumors.
Collapse
Affiliation(s)
- Victoria
A. Toomajian
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biomedical Engineering, Michigan State
University, East Lansing, Michigan 48824, United States
| | - Anthony Tundo
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Evran E. Ural
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biomedical Engineering, Michigan State
University, East Lansing, Michigan 48824, United States
| | - Emily M. Greeson
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Microbiology, Genetics & Immunology, Michigan State University, East
Lansing, Michigan 48824, United States
| | - Christopher H. Contag
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biomedical Engineering, Michigan State
University, East Lansing, Michigan 48824, United States
- Department
of Microbiology, Genetics & Immunology, Michigan State University, East
Lansing, Michigan 48824, United States
| | - Ashley V. Makela
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
10
|
Fallahi S, Zangbar HS, Farajdokht F, Rahbarghazi R, Ghiasi F, Mohaddes G. Mesenchymal stem cell-derived exosomes improve neurogenesis and cognitive function of mice with methamphetamine addiction: A novel treatment approach. CNS Neurosci Ther 2024; 30:e14719. [PMID: 38783536 PMCID: PMC11116483 DOI: 10.1111/cns.14719] [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: 10/08/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Methamphetamine (METH) is a psychostimulant substance with highly addictive and neurotoxic effects, but no ideal treatment option exists to improve METH-induced neurocognitive deficits. Recently, mesenchymal stem cells (MSCs)-derived exosomes have raised many hopes for treating neurodegenerative sequela of brain disorders. This study aimed to determine the therapeutic potential of MSCs-derived exosomes on cognitive function and neurogenesis of METH-addicted rodents. METHODS Male BALB/c mice were subjected to chronic METH addiction, followed by intravenous administration of bone marrow MSCs-derived exosomes. Then, the spatial memory and recognition memory of animals were assessed by the Barnes maze and the novel object recognition test (NORT). The neurogenesis-related factors, including NeuN and DCX, and the expression of Iba-1, a microglial activation marker, were assessed in the hippocampus by immunofluorescence staining. Also, the expression of inflammatory cytokines, including TNF-α and NF-κB, were evaluated by western blotting. RESULTS The results showed that BMSCs-exosomes improved the time spent in the target quadrant and correct-to-wrong relative time in the Barnes maze. Also, NORT's discrimination index (DI) and recognition index (RI) were improved following exosome therapy. Additionally, exosome therapy significantly increased the expression of NeuN and DCX in the hippocampus while decreasing the expression of inflammatory cytokines, including TNF-α and NF-κB. Besides, BMSC-exosomes down-regulated the expression of Iba-1. CONCLUSION Our findings indicate that BMSC-exosomes mitigated METH-caused cognitive dysfunction by improving neurogenesis and inhibiting neuroinflammation in the hippocampus.
Collapse
Affiliation(s)
- Solmaz Fallahi
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
| | - Hamid Soltani Zangbar
- Department of Neuroscience and Cognition, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Fereshteh Farajdokht
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
- Neurosciences Research CenterTabriz University of Medical SciencesTabrizIran
| | - Reza Rahbarghazi
- Department of Applied Cell Sciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Fariba Ghiasi
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
| | - Gisou Mohaddes
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
- Department of Neuroscience and Cognition, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
- Department of Biomedical EducationCalifornia Health Sciences University, College of Osteopathic MedicineClovisCaliforniaUSA
| |
Collapse
|
11
|
Madhu LN, Kodali M, Upadhya R, Rao S, Shuai B, Somayaji Y, Attaluri S, Kirmani M, Gupta S, Maness N, Rao X, Cai J, Shetty AK. Intranasally Administered EVs from hiPSC-derived NSCs Alter the Transcriptomic Profile of Activated Microglia and Conserve Brain Function in an Alzheimer's Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576313. [PMID: 38293018 PMCID: PMC10827207 DOI: 10.1101/2024.01.18.576313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Antiinflammatory extracellular vesicles (EVs) derived from human induced pluripotent stem cell (hiPSC)-derived neural stem cells (NSCs) hold promise as a disease-modifying biologic for Alzheimer's disease (AD). This study directly addressed this issue by examining the effects of intranasal administrations of hiPSC-NSC-EVs to 3-month-old 5xFAD mice. The EVs were internalized by all microglia, which led to reduced expression of multiple genes associated with disease-associated microglia, inflammasome, and interferon-1 signaling. Furthermore, the effects of hiPSC-NSC-EVs persisted for two months post-treatment in the hippocampus, evident from reduced microglial clusters, inflammasome complexes, and expression of proteins and/or genes linked to the activation of inflammasomes, p38/mitogen-activated protein kinase, and interferon-1 signaling. The amyloid-beta (Aβ) plaques, Aβ-42, and phosphorylated-tau concentrations were also diminished, leading to better cognitive and mood function in 5xFAD mice. Thus, early intervention with hiPSC-NSC-EVs in AD may help maintain better brain function by restraining the progression of adverse neuroinflammatory signaling cascades.
Collapse
|
12
|
Ashique S, Pal R, Sharma H, Mishra N, Garg A. Unraveling the Emerging Niche Role of Extracellular Vesicles (EVs) in Traumatic Brain Injury (TBI). CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1357-1370. [PMID: 38351688 DOI: 10.2174/0118715273288155240201065041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 09/12/2024]
Abstract
Extracellular vesicles or exosomes, often known as EVs, have acquired significant attention in the investigations of traumatic brain injury (TBI) and have a distinct advantage in actively researching the fundamental mechanisms underlying various clinical symptoms and diagnosing the wide range of traumatic brain injury cases. The mesenchymal stem cells (MSCs) can produce and release exosomes, which offer therapeutic benefits. Exosomes are tiny membranous vesicles produced by various cellular entities originating from endosomes. Several studies have reported that administering MSC-derived exosomes through intravenous infusions improves neurological recovery and promotes neuroplasticity in rats with traumatic brain damage. The therapeutic advantages of exosomes can be attributed to the microRNAs (miRNAs), which are small non-coding regulatory RNAs that significantly impact the regulation of posttranscriptional genes. Exosome-based therapies, which do not involve cells, have lately gained interest as a potential breakthrough in enhancing neuroplasticity and accelerating neurological recovery for various brain injuries and neurodegenerative diseases. This article explores the benefits and drawbacks of exosome treatment for traumatic brain injury while emphasizing the latest advancements in this field with clinical significance.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutical Science, Pandaveswar School of Pharmacy, Pandaveswar, West Bengal 713378, India
| | - Radheshyam Pal
- Department of Pharmaceutical Science, Pandaveswar School of Pharmacy, Pandaveswar, West Bengal 713378, India
| | - Himanshu Sharma
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad (UP) 244001, India
| | - Neeraj Mishra
- Amity Institute of Pharmacy, Amity University Gwalior 474005, Madhya Pradesh, India
| | - Ashish Garg
- Guru Ramdas Khalsa Institute of Science and Technology, Pharmacy, Jabalpur, M.P. 483001, India
| |
Collapse
|
13
|
Ayyubova G, Kodali M, Upadhya R, Madhu LN, Attaluri S, Somayaji Y, Shuai B, Rao S, Shankar G, Shetty AK. Extracellular vesicles from hiPSC-NSCs can prevent peripheral inflammation-induced cognitive dysfunction with inflammasome inhibition and improved neurogenesis in the hippocampus. J Neuroinflammation 2023; 20:297. [PMID: 38087314 PMCID: PMC10717852 DOI: 10.1186/s12974-023-02971-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Extracellular vesicles (EVs) released by human induced pluripotent stem cell-derived neural stem cells (hiPSC-NSCs) are enriched with miRNAs and proteins capable of mediating robust antiinflammatory activity. The lack of tumorigenic and immunogenic properties and ability to permeate the entire brain to incorporate into microglia following intranasal (IN) administrations makes them an attractive biologic for curtailing chronic neuroinflammation in neurodegenerative disorders. We tested the hypothesis that IN administrations of hiPSC-NSC-EVs can alleviate chronic neuroinflammation and cognitive impairments induced by the peripheral lipopolysaccharide (LPS) challenge. Adult male, C57BL/6J mice received intraperitoneal injections of LPS (0.75 mg/kg) for seven consecutive days. Then, the mice received either vehicle (VEH) or hiPSC-NSC-EVs (~ 10 × 109 EVs/administration, thrice over 6 days). A month later, mice in all groups were investigated for cognitive function with behavioral tests and euthanized for histological and biochemical studies. Mice receiving VEH after LPS displayed deficits in associative recognition memory, temporal pattern processing, and pattern separation. Such impairments were associated with an increased incidence of activated microglia presenting NOD-, LRR-, and pyrin domain containing 3 (NLRP3) inflammasomes, elevated levels of NLRP3 inflammasome mediators and end products, and decreased neurogenesis in the hippocampus. In contrast, the various cognitive measures in mice receiving hiPSC-NSC-EVs after LPS were closer to naive mice. Significantly, these mice displayed diminished microglial activation, NLRP3 inflammasomes, proinflammatory cytokines, and a level of neurogenesis matching age-matched naïve controls. Thus, IN administrations of hiPSC-NSC-EVs are an efficacious approach to reducing chronic neuroinflammation-induced cognitive impairments.
Collapse
Affiliation(s)
- Gunel Ayyubova
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Maheedhar Kodali
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Raghavendra Upadhya
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Yogish Somayaji
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Bing Shuai
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Shama Rao
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Goutham Shankar
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, School of Medicine, Texas A&M Health Science Center, 1114 TAMU, 206 Olsen Boulevard, College Station, TX, 77843, USA.
| |
Collapse
|
14
|
Zanirati G, Shetty PA, Shetty AK. Neural stem cells persist to generate new neurons in the hippocampus of adult and aged human brain - Fiction or accurate? Ageing Res Rev 2023; 92:102133. [PMID: 38000512 PMCID: PMC10843673 DOI: 10.1016/j.arr.2023.102133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 11/18/2023] [Indexed: 11/26/2023]
Abstract
Adult neurogenesis, comprising the generation, differentiation and integration of new neurons in the mature brain, has emerged as a dynamic area of research over decades. The discovery of adult neurogenesis was a paradigm shift to comprehend mechanisms underlying brain plasticity, cognitive function, and neurological disorders. This review deliberates significant findings from articles published over four decades on adult neurogenesis, highlighting key milestones, methodological advances, and controversies that have shaped our comprehension of the phenomenon of adult neurogenesis. Early skepticism gave way to a rich body of evidence via various reliable approaches. Studies on neurogenic niches, microenvironmental factors, molecular regulators, and functional implications have uncovered the involvement of adult neurogenesis in learning, memory, mood, and even neurological and neurodegenerative conditions. Despite significant progress, several questions still need to be answered, including the exact contributions of new neurons to brain function, their integration into existing circuits, and the impact of enhancing adult neurogenesis in the human hippocampus. While the existence of robust neurogenesis in the adult and aged human hippocampus is yet to be confirmed, this review highlights evidence from a significant number of studies supporting the persistence of hippocampal neurogenesis during adulthood and aging in humans, including in some neurological conditions, such as epilepsy and Alzheimer's disease. Nonetheless, additional large-scale studies using single cell-RNA-seq, single nucleus-RNA-seq, and spatial transcriptomics are critical to validate the presence and contribution of hippocampal neurogenesis in the pathophysiology of various neurological and neurodegenerative conditions at different stages of the disease. There is also a need to develop standardized protocols for analyzing postmortem hippocampal tissues for cellular and molecular analyses.
Collapse
Affiliation(s)
- Gabriele Zanirati
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, RS, Brazil
| | - Padmashri A Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA; Department of Psychiatry, Foster School of Medicine, Texas Tech Health Science Center, El Paso, TX, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M University School of Medicine, College Station, TX, USA.
| |
Collapse
|
15
|
Shelash Al-Hawary SI, Yahya Ali A, Mustafa YF, Margiana R, Maksuda Ilyasovna S, Ramadan MF, Almalki SG, Alwave M, Alkhayyat S, Alsalamy A. The microRNAs (miRs) overexpressing mesenchymal stem cells (MSCs) therapy in neurological disorders; hope or hype. Biotechnol Prog 2023; 39:e3383. [PMID: 37642165 DOI: 10.1002/btpr.3383] [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: 06/25/2023] [Revised: 07/30/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023]
Abstract
Altered expression of multiple miRNAs was found to be extensively involved in the pathogenesis of different neurological disorders including Alzheimer's disease, Parkinson's disease, stroke, epilepsy, multiple sclerosis, amyotrophic lateral sclerosis, and Huntington's disease. One of the biggest concerns within gene-based therapy is the delivery of the therapeutic microRNAs to the intended place, which is obligated to surpass the biological barriers without undergoing degradation in the bloodstream or renal excretion. Hence, the delivery of modified and unmodified miRNA molecules using excellent vehicles is required. In this light, mesenchymal stem cells (MSCs) have attracted increasing attention. The MSCs can be genetically modified to express or overexpress a particular microRNA aimed with promote neurogenesis and neuroprotection. The current review has focused on the therapeutic capabilities of microRNAs-overexpressing MSCs to ameliorate functional deficits in neurological conditions.
Collapse
Affiliation(s)
| | - Anas Yahya Ali
- Department of Nursing, Al-maarif University College, Ramadi, Al-Anbar, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | | | | | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Saudi Arabia
| | - Marim Alwave
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Safa Alkhayyat
- College of Pharmacy, The Islamic University, Najaf, Iraq
| | - Ali Alsalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq
| |
Collapse
|