1
|
Goldschmidt-Clermont PJ, Khan A, Jimsheleishvili G, Graham P, Brooks A, Silvera R, Goldschmidt AJ, Pearse DD, Dietrich WD, Levi AD, Guest JD. Treating amyotrophic lateral sclerosis with allogeneic Schwann cell-derived exosomal vesicles: a case report. Neural Regen Res 2025; 20:1207-1216. [PMID: 38922880 PMCID: PMC11438342 DOI: 10.4103/nrr.nrr-d-23-01815] [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: 11/05/2023] [Revised: 01/05/2024] [Accepted: 02/24/2024] [Indexed: 06/28/2024] Open
Abstract
Schwann cells are essential for the maintenance and function of motor neurons, axonal networks, and the neuromuscular junction. In amyotrophic lateral sclerosis, where motor neuron function is progressively lost, Schwann cell function may also be impaired. Recently, important signaling and potential trophic activities of Schwann cell-derived exosomal vesicles have been reported. This case report describes the treatment of a patient with advanced amyotrophic lateral sclerosis using serial intravenous infusions of allogeneic Schwann cell-derived exosomal vesicles, marking, to our knowledge, the first instance of such treatment. An 81-year-old male patient presented with a 1.5-year history of rapidly progressive amyotrophic lateral sclerosis. After initial diagnosis, the patient underwent a combination of generic riluzole, sodium phenylbutyrate for the treatment of amyotrophic lateral sclerosis, and taurursodiol. The patient volunteered to participate in an FDA-approved single-patient expanded access treatment and received weekly intravenous infusions of allogeneic Schwann cell-derived exosomal vesicles to potentially restore impaired Schwann cell and motor neuron function. We confirmed that cultured Schwann cells obtained from the amyotrophic lateral sclerosis patient via sural nerve biopsy appeared impaired (senescent) and that exposure of the patient's Schwann cells to allogeneic Schwann cell-derived exosomal vesicles, cultured expanded from a cadaver donor improved their growth capacity in vitro. After a period of observation lasting 10 weeks, during which amyotrophic lateral sclerosis Functional Rating Scale-Revised and pulmonary function were regularly monitored, the patient received weekly consecutive infusions of 1.54 × 10 12 (×2), and then consecutive infusions of 7.5 × 10 12 (×6) allogeneic Schwann cell-derived exosomal vesicles diluted in 40 mL of Dulbecco's phosphate-buffered saline. None of the infusions were associated with adverse events such as infusion reactions (allergic or otherwise) or changes in vital signs. Clinical lab serum neurofilament and cytokine levels measured prior to each infusion varied somewhat without a clear trend. A more sensitive in-house assay suggested possible inflammasome activation during the disease course. A trend for clinical stabilization was observed during the infusion period. Our study provides a novel approach to address impaired Schwann cells and possibly motor neuron function in patients with amyotrophic lateral sclerosis using allogeneic Schwann cell-derived exosomal vesicles. Initial findings suggest that this approach is safe.
Collapse
Affiliation(s)
| | - Aisha Khan
- Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - George Jimsheleishvili
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Patricia Graham
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Adriana Brooks
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Risset Silvera
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
| | | | - Damien D. Pearse
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - W. Dalton Dietrich
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Allan D. Levi
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - James D. Guest
- The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, USA
- Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
2
|
Tajabadi Z, Dadkhah PA, Gholami Chahkand MS, Esmaeilpour Moallem F, Karimi MA, Amini-Salehi E, Karimi M. Exploring the role of exosomes in diabetic neuropathy: From molecular mechanisms to therapeutic potential. Biomed Pharmacother 2025; 185:117959. [PMID: 40056828 DOI: 10.1016/j.biopha.2025.117959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/24/2025] [Accepted: 03/04/2025] [Indexed: 03/10/2025] Open
Abstract
Diabetic neuropathy (DN) is a debilitating complication of diabetes mellitus (DM), characterized by progressive neuronal damage, sensory dysfunction, and impaired quality of life. Recent advances in exosome research have elucidated their crucial role in DN's pathogenesis, diagnosis, and treatment. Exosomes-nanoscale extracellular vesicles-function as vehicles for molecular cargo, including microRNAs (miRNAs), proteins, and lipids, which mediate intercellular communication and regulate key biological processes. Pathologically, hyperglycemia and hyperlipidemia induce the release of exosomes enriched with pathogenic miRNAs, such as miR-130a and miR-20b-3p, which disrupt neuronal function, axonal regeneration, and inflammatory pathways. Conversely, diagnostic studies highlight the utility of exosomal biomarkers like miR-7 and miR-221 in the early detection and monitoring of DN. Therapeutically, Schwann cell-derived and mesenchymal stromal cell (MSC)-derived exosomes demonstrate neuroprotective and reparative effects by enhancing mitochondrial function, modulating inflammation, and promoting axonal repair. Emerging approaches, including engineered exosomes and miRNA-enriched vesicles, further expand their therapeutic potential. Despite these advances, challenges such as standardization, large-scale production, and clinical validation remain in translating these findings into clinical practice. This review underscores the multifaceted roles of exosomes in DN and highlights their potential as innovative tools for precision diagnostics and targeted therapies, paving the way for future research and clinical applications.
Collapse
Affiliation(s)
- Zohreh Tajabadi
- Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | - Mohammad Amin Karimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | | - Mehdi Karimi
- Faculty of Medicine, Bogomolets National Medical University (NMU), Kyiv, Ukraine
| |
Collapse
|
3
|
Saha P, Yarra SS, Arruri V, Mohan U, Kumar A. Exploring the role of miRNA in diabetic neuropathy: from diagnostics to therapeutics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1129-1144. [PMID: 39249503 DOI: 10.1007/s00210-024-03422-y] [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: 05/17/2024] [Accepted: 08/29/2024] [Indexed: 09/10/2024]
Abstract
Diabetic neuropathy (DN) is one of the major microvascular complications of diabetes mellitus affecting 50% of the diabetic population marred by various unmet clinical needs. There is a need to explore newer pathological mechanisms for designing futuristic regimens for the management of DN. There is a need for post-transcriptional regulation of gene expression by non-coding RNAs (ncRNAs) to finetune different cellular mechanisms with significant biological relevance. MicroRNAs (miRNAs) are a class of small ncRNAs (~ 20 to 24 nucleotide length) that are known to regulate the activity of ~ 50% protein-coding genes through repression of their target mRNAs. Differential expression of these miRNAs is associated with the pathophysiology of diabetic neuropathy via regulating various pathways such as neuronal hyperexcitability, inflammation, axonal growth, regeneration, and oxidative stress. Of note, the circulating and extracellular vesicular miRNAs serve as potential biomarkers underscoring their diagnostic potential. Recent pieces of evidence highlight the potential of miRNAs in modulating the initiation and progression of DN and the possibility of developing miRNAs as treatment options for DN. In this review, we have elaborated on the role of different miRNAs as potential biomarkers and emphasized their druggable aspects for promising future therapies for the clinical management of DN.
Collapse
Affiliation(s)
- Priya Saha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) SAS Nagar, Sec 67, Mohali, Punjab, 160062, India
| | - Sai Sumanjali Yarra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Kolkata, Maniktala Main Road, Kolkata, West Bengal, India
| | - Vijay Arruri
- Department of Neurological Surgery, University of Wisconsin, Madison, USA
| | - Utpal Mohan
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) Kolkata, Maniktala Main Road, Kolkata, West Bengal, India
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) SAS Nagar, Sec 67, Mohali, Punjab, 160062, India.
| |
Collapse
|
4
|
Wang Y, Qu F, Wu Y, Lan K, Shen Y, Wu Z, Zhong Q, Cao X, Fan Z, Xu C. Peripheral nerves modulate the peri-implant osteogenesis under type 2 diabetes through exosomes derived from schwann cells via miR-15b-5p/Txnip signaling axis. J Nanobiotechnology 2025; 23:51. [PMID: 39875954 PMCID: PMC11773925 DOI: 10.1186/s12951-025-03160-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/22/2025] [Indexed: 01/30/2025] Open
Abstract
Studies have shown that the prognosis of dental implant treatment in patients with diabetes is not as good as that in the non-diabetes population. The nerve plays a crucial role in bone metabolism, but the role and the mechanism of peripheral nerves in regulating peri-implant osteogenesis under Type 2 diabetes mellitus (T2DM) situation remains unclear. In this study, it was shown that high glucose-stimulated Schwann cells (SCs) inhibited peri-implant osteogenesis via their exosomes. SCs-derived exosomes were analyzed for their miRNA cargo, identifying miR-15b-5p as significantly downregulated in high glucose conditions. T2DM rats and patients exhibited decreased miR-15b-5p expression, correlating with impaired bone microarchitecture. Luciferase assays and Western blotting confirmed TXNIP as a direct miR-15b-5p target, implicating its involvement in ROS signaling and inflammation-related osteogenesis suppression. Furthermore, normal SCs exosomes improved bone parameters around dental implants in T2DM rats. These findings underscore the therapeutic potential of miR-15b-5p and normal SCs exosomes in mitigating poor peri-implant bone regeneration of T2DM patients, offering insights into the molecular mechanisms of peripheral nerves governing bone regeneration in diabetic conditions.
Collapse
Affiliation(s)
- Yingying Wang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Shanghai, 200011, China
- National Center for Stomatology and National Clinical Research Center for Oral Diseases, No.639 Zhizaoju Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, No.639 Zhizaoju Road, Shanghai, 200011, China
| | - Fang Qu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Shanghai, 200011, China
- National Center for Stomatology and National Clinical Research Center for Oral Diseases, No.639 Zhizaoju Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, No.639 Zhizaoju Road, Shanghai, 200011, China
| | - Yaqin Wu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Shanghai, 200011, China
- National Center for Stomatology and National Clinical Research Center for Oral Diseases, No.639 Zhizaoju Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, No.639 Zhizaoju Road, Shanghai, 200011, China
| | - Kengliang Lan
- College of Stomatology, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Shanghai, 200011, China
- National Center for Stomatology and National Clinical Research Center for Oral Diseases, No.639 Zhizaoju Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, No.639 Zhizaoju Road, Shanghai, 200011, China
| | - Yingyi Shen
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Shanghai, 200011, China
- National Center for Stomatology and National Clinical Research Center for Oral Diseases, No.639 Zhizaoju Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, No.639 Zhizaoju Road, Shanghai, 200011, China
| | - Ziang Wu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Shanghai, 200011, China
- National Center for Stomatology and National Clinical Research Center for Oral Diseases, No.639 Zhizaoju Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, No.639 Zhizaoju Road, Shanghai, 200011, China
| | - Qi Zhong
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Shanghai, 200011, China
- National Center for Stomatology and National Clinical Research Center for Oral Diseases, No.639 Zhizaoju Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, No.639 Zhizaoju Road, Shanghai, 200011, China
| | - Ximeng Cao
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Shanghai, 200011, China
- National Center for Stomatology and National Clinical Research Center for Oral Diseases, No.639 Zhizaoju Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, No.639 Zhizaoju Road, Shanghai, 200011, China
| | - Zhen Fan
- Department of Implantology, School & Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, No.399 Middle Yanchang Road, Shanghai, 200072, China.
| | - Chun Xu
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200011, China.
- College of Stomatology, Shanghai Jiao Tong University, No.639 Zhizaoju Road, Shanghai, 200011, China.
- National Center for Stomatology and National Clinical Research Center for Oral Diseases, No.639 Zhizaoju Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, No.639 Zhizaoju Road, Shanghai, 200011, China.
| |
Collapse
|
5
|
Coy-Dibley J, Jayaraj ND, Ren D, Pacifico P, Belmadani A, Wang YZ, Gebis KK, Savas JN, Paller AS, Miller RJ, Menichella DM. Keratinocyte-derived extracellular vesicles in painful diabetic neuropathy. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2025; 17:100176. [PMID: 39811188 PMCID: PMC11731614 DOI: 10.1016/j.ynpai.2024.100176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/09/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025]
Abstract
Painful diabetic neuropathy (PDN) is a challenging complication of diabetes with patients experiencing a painful and burning sensation in their extremities. Existing treatments provide limited relief without addressing the underlying mechanisms of the disease. PDN involves the gradual degeneration of nerve fibers in the skin. Keratinocytes, the most abundant epidermal cell type, are closely positioned to cutaneous nerve terminals, suggesting the possibility of bi-directional communication. Extracellular vesicles are lipid-bilayer encapsulated nanovesicles released from many cell types that mediate cell to cell communication. The role of keratinocyte-derived extracellular vesicles (KDEVs) in influencing signaling between the skin and cutaneous nerve terminals and their contribution to the genesis of PDN has not been explored. In this study, we characterized KDEVs in a well-established high-fat diet mouse model of PDN using primary adult mouse keratinocyte cultures. We obtained highly enriched KDEVs through size-exclusion chromatography and then analyzed their molecular cargo using proteomic analysis and small RNA sequencing. We found significant differences in the protein and microRNA content of high-fat diet KDEVs compared to KDEVs obtained from control mice on a regular diet, including pathways involved in axon guidance and synaptic transmission. Additionally, using an in vivo conditional extracellular vesicle reporter mouse model, we demonstrated that epidermal-originating GFP-tagged KDEVs are retrogradely trafficked into the dorsal root ganglion (DRG) neuron cell bodies. This study presents the first comprehensive isolation and molecular characterization of the KDEV protein and microRNA cargo in RD and HFD mice. Our findings suggest a potential novel communication pathway between keratinocytes and DRG neurons in the skin, which could have implications for PDN.
Collapse
Affiliation(s)
- James Coy-Dibley
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Nirupa D. Jayaraj
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Dongjun Ren
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Paola Pacifico
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Abdelhak Belmadani
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yi-Zhi Wang
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kamil K. Gebis
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jeffrey N. Savas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Amy S. Paller
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Richard J. Miller
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Daniela M. Menichella
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
6
|
Pacifico P, Menichella DM. Molecular mechanisms of neuropathic pain. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:279-309. [PMID: 39580215 DOI: 10.1016/bs.irn.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
Peripheral neuropathic pain, which occurs after a lesion or disease affecting the peripheral somatosensory nervous system, is a complex and challenging condition to treat. This chapter will cover molecular mechanisms underlying the pathophysiology of peripheral neuropathic pain, focusing on (1) sensitization of nociceptors, (2) neuro-immune crosstalk, and (3) axonal degeneration and regeneration. The chapter will also emphasize the importance of identifying novel therapeutic targets in non-neuronal cells. A comprehensive understanding of how changes at both neuronal and non-neuronal levels contribute to peripheral neuropathic pain may significantly improve pain management and treatment options, expanding to topical application that bypass the side effects associated with systemic administration.
Collapse
Affiliation(s)
- Paola Pacifico
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| | - Daniela M Menichella
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States; Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| |
Collapse
|
7
|
Coy-Dibley J, Jayaraj ND, Ren D, Pacifico P, Belmadani A, Wang YZ, Gebis KK, Savas JN, Paller AS, Miller RJ, Menichella DM. Keratinocyte-Derived Exosomes in Painful Diabetic Neuropathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.21.608803. [PMID: 39229068 PMCID: PMC11370388 DOI: 10.1101/2024.08.21.608803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Painful diabetic neuropathy (PDN) is a challenging complication of diabetes with patients experiencing a painful and burning sensation in their extremities. Existing treatments provide limited relief without addressing the underlying mechanisms of the disease. PDN involves the gradual degeneration of nerve fibers in the skin. Keratinocytes, the most abundant epidermal cell type, are closely positioned to cutaneous nerve terminals, suggesting the possibility of bi-directional communication. Exosomes are small extracellular vesicles released from many cell types that mediate cell to cell communication. The role of keratinocyte-derived exosomes (KDEs) in influencing signaling between the skin and cutaneous nerve terminals and their contribution to the genesis of PDN has not been explored. In this study, we characterized KDEs in a well-established high-fat diet (HFD) mouse model of PDN using primary adult mouse keratinocyte cultures. We obtained highly enriched KDEs through size exclusion chromatography and then analyzed their molecular cargo using proteomic analysis and small RNA sequencing. We found significant differences in the protein and microRNA content of HFD KDEs compared to KDEs obtained from control mice on a regular diet (RD), including pathways involved in axon guidance and synaptic transmission. Additionally, using an in vivo conditional extracellular vesicle (EV) reporter mouse model, we demonstrated that epidermal-originating GFP-tagged KDEs are retrogradely trafficked into the DRG neuron cell body. Overall, our study presents a potential novel mode of communication between keratinocytes and DRG neurons in the skin, revealing a possible role for KDEs in contributing to the axonal degeneration that underlies neuropathic pain in PDN. Moreover, this study presents potential therapeutic targets in the skin for developing more effective, disease-modifying, and better-tolerated topical interventions for patients suffering from PDN, one of the most common and untreatable peripheral neuropathies.
Collapse
Affiliation(s)
- James Coy-Dibley
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Nirupa D Jayaraj
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Dongjun Ren
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Paola Pacifico
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Abdelhak Belmadani
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yi-Zhi Wang
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Kamil K Gebis
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jeffrey N Savas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Amy S Paller
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Richard J Miller
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Daniela M Menichella
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
8
|
Wang L, Lu X, Szalad A, Liu XS, Zhang Y, Wang X, Golembieski WA, Powell B, Mccann M, Lu M, Chopp M, Zhang ZG. Schwann cell-derived exosomes ameliorate peripheral neuropathy induced by ablation of dicer in Schwann cells. Front Cell Neurosci 2024; 18:1462228. [PMID: 39285940 PMCID: PMC11402728 DOI: 10.3389/fncel.2024.1462228] [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: 07/09/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Background MicroRNAs (miRNAs) in Schwann cells (SCs) mediate peripheral nerve function. Ablating Dicer, a key gene in miRNA biogenesis, in SCs causes peripheral neuropathy. Exosomes from healthy SCs (SC-Exo) ameliorate diabetic peripheral neuropathy in part via miRNAs. Thus, using transgenic mice with conditional and inducible ablation of Dicer in proteolipid protein (PLP) expressing SCs (PLP-cKO), we examined whether SC-Exo could reduce peripheral neuropathy in PLP-cKO mice. Methods PLP-cKO mice at the age of 16 weeks (8 week post-Tamoxifen) were randomly treated with SC-Exo or saline weekly for 8 weeks. Age-and sex-matched wild-type (WT) littermates were used as controls. Peripheral neurological functions, sciatic nerve integrity, and myelination were analyzed. Quantitative RT-PCR and Western blot analyses were performed to examine miRNA and protein expression in sciatic nerve tissues, respectively. Results Compared to the WT mice, PLP-cKO mice exhibited a significant decrease in motor and sensory conduction velocities, thermal sensitivity, and motor coordination. PLP-cKO mice exhibited substantial demyelination and axonal damage of the sciatic nerve. Treatment of PLP-cKO mice with SC-Exo significantly ameliorated the peripheral neuropathy and sciatic nerve damage. PLP-cKO mice showed a substantial reduction in a set of Dicer-related miRNAs known to regulate myelination, axonal integrity, and inflammation such as miR-138, -146a and - 338 in the sciatic nerve. In addition, PLP-cKO mice exhibited significant reduction of myelin forming proteins, early growth response 2 (EGR2) and sex determining region Y-box10 (Sox10), but significantly increased myelination inhibitors, Notch1, c-Jun, and Sox2 and the axonal growth inhibitor phosphatase and tens in homolog (PTEN). However, SC-Exo treatment reversed the PLP-cKO altered miRNAs and proteins. Conclusion This study demonstrates that exogenous SC-Exo ameliorate peripheral neuropathy induced by Dicer ablation in PLP expressing SCs. The therapeutic benefit may be mediated by the SC-Exo altered miRNAs and their targeted genes.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - XueRong Lu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Alexandra Szalad
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Xian Shuang Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Xinli Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Brianna Powell
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Mikkala Mccann
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Mei Lu
- Department of Biostatistics and Research Epidemiology, Henry Ford Hospital, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| |
Collapse
|
9
|
Abdal Dayem A, Yan E, Do M, Kim Y, Lee Y, Cho SG, Kim DH. Engineering extracellular vesicles for ROS scavenging and tissue regeneration. NANO CONVERGENCE 2024; 11:24. [PMID: 38922501 PMCID: PMC11208369 DOI: 10.1186/s40580-024-00430-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024]
Abstract
Stem cell therapy holds promise for tissue regeneration, yet significant challenges persist. Emerging as a safer and potentially more effective alternative, extracellular vesicles (EVs) derived from stem cells exhibit remarkable abilities to activate critical signaling cascades, thereby facilitating tissue repair. EVs, nano-scale membrane vesicles, mediate intercellular communication by encapsulating a diverse cargo of proteins, lipids, and nucleic acids. Their therapeutic potential lies in delivering cargos, activating signaling pathways, and efficiently mitigating oxidative stress-an essential aspect of overcoming limitations in stem cell-based tissue repair. This review focuses on engineering and applying EVs in tissue regeneration, emphasizing their role in regulating reactive oxygen species (ROS) pathways. Additionally, we explore strategies to enhance EV therapeutic activity, including functionalization and incorporation of antioxidant defense proteins. Understanding these molecular mechanisms is crucial for optimizing EV-based regenerative therapies. Insights into EV and ROS signaling modulation pave the way for targeted and efficient regenerative therapies harnessing the potential of EVs.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ellie Yan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Minjae Do
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yoojung Kim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Yeongseo Lee
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
- R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120, Neungdong-ro, Gwangjin- gu, Seoul, 05029, Republic of Korea.
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, 21205, USA.
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Institute for NanoBiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
10
|
González-Blanco C, Iglesias-Fortes S, Lockwood ÁC, Figaredo C, Vitulli D, Guillén C. The Role of Extracellular Vesicles in Metabolic Diseases. Biomedicines 2024; 12:992. [PMID: 38790954 PMCID: PMC11117504 DOI: 10.3390/biomedicines12050992] [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: 04/05/2024] [Revised: 04/23/2024] [Accepted: 04/27/2024] [Indexed: 05/26/2024] Open
Abstract
Extracellular vesicles represent a group of structures with the capacity to communicate with different cells and organs. This complex network of interactions can regulate multiple physiological processes in the organism. Very importantly, these processes can be altered during the appearance of different diseases including cancer, metabolic diseases, etc. In addition, these extracellular vesicles can transport different cargoes, altering the initiation of the disease, driving the progression, or even accelerating the pathogenesis. Then, we have explored the implication of these structures in different alterations such as pancreatic cancer, and in different metabolic alterations such as diabetes and its complications and non-alcoholic fatty liver disease. Finally, we have explored in more detail the communication between the liver and the pancreas. In summary, extracellular vesicles represent a very efficient system for the communication among different tissues and permit an efficient system as biomarkers of the disease, as well as being involved in the extracellular-vesicle-mediated transport of molecules, serving as a potential therapy for different diseases.
Collapse
Affiliation(s)
- Carlos González-Blanco
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28040 Madrid, Spain; (C.G.-B.); (Á.C.L.)
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (S.I.-F.); (C.F.); (D.V.)
- IdISSC, 28040 Madrid, Spain
- Dirección General de Investigación e Innovación Tecnológica (DGIIT), Consejería de Educación y Universidades, Comunidad de Madrid, 28001 Madrid, Spain
| | - Sarai Iglesias-Fortes
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (S.I.-F.); (C.F.); (D.V.)
| | - Ángela Cristina Lockwood
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28040 Madrid, Spain; (C.G.-B.); (Á.C.L.)
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (S.I.-F.); (C.F.); (D.V.)
- Dirección General de Investigación e Innovación Tecnológica (DGIIT), Consejería de Educación y Universidades, Comunidad de Madrid, 28001 Madrid, Spain
| | - César Figaredo
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (S.I.-F.); (C.F.); (D.V.)
| | - Daniela Vitulli
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (S.I.-F.); (C.F.); (D.V.)
| | - Carlos Guillén
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 28040 Madrid, Spain; (C.G.-B.); (Á.C.L.)
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain; (S.I.-F.); (C.F.); (D.V.)
- IdISSC, 28040 Madrid, Spain
- Dirección General de Investigación e Innovación Tecnológica (DGIIT), Consejería de Educación y Universidades, Comunidad de Madrid, 28001 Madrid, Spain
| |
Collapse
|
11
|
Cai H, Pang Y, Ren Z, Fu X, Jia L. Delivering synaptic protein mRNAs via extracellular vesicles ameliorates cognitive impairment in a mouse model of Alzheimer's disease. BMC Med 2024; 22:138. [PMID: 38528511 PMCID: PMC10964680 DOI: 10.1186/s12916-024-03359-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 03/15/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Synaptic dysfunction with reduced synaptic protein levels is a core feature of Alzheimer's disease (AD). Synaptic proteins play a central role in memory processing, learning, and AD pathogenesis. Evidence suggests that synaptic proteins in plasma neuronal-derived extracellular vesicles (EVs) are reduced in patients with AD. However, it remains unclear whether levels of synaptic proteins in EVs are associated with hippocampal atrophy of AD and whether upregulating the expression of these synaptic proteins has a beneficial effect on AD. METHODS In this study, we included 57 patients with AD and 56 healthy controls. We evaluated their brain atrophy through magnetic resonance imaging using the medial temporal lobe atrophy score. We measured the levels of four synaptic proteins, including synaptosome-associated protein 25 (SNAP25), growth-associated protein 43 (GAP43), neurogranin, and synaptotagmin 1 in both plasma neuronal-derived EVs and cerebrospinal fluid (CSF). We further examined the association of synaptic protein levels with brain atrophy. We also evaluated the levels of these synaptic proteins in the brains of 5×FAD mice. Then, we loaded rabies virus glycoprotein-engineered EVs with messenger RNAs (mRNAs) encoding GAP43 and SNAP25 and administered these EVs to 5×FAD mice. After treatment, synaptic proteins, dendritic density, and cognitive function were evaluated. RESULTS The results showed that GAP43, SNAP25, neurogranin, and synaptotagmin 1 were decreased in neuronal-derived EVs but increased in CSF in patients with AD, and the changes corresponded to the severity of brain atrophy. GAP43 and SNAP25 were decreased in the brains of 5×FAD mice. The engineered EVs efficiently and stably delivered these synaptic proteins to the brain, where synaptic protein levels were markedly upregulated. Upregulation of synaptic protein expression could ameliorate cognitive impairment in AD by promoting dendritic density. This marks the first successful delivery of synaptic protein mRNAs via EVs in AD mice, yielding remarkable therapeutic effects. CONCLUSIONS Synaptic proteins are closely related to AD processes. Delivery of synaptic protein mRNAs via EVs stands as a promising effective precision treatment strategy for AD, which significantly advances the current understanding of therapeutic approaches for the disease.
Collapse
Affiliation(s)
- Huimin Cai
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Yana Pang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Ziye Ren
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Xiaofeng Fu
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, 45 Changchun St., Beijing, 100053, China.
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China.
| |
Collapse
|
12
|
Vali R, Azadi A, Tizno A, Farkhondeh T, Samini F, Samarghandian S. miRNA contributes to neuropathic pains. Int J Biol Macromol 2023; 253:126893. [PMID: 37730007 DOI: 10.1016/j.ijbiomac.2023.126893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/29/2023] [Accepted: 09/11/2023] [Indexed: 09/22/2023]
Abstract
Neuropathic pain (NP) is a kind of chronic pain caused by direct injury to the peripheral or central nervous system (CNS). microRNAs (miRNAs) are small noncoding RNAs that mostly interact with the 3 untranslated region of messenger RNAs (mRNAs) to regulate the expression of multiple genes. NP is characterized by changes in the expression of receptors and mediators, and there is evidence that miRNAs may contribute to some of these alterations. In this review, we aimed to fully comprehend the connection between NP and miRNA; and also, to establish a link between neurology, biology, and dentistry. Studies have shown that targeting miRNAs may be an effective therapeutic strategy for the treatment of chronic pain and potential target for the prevention of NP.
Collapse
Affiliation(s)
- Reyhaneh Vali
- Department of Biology, Faculty of Modern Science, Tehran Medical Branch, Islamic Azad University, Tehran, Iran; Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Ali Azadi
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ashkan Tizno
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tahereh Farkhondeh
- Neuroscience Research Center, Kamyab Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fariborz Samini
- Department of Toxicology and Pharmacology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Department of Toxicology and Pharmacology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
13
|
Ghosh M, Pearse DD. Schwann Cell-Derived Exosomal Vesicles: A Promising Therapy for the Injured Spinal Cord. Int J Mol Sci 2023; 24:17317. [PMID: 38139147 PMCID: PMC10743801 DOI: 10.3390/ijms242417317] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/02/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Exosomes are nanoscale-sized membrane vesicles released by cells into their extracellular milieu. Within these nanovesicles reside a multitude of bioactive molecules, which orchestrate essential biological processes, including cell differentiation, proliferation, and survival, in the recipient cells. These bioactive properties of exosomes render them a promising choice for therapeutic use in the realm of tissue regeneration and repair. Exosomes possess notable positive attributes, including a high bioavailability, inherent safety, and stability, as well as the capacity to be functionalized so that drugs or biological agents can be encapsulated within them or to have their surface modified with ligands and receptors to imbue them with selective cell or tissue targeting. Remarkably, their small size and capacity for receptor-mediated transcytosis enable exosomes to cross the blood-brain barrier (BBB) and access the central nervous system (CNS). Unlike cell-based therapies, exosomes present fewer ethical constraints in their collection and direct use as a therapeutic approach in the human body. These advantageous qualities underscore the vast potential of exosomes as a treatment option for neurological injuries and diseases, setting them apart from other cell-based biological agents. Considering the therapeutic potential of exosomes, the current review seeks to specifically examine an area of investigation that encompasses the development of Schwann cell (SC)-derived exosomal vesicles (SCEVs) as an approach to spinal cord injury (SCI) protection and repair. SCs, the myelinating glia of the peripheral nervous system, have a long history of demonstrated benefit in repair of the injured spinal cord and peripheral nerves when transplanted, including their recent advancement to clinical investigations for feasibility and safety in humans. This review delves into the potential of utilizing SCEVs as a therapy for SCI, explores promising engineering strategies to customize SCEVs for specific actions, and examines how SCEVs may offer unique clinical advantages over SC transplantation for repair of the injured spinal cord.
Collapse
Affiliation(s)
- Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
14
|
Caparaso SM, Redwine AL, Wachs RA. Engineering a multicompartment in vitro model for dorsal root ganglia phenotypic assessment. J Biomed Mater Res B Appl Biomater 2023; 111:1903-1920. [PMID: 37326300 PMCID: PMC10527728 DOI: 10.1002/jbm.b.35294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 05/19/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023]
Abstract
Despite the significant global prevalence of chronic pain, current methods to identify pain therapeutics often fail translation to the clinic. Phenotypic screening platforms rely on modeling and assessing key pathologies relevant to chronic pain, improving predictive capability. Patients with chronic pain often present with sensitization of primary sensory neurons (that extend from dorsal root ganglia [DRG]). During neuronal sensitization, painful nociceptors display lowered stimulation thresholds. To model neuronal excitability, it is necessary to maintain three key anatomical features of DRGs to have a physiologically relevant platform: (1) isolation between DRG cell bodies and neurons, (2) 3D platform to preserve cell-cell and cell-matrix interactions, and (3) presence of native non-neuronal support cells, including Schwann cells and satellite glial cells. Currently, no culture platforms maintain the three anatomical features of DRGs. Herein, we demonstrate an engineered 3D multicompartment device that isolates DRG cell bodies and neurites and maintains native support cells. We observed neurite growth into isolated compartments from the DRG using two formulations of collagen, hyaluronic acid, and laminin-based hydrogels. Further, we characterized the rheological, gelation and diffusivity properties of the two hydrogel formulations and found the mechanical properties mimic native neuronal tissue. Importantly, we successfully limited fluidic diffusion between the DRG and neurite compartment for up to 72 h, suggesting physiological relevance. Lastly, we developed a platform with the capability of phenotypic assessment of neuronal excitability using calcium imaging. Ultimately, our culture platform can screen neuronal excitability, providing a more translational and predictive system to identify novel pain therapeutics to treat chronic pain.
Collapse
Affiliation(s)
- Sydney M. Caparaso
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln Nebraska, USA
| | - Adan L. Redwine
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln Nebraska, USA
| | - Rebecca A. Wachs
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln Nebraska, USA
| |
Collapse
|
15
|
Oliveira JT, Yanick C, Wein N, Gomez Limia CE. Neuron-Schwann cell interactions in peripheral nervous system homeostasis, disease, and preclinical treatment. Front Cell Neurosci 2023; 17:1248922. [PMID: 37900588 PMCID: PMC10600466 DOI: 10.3389/fncel.2023.1248922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023] Open
Abstract
Schwann cells (SCs) have a critical role in the peripheral nervous system. These cells are able to support axons during homeostasis and after injury. However, mutations in genes associated with the SCs repair program or myelination result in dysfunctional SCs. Several neuropathies such as Charcot-Marie-Tooth (CMT) disease, diabetic neuropathy and Guillain-Barré syndrome show abnormal SC functions and an impaired regeneration process. Thus, understanding SCs-axon interaction and the nerve environment in the context of homeostasis as well as post-injury and disease onset is necessary. Several neurotrophic factors, cytokines, and regulators of signaling pathways associated with proliferation, survival and regeneration are involved in this process. Preclinical studies have focused on the discovery of therapeutic targets for peripheral neuropathies and injuries. To study the effect of new therapeutic targets, modeling neuropathies and peripheral nerve injuries (PNIs) in vitro and in vivo are useful tools. Furthermore, several in vitro protocols have been designed using SCs and neuron cell lines to evaluate these targets in the regeneration process. SCs lines have been used to generate effective myelinating SCs without success. Alternative options have been investigated using direct conversion from somatic cells to SCs or SCs derived from pluripotent stem cells to generate functional SCs. This review will go over the advantages of these systems and the problems associated with them. In addition, there have been challenges in establishing adequate and reproducible protocols in vitro to recapitulate repair SC-neuron interactions observed in vivo. So, we also discuss the mechanisms of repair SCs-axon interactions in the context of peripheral neuropathies and nerve injury (PNI) in vitro and in vivo. Finally, we summarize current preclinical studies evaluating transgenes, drug, and novel compounds with translational potential into clinical studies.
Collapse
Affiliation(s)
| | | | - Nicolas Wein
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| | | |
Collapse
|
16
|
Satyadev N, Rivera MI, Nikolov NK, Fakoya AOJ. Exosomes as biomarkers and therapy in type 2 diabetes mellitus and associated complications. Front Physiol 2023; 14:1241096. [PMID: 37745252 PMCID: PMC10515224 DOI: 10.3389/fphys.2023.1241096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is one of the most prevalent metabolic disorders worldwide. However, T2DM still remains underdiagnosed and undertreated resulting in poor quality of life and increased morbidity and mortality. Given this ongoing burden, researchers have attempted to locate new therapeutic targets as well as methodologies to identify the disease and its associated complications at an earlier stage. Several studies over the last few decades have identified exosomes, small extracellular vesicles that are released by cells, as pivotal contributors to the pathogenesis of T2DM and its complications. These discoveries suggest the possibility of novel detection and treatment methods. This review provides a comprehensive presentation of exosomes that hold potential as novel biomarkers and therapeutic targets. Additional focus is given to characterizing the role of exosomes in T2DM complications, including diabetic angiopathy, diabetic cardiomyopathy, diabetic nephropathy, diabetic peripheral neuropathy, diabetic retinopathy, and diabetic wound healing. This study reveals that the utilization of exosomes as diagnostic markers and therapies is a realistic possibility for both T2DM and its complications. However, the majority of the current research is limited to animal models, warranting further investigation of exosomes in clinical trials. This review represents the most extensive and up-to-date exploration of exosomes in relation to T2DM and its complications.
Collapse
Affiliation(s)
- Nihal Satyadev
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, United States
| | - Milagros I. Rivera
- University of Medicine and Health Sciences, Basseterre, St. Kitts and Nevis
| | | | | |
Collapse
|
17
|
Eid SA, Rumora AE, Beirowski B, Bennett DL, Hur J, Savelieff MG, Feldman EL. New perspectives in diabetic neuropathy. Neuron 2023; 111:2623-2641. [PMID: 37263266 PMCID: PMC10525009 DOI: 10.1016/j.neuron.2023.05.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/20/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023]
Abstract
Diabetes prevalence continues to climb with the aging population. Type 2 diabetes (T2D), which constitutes most cases, is metabolically acquired. Diabetic peripheral neuropathy (DPN), the most common microvascular complication, is length-dependent damage to peripheral nerves. DPN pathogenesis is complex, but, at its core, it can be viewed as a state of impaired metabolism and bioenergetics failure operating against the backdrop of long peripheral nerve axons supported by glia. This unique peripheral nerve anatomy and the injury consequent to T2D underpins the distal-to-proximal symptomatology of DPN. Earlier work focused on the impact of hyperglycemia on nerve damage and bioenergetics failure, but recent evidence additionally implicates contributions from obesity and dyslipidemia. This review will cover peripheral nerve anatomy, bioenergetics, and glia-axon interactions, building the framework for understanding how hyperglycemia and dyslipidemia induce bioenergetics failure in DPN. DPN and painful DPN still lack disease-modifying therapies, and research on novel mechanism-based approaches is also covered.
Collapse
Affiliation(s)
- Stephanie A Eid
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Amy E Rumora
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Bogdan Beirowski
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Neuroscience Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - David L Bennett
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford OX3 9DU, UK
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Masha G Savelieff
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
18
|
Liu YT, Xu Z, Liu W, Ren S, Xiong HW, Jiang T, Chen J, Kang Y, Li QY, Wu ZH, Machens HG, Yang XF, Chen ZB. The circ_0002538/miR-138-5p/plasmolipin axis regulates Schwann cell migration and myelination in diabetic peripheral neuropathy. Neural Regen Res 2023; 18:1591-1600. [PMID: 36571367 PMCID: PMC10075099 DOI: 10.4103/1673-5374.355979] [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] [Indexed: 11/19/2022] Open
Abstract
Circular RNAs (circRNAs) play a vital role in diabetic peripheral neuropathy. However, their expression and function in Schwann cells in individuals with diabetic peripheral neuropathy remain poorly understood. Here, we performed protein profiling and circRNA sequencing of sural nerves in patients with diabetic peripheral neuropathy and controls. Protein profiling revealed 265 differentially expressed proteins in the diabetic peripheral neuropathy group. Gene Ontology indicated that differentially expressed proteins were mainly enriched in myelination and mitochondrial oxidative phosphorylation. A real-time polymerase chain reaction assay performed to validate the circRNA sequencing results yielded 11 differentially expressed circRNAs. circ_0002538 was markedly downregulated in patients with diabetic peripheral neuropathy. Further in vitro experiments showed that overexpression of circ_0002538 promoted the migration of Schwann cells by upregulating plasmolipin (PLLP) expression. Moreover, overexpression of circ_0002538 in the sciatic nerve in a streptozotocin-induced mouse model of diabetic peripheral neuropathy alleviated demyelination and improved sciatic nerve function. The results of a mechanistic experiment showed that circ_0002538 promotes PLLP expression by sponging miR-138-5p, while a lack of circ_0002538 led to a PLLP deficiency that further suppressed Schwann cell migration. These findings suggest that the circ_0002538/miR-138-5p/PLLP axis can promote the migration of Schwann cells in diabetic peripheral neuropathy patients, improving myelin sheath structure and nerve function. Thus, this axis is a potential target for therapeutic treatment of diabetic peripheral neuropathy.
Collapse
Affiliation(s)
- Yu-Tian Liu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhao Xu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Wei Liu
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Sen Ren
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - He-Wei Xiong
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Tao Jiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yu Kang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qian-Yun Li
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zi-Han Wu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hans-GüNther Machens
- Department of Plastic and Hand Surgery, Technical University of Munich, Munich, Germany
| | - Xiao-Fan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhen-Bing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
19
|
Li B, Li W, Liu T, Zha L. Extracellular vesicles regulate the transmission of insulin resistance and redefine noncommunicable diseases. Front Mol Biosci 2023; 9:1024786. [PMID: 36699697 PMCID: PMC9868246 DOI: 10.3389/fmolb.2022.1024786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/19/2022] [Indexed: 01/10/2023] Open
Abstract
Noncommunicable diseases (NCDs), such as diabetes and related neurological disorders, are considered to not be directly transmissible from one person to another. However, NCDs may be transmissible in vivo through extracellular vesicles (EVs). A long-term high-fat diet (HFD) can induce a series of health issues like hyperlipidemia, type 2 diabetes mellitus (T2DM), and diabetic peripheral neuropathy (DPN) due to insulin resistance. Multiple molecular signaling changes can stimulate insulin resistance, especially blocking insulin signaling by increased insulin resistance inducer (phosphorylation of negative regulatory sites of insulin receptor substrate (IRS) proteins) and decreased tyrosine phosphorylation of insulin receptor substrate (phosphorylation of positive regulatory sites of IRS), thus leading to reduced phosphorylation of AKT enzymes. Current efforts to treat T2DM and prevent its complications mainly focus on improving insulin sensitivity, enhancing insulin secretion, or supplementing exogenous insulin based on a common assumption that insulin resistance is noncommunicable. However, insulin resistance is transmissible within multiple tissues or organs throughout the body. Exploring the regulatory roles of EVs in developing insulin resistance may provide novel and effective preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Biao Li
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, National Medical Products Administration Key Laboratory of Cosmetic Safety Evaluation, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Wan Li
- School of Physical Education, Hubei Minzu University, Enshi, China
| | - Tiancai Liu
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Longying Zha
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, National Medical Products Administration Key Laboratory of Cosmetic Safety Evaluation, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
20
|
Razi Soofiyani S, Minaei Beirami S, Hosseini K, Mohammadi Nasr M, Ranjbar M, Forouhandeh H, Tarhriz V, Sadeghi M. Revisiting Inhibition Effects of miR-28 as a Metastasis Suppressor in Gastrointestinal Cancers. Microrna 2023; 12:131-142. [PMID: 37073155 DOI: 10.2174/2211536612666230413125126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 11/09/2022] [Accepted: 01/20/2023] [Indexed: 04/20/2023]
Abstract
MicroRNAs are critical epigenetic regulators that can be used as diagnostic, prognostic, and therapeutic biomarkers for the treatment of various diseases, including gastrointestinal cancers, among a variety of cellular and molecular biomarkers. MiRNAs have also shown oncogenic or tumor suppressor roles in tumor tissue and other cell types. Studies showed that the dysregulation of miR-28 is involved in cell growth and metastasis of gastrointestinal cancers. MiR-28 plays a key role in controlling the physiological processes of cancer cells including growth and proliferation, migration, invasion, apoptosis, and metastasis. Therefore, miR-28 expression patterns can be used to distinguish patient subgroups. Based on the previous studies, miR-28 expression can be a suitable biomarker to detect tumor size and predict histological grade metastasis. In this review, we summarize the inhibitory effects of miR-28 as a metastasis suppressor in gastrointestinal cancers. miR-28 plays a role as a tumor suppressor in gastrointestinal cancers by regulating cancer cell growth, cell differentiation, angiogenesis, and metastasis. As a result, using it as a prognostic, diagnostic, and therapeutic biomarker in the treatment of gastrointestinal cancers can be a way to solve the problems in this field.
Collapse
Affiliation(s)
- Saiedeh Razi Soofiyani
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Clinical Research Development Unit, Sina Educational, Research and Treatment Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sohrab Minaei Beirami
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kamran Hosseini
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Faculty of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mina Mohammadi Nasr
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences. Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Ranjbar
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Haleh Forouhandeh
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Tarhriz
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Sadeghi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences. Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Liu YP, Tian MY, Yang YD, Li H, Zhao TT, Zhu J, Mou FF, Cui GH, Guo HD, Shao SJ. Schwann cells-derived exosomal miR-21 participates in high glucose regulation of neurite outgrowth. iScience 2022; 25:105141. [PMID: 36204278 PMCID: PMC9529988 DOI: 10.1016/j.isci.2022.105141] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/06/2022] [Accepted: 09/09/2022] [Indexed: 11/29/2022] Open
Abstract
As a common complication of diabetes, the pathogenesis of diabetic peripheral neuropathy (DPN) is closely related to high glucose but has not been clarified. Exosomes can mediate crosstalk between Schwann cells (SC) and neurons in the peripheral nerve. Herein, we found that miR-21 in serum exosomes from DPN rats was decreased. SC proliferation was inhibited, cell apoptosis was increased, and the expression of miR-21 in cells and exosomes was downregulated when cultured in high glucose. Increasing miR-21 expression reversed these changes, while knockdown of miR-21 led to the opposite results. When co-cultured with exosomes derived from SC exposed to high glucose, neurite outgrowth was inhibited. On the contrary, neurite outgrowth was accelerated when incubated with exosomes rich in miR-21. We further demonstrated that the SC-derived exosomal miR-21 participates in neurite outgrowth probably through the AKT signaling pathway. Thus, SC-derived exosomal miR-21 contributes to high glucose regulation of neurite outgrowth. The miR-21 was decreased in serum exosomes and sciatic nerve of DPN rats High glucose inhibited SC viability and downregulated the expression of miR-21 Exosomes derived from SC cultured in high glucose inhibited the neurite outgrowth SC-derived exosomes rich in miR-21 accelerated the neurite outgrowth of neuron
Collapse
Affiliation(s)
- Yu-pu Liu
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Ming-yue Tian
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yi-duo Yang
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Han Li
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tian-tian Zhao
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jing Zhu
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fang-fang Mou
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Guo-hong Cui
- Department of Neurology, Shanghai No. 9 People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
- Corresponding author
| | - Hai-dong Guo
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Corresponding author
| | - Shui-jin Shao
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Corresponding author
| |
Collapse
|
22
|
Ding G, Li L, Zhang L, Chopp M, Davoodi-Bojd E, Li Q, Li C, Wei M, Zhang Z, Jiang Q. MRI Metrics of Cerebral Endothelial Cell-Derived Exosomes for the Treatment of Cognitive Dysfunction Induced in Aging Rats Subjected to Type 2 Diabetes. Diabetes 2022; 71:873-880. [PMID: 35175337 PMCID: PMC9044132 DOI: 10.2337/db21-0754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 02/10/2022] [Indexed: 11/13/2022]
Abstract
Ongoing neurovascular dysfunction contributes to type 2 diabetes mellitus (T2DM)-induced cognitive deficits. However, it is not known whether early post onset of T2DM interventions may reduce evolving neurovascular dysfunction and thereby lead to diminution of T2DM-induced cognitive deficits. Using multiple MRI metrics, we evaluated neurovascular changes in T2DM rats treated with exosomes derived from cerebral endothelial cells (CEC-Exos). Two months after induction of T2DM in middle-aged male rats by administration of streptozotocin nicotinamide, rats were randomly treated with CEC-Exos twice weekly or saline for 4 consecutive weeks (n = 10/group). MRI measurements were performed at the end of the treatment, which included cerebral blood flow (CBF), contrast-enhanced T1-weighted imaging, and relaxation time constants T1 and T2. MRI analysis showed that compared with controls, the CEC-Exo-treated T2DM rats exhibited significant elevation of T2 and CBF in white matter and significant augmentation of T1 and reduction of blood-brain barrier permeability in gray matter. In the hippocampus, CEC-Exo treatment significantly increased T1 and CBF. Furthermore, CEC-Exo treatment significantly reduced T2DM-induced cognitive deficits measured by the Morris water maze and odor recognition tests. Collectively, our corresponding MRI data demonstrate that treatment of T2DM rats with CEC-Exos robustly reduced neurovascular dysfunction in gray and white matter and the hippocampus.
Collapse
Affiliation(s)
| | - Lian Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI
| | - Li Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI
- Department of Physics, Oakland University, Rochester, MI
| | | | - Qingjiang Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI
| | - Chao Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI
| | - Min Wei
- Department of Neurology, Henry Ford Hospital, Detroit, MI
| | | | - Quan Jiang
- Department of Neurology, Henry Ford Hospital, Detroit, MI
- Department of Physics, Oakland University, Rochester, MI
| |
Collapse
|
23
|
Ahmad S, Srivastava RK, Singh P, Naik UP, Srivastava AK. Role of Extracellular Vesicles in Glia-Neuron Intercellular Communication. Front Mol Neurosci 2022; 15:844194. [PMID: 35493327 PMCID: PMC9043804 DOI: 10.3389/fnmol.2022.844194] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cross talk between glia and neurons is crucial for a variety of biological functions, ranging from nervous system development, axonal conduction, synaptic transmission, neural circuit maturation, to homeostasis maintenance. Extracellular vesicles (EVs), which were initially described as cellular debris and were devoid of biological function, are now recognized as key components in cell-cell communication and play a critical role in glia-neuron communication. EVs transport the proteins, lipids, and nucleic acid cargo in intercellular communication, which alters target cells structurally and functionally. A better understanding of the roles of EVs in glia-neuron communication, both in physiological and pathological conditions, can aid in the discovery of novel therapeutic targets and the development of new biomarkers. This review aims to demonstrate that different types of glia and neuronal cells secrete various types of EVs, resulting in specific functions in intercellular communications.
Collapse
Affiliation(s)
- Shahzad Ahmad
- Department of Medical Elementology and Toxicology, Jamia Hamdard University, New Delhi, India
| | - Rohit K. Srivastava
- Department of Pediatric Surgery, Texas Children’s Hospital, Houston, TX, United States
- M.E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Pratibha Singh
- Department of Biochemistry and Cell Biology, Biosciences Research Collaborative, Rice University, Houston, TX, United States
| | - Ulhas P. Naik
- Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Cardeza Foundation for Hematologic Research, Philadelphia, PA, United States
| | - Amit K. Srivastava
- Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Cardeza Foundation for Hematologic Research, Philadelphia, PA, United States
- *Correspondence: Amit K. Srivastava,
| |
Collapse
|
24
|
Guo Y, Gil Z. The Role of Extracellular Vesicles in Cancer-Nerve Crosstalk of the Peripheral Nervous System. Cells 2022; 11:cells11081294. [PMID: 35455973 PMCID: PMC9027707 DOI: 10.3390/cells11081294] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 03/30/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023] Open
Abstract
Although the pathogenic operations of cancer–nerve crosstalk (e.g., neuritogenesis, neoneurogensis, and perineural invasion—PNI) in the peripheral nervous system (PNS) during tumorigenesis, as well as the progression of all cancer types is continuing to emerge as an area of unique scientific interest and study, extensive, wide-ranging, and multidisciplinary investigations still remain fragmented and unsystematic. This is especially so in regard to the roles played by extracellular vesicles (EVs), which are lipid bilayer-enclosed nano- to microsized particles that carry multiple-function molecular cargos, facilitate intercellular communication in diverse processes. Accordingly, the biological significance of EVs has been greatly elevated in recent years, as there is strong evidence that they could contribute to important and possibly groundbreaking diagnostic and therapeutic innovations. This can be achieved and the pace of discoveries accelerated through cross-pollination from existing knowledge and studies regarding nervous system physiology and pathology, as well as thoroughgoing collaborations between oncologists, neurobiologists, pathologists, clinicians, and researchers. This article offers an overview of current and recent past investigations on the roles of EVs in cancer–nerve crosstalk, as well as in neural development, physiology, inflammation, injury, and regeneration in the PNS. By highlighting the mechanisms involved in physiological and noncancerous pathological cellular crosstalk, we provide hints that may inspire additional translational studies on cancer–nerve interplay.
Collapse
Affiliation(s)
- Yuanning Guo
- Rappaport Family Institute for Research in the Medical Sciences, Technion—Israel Institute of Technology, Haifa 31096, Israel;
| | - Ziv Gil
- Rappaport Family Institute for Research in the Medical Sciences, Technion—Israel Institute of Technology, Haifa 31096, Israel;
- Head and Neck Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel
- Correspondence: ; Tel.: +972-4-854-2480
| |
Collapse
|
25
|
Rampin A, Carrabba M, Mutoli M, Eman CL, Testa G, Madeddu P, Spinetti G. Recent Advances in KEAP1/NRF2-Targeting Strategies by Phytochemical Antioxidants, Nanoparticles, and Biocompatible Scaffolds for the Treatment of Diabetic Cardiovascular Complications. Antioxid Redox Signal 2022; 36:707-728. [PMID: 35044251 DOI: 10.1089/ars.2021.0134] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: Modulation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2)-mediated antioxidant response is a key aspect in the onset of diabetes-related cardiovascular complications. With this review, we provide an overview of the recent advances made in the development of Nrf2-targeting strategies for the treatment of diabetes, with particular attention toward the activation of Nrf2 by natural antioxidant compounds, nanoparticles, and oxidative stress-modulating biocompatible scaffolds. Recent Advances: In the past 30 years, studies addressing the use of antioxidant therapies to treat diabetes have grown exponentially, showing promising but yet inconclusive results. Animal studies and clinical trials on the Nrf2 pathway have shown promising results, suggesting that its activation can delay or reverse some of the cardiovascular impairments in diabetes. Critical Issues: Hyperglycemia- and oscillating glucose levels-induced reactive oxygen species (ROS) accumulation is progressively emerging as a central factor in the onset and progression of diabetes-related cardiovascular complications, including endothelial dysfunction, retinopathy, heart failure, stroke, critical limb ischemia, ulcers, and delayed wound healing. In this context, accumulating evidence suggests a central role for Nrf2-mediated antioxidant response, one of the most studied cellular defensive mechanisms against ROS accumulation. Future Directions: Innovative approaches such as tissue engineering and nanotechnology are converging toward targeting oxidative stress in diabetes. Antioxid. Redox Signal. 36, 707-728.
Collapse
Affiliation(s)
- Andrea Rampin
- Laboratory of Cardiovascular Physiopathology-Regenerative Medicine, IRCCS MultiMedica, Milan, Italy
| | - Michele Carrabba
- Laboratory of Experimental Cardiovascular Medicine, University of Bristol, Bristol, England, United Kingdom
| | - Martina Mutoli
- Laboratory of Cardiovascular Physiopathology-Regenerative Medicine, IRCCS MultiMedica, Milan, Italy
| | - Charlotte L Eman
- Laboratory of Cardiovascular Physiopathology-Regenerative Medicine, IRCCS MultiMedica, Milan, Italy
| | - Gianluca Testa
- Department of Medicine and Health Sciences, "V. Tiberio" University of Molise, Campobasso, Italy.,Interdepartmental Center for Nanotechnology Research-NanoBem, University of Molise, Campobasso, Italy
| | - Paolo Madeddu
- Laboratory of Experimental Cardiovascular Medicine, University of Bristol, Bristol, England, United Kingdom
| | - Gaia Spinetti
- Laboratory of Cardiovascular Physiopathology-Regenerative Medicine, IRCCS MultiMedica, Milan, Italy
| |
Collapse
|
26
|
Gada Y, Pandey A, Jadhav N, Ajgaonkar S, Mehta D, Nair S. New Vistas in microRNA Regulatory Interactome in Neuropathic Pain. Front Pharmacol 2022; 12:778014. [PMID: 35280258 PMCID: PMC8914318 DOI: 10.3389/fphar.2021.778014] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022] Open
Abstract
Neuropathic pain is a chronic pain condition seen in patients with diabetic neuropathy, cancer chemotherapy-induced neuropathy, idiopathic neuropathy as well as other diseases affecting the nervous system. Only a small percentage of people with neuropathic pain benefit from current medications. The complexity of the disease, poor identification/lack of diagnostic and prognostic markers limit current strategies for the management of neuropathic pain. Multiple genes and pathways involved in human diseases can be regulated by microRNA (miRNA) which are small non-coding RNA. Several miRNAs are found to be dysregulated in neuropathic pain. These miRNAs regulate expression of various genes associated with neuroinflammation and pain, thus, regulating neuropathic pain. Some of these key players include adenylate cyclase (Ac9), toll-like receptor 8 (Tlr8), suppressor of cytokine signaling 3 (Socs3), signal transducer and activator of transcription 3 (Stat3) and RAS p21 protein activator 1 (Rasa1). With advancements in high-throughput technology and better computational power available for research in present-day pharmacology, biomarker discovery has entered a very exciting phase. We dissect the architecture of miRNA biological networks encompassing both human and rodent microRNAs involved in the development of neuropathic pain. We delineate various microRNAs, and their targets, that may likely serve as potential biomarkers for diagnosis, prognosis, and therapeutic intervention in neuropathic pain. miRNAs mediate their effects in neuropathic pain by signal transduction through IRAK/TRAF6, TLR4/NF-κB, TXIP/NLRP3 inflammasome, MAP Kinase, TGFβ and TLR5 signaling pathways. Taken together, the elucidation of the landscape of signature miRNA regulatory networks in neuropathic pain will facilitate the discovery of novel miRNA/target biomarkers for more effective management of neuropathic pain.
Collapse
|
27
|
Benameur T, Panaro MA, Porro C. Exosomes and their Cargo as a New Avenue for Brain and Treatment of CNS-Related Diseases. Open Neurol J 2022. [DOI: 10.2174/1874205x-v16-e2201190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Extracellular Vesicles (EVs), which belong to nanoscale vesicles, including microvesicles (MVs) and exosomes, are now considered a new important tool for intercellular neuronal communication in the Central Nervous System (CNS) under physiological and pathological conditions. EVs are shed into blood, peripheral body fluids and cerebrospinal fluid (CSF) by a large variety of cells.
EVs can act locally on neighboring and distant cells. EVs represent the fingerprints of the originating cells and can carry a variety of molecular constituents of their cell of origin, including protein, lipids, DNA and microRNAs (miRNAs).
The most studied EVs are the exosomes because they are ubiquitous and have the capacity to transfer cell-derived components and bioactive molecules to target cells. In this minireview, we focused on cell-cell communication in CNS mediated by exosomes and their important cargo as an innovative way to treat or follow up with CNS diseases.
Collapse
|
28
|
Sadri M, Hirosawa N, Le J, Romero H, Martellucci S, Kwon HJ, Pizzo D, Ohtori S, Gonias SL, Campana WM. Tumor necrosis factor receptor-1 is selectively sequestered into Schwann cell extracellular vesicles where it functions as a TNFα decoy. Glia 2022; 70:256-272. [PMID: 34559433 PMCID: PMC10656730 DOI: 10.1002/glia.24098] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 12/19/2022]
Abstract
Schwann cells (SCs) are known to produce extracellular vesicles (EV) that participate in cell-cell communication by transferring cargo to target cells, including mRNAs, microRNAs, and biologically active proteins. Herein, we report a novel mechanism whereby SC EVs may regulate PNS physiology, especially in injury, by controlling the activity of TNFα. SCs actively sequester tumor necrosis factor receptor-1 (TNFR1) into EVs at high density, accounting for about 2% of the total protein in SC EVs (~1000 copies TNFR1/EV). Although TNFR2 was robustly expressed by SCs in culture, TNFR2 was excluded from SC EVs. SC EV TNFR1 bound TNFα, decreasing the concentration of free TNFα available to bind to cells and thus served as a TNFα decoy. SC EV TNFR1 significantly inhibited TNFα-induced p38 MAPK phosphorylation in cultured SCs. When TNFR1 was proteolytically removed from SC EVs using tumor necrosis factor-α converting enzyme (TACE) or neutralized with antibody, the ability of TNFα to activate p38 MAPK in the presence of these EVs was restored. As further evidence of its decoy activity, SC EV TNFR1 modified TNFα activities in vitro including: (1) regulation of expression of other cytokines; (2) effects on SC morphology; and (3) effects on SC viability. SC EVs also modified the effects of TNFα on sciatic nerve morphology and neuropathic pain-related behavior in vivo. By sequestering TNFR1 in EVs, SCs may buffer against the potentially toxic effects of TNFα. SC EVs provide a novel mechanism for the spatial and temporal regulation of neuro-inflammation.
Collapse
Affiliation(s)
- Mahrou Sadri
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA
| | - Naoya Hirosawa
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA
- Department of Orthopaedic Surgery and Graduate School in Medicine, Chiba University, Chiba, Japan
| | - Jasmine Le
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA
- Veterans Administration San Diego Healthcare System, San Diego, California, USA
| | - Haylie Romero
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA
- Program in Neuroscience, University of California, San Diego, La Jolla, California, USA
| | - Stefano Martellucci
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA
| | - Hyo Jun Kwon
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA
| | - Donald Pizzo
- Department of Pathology, University of California, San Diego, California, USA
| | - Seiji Ohtori
- Department of Orthopaedic Surgery and Graduate School in Medicine, Chiba University, Chiba, Japan
| | - Steven L. Gonias
- Department of Pathology, University of California, San Diego, California, USA
| | - Wendy M. Campana
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA
- Veterans Administration San Diego Healthcare System, San Diego, California, USA
- Program in Neuroscience, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
29
|
Liu YP, Yang YD, Mou FF, Zhu J, Li H, Zhao TT, Zhao Y, Shao SJ, Cui GH, Guo HD. Exosome-Mediated miR-21 Was Involved in the Promotion of Structural and Functional Recovery Effect Produced by Electroacupuncture in Sciatic Nerve Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7530102. [PMID: 35132352 PMCID: PMC8817850 DOI: 10.1155/2022/7530102] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/24/2021] [Accepted: 01/05/2022] [Indexed: 12/11/2022]
Abstract
PURPOSE Our study is aimed at investigating the mechanism by which electroacupuncture (EA) promoted nerve regeneration by regulating the release of exosomes and exosome-mediated miRNA-21 (miR-21) transmission. Furthermore, the effects of Schwann cells- (SC-) derived exosomes on the overexpression of miR-21 for the treatment of PNI were investigated. METHODS A sciatic nerve injury model of rat was constructed, and the expression of miR-21 in serum exosomes and damaged local nerves was detected using RT-qPCR after EA treatment. The exosomes were identified under a transmission electron microscope and using western blotting analysis. Then, the exosome release inhibitor, GW4869, and the miR-21-5p-sponge used for the knockdown of miR-21 were used to clarify the effects of exosomal miR-21 on nerve regeneration promoted by EA. The nerve conduction velocity recovery rate, sciatic nerve function index, and wet weight ratio of gastrocnemius muscle were determined to evaluate sciatic nerve function recovery. SC proliferation and the level of neurotrophic factors were assessed using immunofluorescence staining, and the expression levels of SPRY2 and miR-21 were detected using RT-qPCR analysis. Subsequently, the transmission of exosomal miR-21 from SC to the axon was verified in vitro. Finally, the exosomes derived from the SC infected with the miR-21 overexpression lentivirus were collected and used to treat the rat SNI model to explore the therapeutic role of SC-derived exosomes overexpressing miR-21. RESULTS We found that EA inhibited the release of serum exosomal miR-21 in a PNI model of rats during the early stage of PNI, while it promoted its release during later stages. EA enhanced the accumulation of miR-21 in the injured nerve and effectively promoted the recovery of nerve function after PNI. The treatment effect of EA was attenuated when the release of circulating exosomes was inhibited or when miR-21 was downregulated in local injury tissue via the miR-21-5p-sponge. Normal exosomes secreted by SC exhibited the ability to promote the recovery of nerve function, while the overexpression of miR-21 enhanced the effects of the exosomes. In addition, exosomal miR-21 secreted by SC could promote neurite outgrowth in vitro. CONCLUSION Our results demonstrated the mechanism of EA on PNI from the perspective of exosome-mediated miR-21 transport and provided a theoretical basis for the use of exosomal miR-21 as a novel strategy for the treatment of PNI.
Collapse
Affiliation(s)
- Yu-pu Liu
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yi-duo Yang
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fang-fang Mou
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jing Zhu
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Han Li
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tian-tian Zhao
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yue Zhao
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shui-jin Shao
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Guo-hong Cui
- Department of Neurology, Shanghai No. 9 People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Hai-dong Guo
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
30
|
Jones JI, Costa CJ, Cooney C, Goldberg DC, Ponticiello M, Cohen MW, Mellado W, Ma TC, Willis DE. Failure to Upregulate the RNA Binding Protein ZBP After Injury Leads to Impaired Regeneration in a Rodent Model of Diabetic Peripheral Neuropathy. Front Mol Neurosci 2021; 14:728163. [PMID: 34949989 PMCID: PMC8688773 DOI: 10.3389/fnmol.2021.728163] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/16/2021] [Indexed: 12/03/2022] Open
Abstract
Most diabetes patients eventually suffer from peripheral nerve degeneration. Unfortunately, there is no treatment for the condition and its mechanisms are not well understood. There is, however, an emerging consensus that the inability of peripheral nerves to regenerate normally after injury contributes to the pathophysiology. We have previously shown that regeneration of peripheral axons requires local axonal translation of a pool of axonal mRNAs and that the levels and members of this axonal mRNA pool are altered in response to injury. Here, we show that following sciatic nerve injury in a streptozotocin rodent model of type I diabetes, this mobilization of RNAs into the injured axons is attenuated and correlates with decreased axonal regeneration. This failure of axonal RNA localization results from decreased levels of the RNA binding protein ZBP1. Over-expression of ZBP1 rescues the in vitro growth defect in injured dorsal root ganglion neurons from diabetic rodents. These results provide evidence that decreased neuronal responsiveness to injury in diabetes is due to a decreased ability to alter the pool of axonal mRNAs available for local translation, and may open new therapeutic opportunities for diabetic peripheral neuropathy.
Collapse
Affiliation(s)
- James I Jones
- Burke Neurological Institute, White Plains, NY, United States
| | | | - Caitlin Cooney
- Burke Neurological Institute, White Plains, NY, United States
| | | | | | - Melanie W Cohen
- Burke Neurological Institute, White Plains, NY, United States
| | | | - Thong C Ma
- Burke Neurological Institute, White Plains, NY, United States.,Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Dianna E Willis
- Burke Neurological Institute, White Plains, NY, United States.,Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
31
|
Li X, Shi S, Jing D, Li X, Zhang B, Bie Q. Signal transduction mechanism of exosomes in diabetic complications (Review). Exp Ther Med 2021; 23:155. [PMID: 35069836 DOI: 10.3892/etm.2021.11078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/24/2021] [Indexed: 11/05/2022] Open
Affiliation(s)
- Xueting Li
- Department of Clinical Medicine, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Shuo Shi
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Dehuai Jing
- Department of Digestive Endoscopy and 4Nephrology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Xinjian Li
- Department of Nephrology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Bin Zhang
- Department of Clinical Medicine, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Qingli Bie
- Department of Clinical Medicine, Jining Medical University, Jining, Shandong 272000, P.R. China
| |
Collapse
|
32
|
Myelination, axonal loss and Schwann cell characteristics in axonal polyneuropathy compared to controls. PLoS One 2021; 16:e0259654. [PMID: 34735549 PMCID: PMC8568174 DOI: 10.1371/journal.pone.0259654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 10/23/2021] [Indexed: 12/12/2022] Open
Abstract
Introduction Polyneuropathy is a debilitating condition characterized by distal sensory and motor deficits. Schwann cell dysfunction and axonal loss are integral factors in pathophysiology and disease progression of polyneuropathy. Aims The aim of this study was the assessment of Schwann cell characteristics, nerve fibers and myelination parameters in polyneuropathy patients compared to controls. Methods Nerve tissue was obtained from polyneuropathy patients (n = 10) undergoing diagnostic sural nerve biopsies. Biopsies of healthy peripheral nerves (n = 5) were harvested during elective sural nerve grafting for chronic peripheral nerve lesions. Exclusion criteria for the healthy control group were recent neurological trauma, diabetes, neurological and cardiovascular disease, as well as active malignancies and cytotoxic medication within the last 12 months. The over-all architecture of nerve sections and myelination parameters were histomorphometrically analyzed. Immunofluorescent imaging was used to evaluate Schwann cell phenotypes, senescence markers and myelination parameters. Results Histomorphometric analysis of nerve biopsies showed significant axonal loss in polyneuropathy patients compared to controls, which was in accordance with the neuropathological findings. Immunofluorescent staining of Schwann cells and myelin basic protein indicated a significant impairment of myelination and lower Schwann cell counts compared to controls. Phenotypic alterations and increased numbers of non-myelinating p75-positive Schwann cells were found in polyneuropathy patients. Discussion This study provided quantitative data of axonal loss, reduced myelination and Schwann cell dysfunction of polyneuropathy patients compared to neurologically healthy controls. Phenotypic alterations of Schwann cells were similar to those seen after peripheral nerve injury, highlighting the clinical relevance of Schwann cell dysfunction.
Collapse
|
33
|
Anakor E, Le Gall L, Dumonceaux J, Duddy WJ, Duguez S. Exosomes in Ageing and Motor Neurone Disease: Biogenesis, Uptake Mechanisms, Modifications in Disease and Uses in the Development of Biomarkers and Therapeutics. Cells 2021; 10:2930. [PMID: 34831153 PMCID: PMC8616058 DOI: 10.3390/cells10112930] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 02/07/2023] Open
Abstract
Intercellular communication between neurons and their surrounding cells occurs through the secretion of soluble molecules or release of vesicles such as exosomes into the extracellular space, participating in brain homeostasis. Under neuro-degenerative conditions associated with ageing, such as amyotrophic lateral sclerosis (ALS), Alzheimer's or Parkinson's disease, exosomes are suspected to propagate toxic proteins. The topic of this review is the role of exosomes in ageing conditions and more specifically in ALS. Our current understanding of exosomes and exosome-related mechanisms is first summarized in a general sense, including their biogenesis and secretion, heterogeneity, cellular interaction and intracellular fate. Their role in the Central Nervous System (CNS) and ageing of the neuromotor system is then considered in the context of exosome-induced signaling. The review then focuses on exosomes in age-associated neurodegenerative disease. The role of exosomes in ALS is highlighted, and their use as potential biomarkers to diagnose and prognose ALS is presented. The therapeutic implications of exosomes for ALS are considered, whether as delivery vehicles, neurotoxic targets or as corrective drugs in and of themselves. A diverse set of mechanisms underpin the functional roles, both confirmed and potential, of exosomes, generally in ageing and specifically in motor neurone disease. Aspects of their contents, biogenesis, uptake and modifications offer many plausible routes towards the development of novel biomarkers and therapeutics.
Collapse
Affiliation(s)
- Ekene Anakor
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry-Londonderry BT47 6SB, UK; (E.A.); (L.L.G.); (J.D.); (W.J.D.)
| | - Laura Le Gall
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry-Londonderry BT47 6SB, UK; (E.A.); (L.L.G.); (J.D.); (W.J.D.)
- NIHR Biomedical Research Centre, Great Ormond Street Institute of Child Health, Great Ormond Street Hospital NHS Trust, University College London, London WC1N 1EH, UK
| | - Julie Dumonceaux
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry-Londonderry BT47 6SB, UK; (E.A.); (L.L.G.); (J.D.); (W.J.D.)
- NIHR Biomedical Research Centre, Great Ormond Street Institute of Child Health, Great Ormond Street Hospital NHS Trust, University College London, London WC1N 1EH, UK
| | - William John Duddy
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry-Londonderry BT47 6SB, UK; (E.A.); (L.L.G.); (J.D.); (W.J.D.)
| | - Stephanie Duguez
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry-Londonderry BT47 6SB, UK; (E.A.); (L.L.G.); (J.D.); (W.J.D.)
| |
Collapse
|
34
|
Wong FC, Ye L, Demir IE, Kahlert C. Schwann cell-derived exosomes: Janus-faced mediators of regeneration and disease. Glia 2021; 70:20-34. [PMID: 34519370 DOI: 10.1002/glia.24087] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 12/20/2022]
Abstract
The phenotypic plasticity of Schwann cells (SCs) has contributed to the regenerative potential of the peripheral nervous system (PNS), but also pathological processes. This double-sided effect has led to an increasing attention to the role of extracellular vesicles (EVs) or exosomes in SCs to examine the intercellular communication between SCs and their surroundings. Here, we first describe the current knowledge of SC and EV biology, which forms the basis for the updates on advances in SC-derived exosomes research. We seek to explore in-depth the exosome-mediated molecular mechanisms involved in the regulation of SCs and their microenvironment. This review concludes with potential applications of SC-derived exosomes as delivery vehicles for therapeutics and biomarkers. The goal of this review is to emphasize the crucial role of SC-derived exosomes in the functional integration of the PNS, highlighting an emerging area in which there is much to explore and re-explore.
Collapse
Affiliation(s)
- Fang Cheng Wong
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Linhan Ye
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany.,Germany German Cancer Consortium (DKTK), Partner Site, Munich, Germany.,CRC 1321 Modelling and Targeting Pancreatic Cancer, Munich, Germany
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany.,Germany German Cancer Consortium (DKTK), Partner Site, Munich, Germany.,Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.,CRC 1321 Modelling and Targeting Pancreatic Cancer, Munich, Germany.,Else Kröner Clinician Scientist Professor for "Translational Pancreatic Surgery
| | - Christoph Kahlert
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
| |
Collapse
|
35
|
He X, Kuang G, Wu Y, Ou C. Emerging roles of exosomal miRNAs in diabetes mellitus. Clin Transl Med 2021; 11:e468. [PMID: 34185424 PMCID: PMC8236118 DOI: 10.1002/ctm2.468] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Exosomes are small extracellular vesicles 40-160 nm in diameter that are secreted by almost all cell types. Exosomes can carry diverse cargo including RNA, DNA, lipids, proteins, and metabolites. Exosomes transfer substances and information between cells by circulating in body fluids and are thus involved in diverse physiological and pathological processes in the human body. Recent studies have closely associated exosomal microRNAs (miRNAs) with various human diseases, including diabetes mellitus (DM), which is a complex multifactorial metabolic disorder disease. Exosomal miRNAs are emerging as pivotal regulators in the progression of DM, mainly in terms of pancreatic β-cell injury and insulin resistance. Exosomal miRNAs are closely associated with DM-associated complications, such as diabetic retinopathy (DR), diabetic nephropathy (DN), and diabetic cardiomyopathy (DCM), etc. Further investigations of the mechanisms of action of exosomal miRNAs and their role in DM will be valuable for the thorough understanding of the physiopathological process of DM. Here, we have summarized recent findings regarding exosomal miRNAs associated with DM to provide a new strategy for identifying potential diagnostic biomarkers and drug targets for the early diagnosis and treatment, respectively, of DM.
Collapse
Affiliation(s)
- Xiaoyun He
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunan410008China
- Departments of Ultrasound Imaging, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Gaoyan Kuang
- Department of OrthopedicsThe First Affiliated Hospital of Hunan University of Chinese MedicineChangshaHunan410007China
- Postdoctoral Research WorkstationHinye Pharmaceutical Co. LtdChangshaHunan410331China
| | - Yongrong Wu
- Hunan university of Chinese MedicineChangshaHunan410208China
| | - Chunlin Ou
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaHunan410008China
| |
Collapse
|
36
|
Wang H, Davison M, Wang K, Xia TH, Call KM, Luo J, Wu X, Zuccarino R, Bacha A, Bai Y, Gutmann L, Feely SME, Grider T, Rossor AM, Reilly MM, Shy ME, Svaren J. MicroRNAs as Biomarkers of Charcot-Marie-Tooth Disease Type 1A. Neurology 2021; 97:e489-e500. [PMID: 34031204 DOI: 10.1212/wnl.0000000000012266] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/26/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To determine whether microRNAs (miRs) are elevated in the plasma of individuals with the inherited peripheral neuropathy Charcot-Marie-Tooth disease type 1A (CMT1A), miR profiling was employed to compare control and CMT1A plasma. METHODS We performed a screen of CMT1A and control plasma samples to identify miRs that are elevated in CMT1A using next-generation sequencing, followed by validation of selected miRs by quantitative PCR, and correlation with protein biomarkers and clinical data: Rasch-modified CMT Examination and Neuropathy Scores, ulnar compound muscle action potentials, and motor nerve conduction velocities. RESULTS After an initial pilot screen, a broader screen confirmed elevated levels of several muscle-associated miRNAs (miR1, -133a, -133b, and -206, known as myomiRs) along with a set of miRs that are highly expressed in Schwann cells of peripheral nerve. Comparison to other candidate biomarkers for CMT1A (e.g., neurofilament light) measured on the same sample set shows a comparable elevation of several miRs (e.g., miR133a, -206, -223) and ability to discriminate cases from controls. Neurofilament light levels were most highly correlated with miR133a. In addition, the putative Schwann cell miRs (e.g., miR223, -199a, -328, -409, -431) correlate with the recently described transmembrane protease serine 5 (TMPRSS5) protein biomarker that is most highly expressed in Schwann cells and also elevated in CMT1A plasma. CONCLUSIONS These studies identify a set of miRs that are candidate biomarkers for clinical trials in CMT1A. Some of the miRs may reflect Schwann cell processes that underlie the pathogenesis of the disease. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that a set of plasma miRs are elevated in patients with CMT1A.
Collapse
Affiliation(s)
- Hongge Wang
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Matthew Davison
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Kathryn Wang
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Tai-He Xia
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Katherine M Call
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Jun Luo
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Xingyao Wu
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Riccardo Zuccarino
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Alexa Bacha
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Yunhong Bai
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Laurie Gutmann
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Shawna M E Feely
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Tiffany Grider
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Alexander M Rossor
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Mary M Reilly
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - Michael E Shy
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison
| | - John Svaren
- From Translational Sciences (H.W., M.D., K.W., T.X., K.M.C.), Sanofi Research; Biostatistics and Programming (J.L.), Sanofi Development, Framingham, MA; Department of Neurology (X.W., R.Z., A.B., Y.B., L.G., S.M.E.F., T.G., M.E.S.), Carver College of Medicine, University of Iowa, Iowa City; Department of Neuromuscular Diseases (A.M.R., M.M.R.), UCL Queen Square Institute of Neurology and National Hospital for Neurology and Neurosurgery, University College London, UK; and Waisman Center and Department of Comparative Biosciences (J.S.), University of Wisconsin, Madison.
| |
Collapse
|
37
|
Hosseini M, Roshangar L, Raeisi S, Ghahremanzadeh K, Negargar S, Tarmahi V, Hosseini V, Raeisi M, Rahimi E, Ebadi Z. The Therapeutic Applications of Exosomes in Different Types of Diseases: A Review. Curr Mol Med 2021; 21:87-95. [PMID: 32520687 DOI: 10.2174/1566524020666200610164743] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 11/22/2022]
Abstract
Exosomes are nano-sized vesicles secreted by nearly all cells and have received massive attention recently. In addition to their roles in pathophysiological processes and diagnostic evaluations, recently, several studies have applied exosomes to design novel therapeutic applications. Exosomes can be derived from a variety of cells and tissues and based on the source, they can carry different native contents such as DNAs, non-coding small RNAs, mRNAs, and proteins. They can also be engineered by adding desirable agents including specific biomolecules or drugs. Both forms can be therapeutically used for delivering their cargoes to the target cells and desirably alter their functions. The present study aimed to provide a comprehensive review of the various studies which applied exosomes as a therapeutic tool in the treatment of different types of diseases including cancer, cardiovascular, neurologic, psychiatric, liver, and kidney diseases.
Collapse
Affiliation(s)
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Raeisi
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kazem Ghahremanzadeh
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sohrab Negargar
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Tarmahi
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Hosseini
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Raeisi
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elnaz Rahimi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zakiyeh Ebadi
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
38
|
Zhang Y, Li C, Qin Y, Cepparulo P, Millman M, Chopp M, Kemper A, Szalad A, Lu X, Wang L, Zhang ZG. Small extracellular vesicles ameliorate peripheral neuropathy and enhance chemotherapy of oxaliplatin on ovarian cancer. J Extracell Vesicles 2021; 10:e12073. [PMID: 33728031 PMCID: PMC7931803 DOI: 10.1002/jev2.12073] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 01/07/2021] [Accepted: 02/13/2021] [Indexed: 12/17/2022] Open
Abstract
There are no effective treatments for chemotherapy induced peripheral neuropathy (CIPN). Small extracellular vesicles (sEVs) facilitate intercellular communication and mediate nerve function and tumour progression. We found that the treatment of mice bearing ovarian tumour with sEVs derived from cerebral endothelial cells (CEC-sEVs) in combination with a chemo-drug, oxaliplatin, robustly reduced oxaliplatin-induced CIPN by decreasing oxaliplatin-damaged myelination and nerve fibres of the sciatic nerve and significantly amplified chemotherapy of oxaliplatin by reducing tumour size. The combination therapy substantially increased a set of sEV cargo-enriched miRNAs, but significantly reduced oxaliplatin-increased proteins in the sciatic nerve and tumour tissues. Bioinformatics analysis revealed the altered miRNAs and proteins formed two distinct networks that regulate neuropathy and tumour growth, respectively. Intravenously administered CEC-sEVs were internalized by axons of the sciatic nerve and cancer cells. Reduction of CEC-sEV cargo miRNAs abolished the effects of CEC-sEVs on oxaliplatin-inhibited axonal growth and on amplification of the anti-cancer effect in ovarian cancer cells, suggesting that alterations in the networks of miRNAs and proteins in recipient cells contribute to the therapeutic effect of CEC-sEVs on CIPN. Together, the present study demonstrates that CEC-sEVs suppressed CIPN and enhanced chemotherapy of oxaliplatin in the mouse bearing ovarian tumour.
Collapse
Affiliation(s)
- Yi Zhang
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | - Chao Li
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | - Yi Qin
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | | | | | - Michael Chopp
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
- Department of PhysicsOakland UniversityRochesterMichiganUSA
| | - Amy Kemper
- Department of PathologyHenry Ford Health SystemDetroitMichiganUSA
| | - Alexandra Szalad
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | - Xuerong Lu
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | - Lei Wang
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | - Zheng Gang Zhang
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| |
Collapse
|
39
|
Zhu X, Chen Y, Xu X, Xu X, Lu Y, Huang X, Zhou J, Hu L, Wang J, Shen X. SP6616 as a Kv2.1 inhibitor efficiently ameliorates peripheral neuropathy in diabetic mice. EBioMedicine 2020; 61:103061. [PMID: 33096484 PMCID: PMC7581884 DOI: 10.1016/j.ebiom.2020.103061] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 09/20/2020] [Accepted: 09/24/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Diabetic peripheral neuropathy (DPN) is a common complication of diabetes severely afflicting the patients, while there is yet no effective medication against this disease. As Kv2.1 channel functions potently in regulating neurological disorders, the present work was to investigate the regulation of Kv2.1 channel against DPN-like pathology of DPN model mice by using selective Kv2.1 inhibitor SP6616 (ethyl 5-(3-ethoxy-4-methoxyphenyl)-2-(4-hydroxy-3-methoxybenzylidene)-7-methyl-3-oxo-2,3-dihydro-5H-[1,3]thiazolo[3,2-a]pyrimidine-6-carboxylate) as a probe. METHODS STZ-induced type 1 diabetic mice with DPN (STZ mice) were defined at 12 weeks of age (4 weeks after STZ injection) through behavioral tests, and db/db (BKS Cg-m+/+Leprdb/J) type 2 diabetic mice with DPN (db/db mice) were at 18 weeks of age. SP6616 was administered daily via intraperitoneal injection for 4 weeks. The mechanisms underlying the amelioration of SP6616 on DPN-like pathology were investigated by RT-PCR, western blot and immunohistochemistry technical approaches against diabetic mice, and verified against the STZ mice with Kv2.1 knockdown in dorsal root ganglion (DRG) tissue by injection of adeno associated virus AAV9-Kv2.1-RNAi. Amelioration of SP6616 on the pathological behaviors of diabetic mice was assessed against tactile allodynia, thermal sensitivity and motor nerve conduction velocity (MNCV). FINDINGS SP6616 treatment effectively ameliorated the threshold of mechanical stimuli, thermal sensitivity and MNCV of diabetic mice. Mechanism research results indicated that SP6616 suppressed Kv2.1 expression, increased the number of intraepidermal nerve fibers (IENFs), improved peripheral nerve structure and vascular function in DRG tissue. In addition, SP6616 improved mitochondrial dysfunction through Kv2.1/CaMKKβ/AMPK/PGC-1α pathway, repressed inflammatory response by inhibiting Kv2.1/NF-κB signaling and alleviated apoptosis of DRG neuron through Kv2.1-mediated regulation of Bcl-2 family proteins and Caspase-3 in diabetic mice. INTERPRETATION Our work has highly supported the beneficial of Kv2.1 inhibition in ameliorating DPN-like pathology and highlighted the potential of SP6616 in the treatment of DPN. FUNDING Please see funding sources.
Collapse
Affiliation(s)
- Xialin Zhu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yun Chen
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China
| | - Xu Xu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaoju Xu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xi Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinpei Zhou
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, China.
| | - Lihong Hu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiaying Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xu Shen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
40
|
Gonçalves NP, Yan Y, Ulrichsen M, Venø MT, Poulsen ET, Enghild JJ, Kjems J, Vægter CB. Modulation of Small RNA Signatures in Schwann-Cell-Derived Extracellular Vesicles by the p75 Neurotrophin Receptor and Sortilin. Biomedicines 2020; 8:E450. [PMID: 33114403 PMCID: PMC7694014 DOI: 10.3390/biomedicines8110450] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Schwann cells (SCs) are the main glial cells of the peripheral nervous system (PNS) and are known to be involved in various pathophysiological processes, such as diabetic neuropathy and nerve regeneration, through neurotrophin signaling. Such glial trophic support to axons, as well as neuronal survival/death signaling, has previously been linked to the p75 neurotrophin receptor (p75NTR) and its co-receptor Sortilin. Recently, SC-derived extracellular vesicles (EVs) were shown to be important for axon growth and nerve regeneration, but cargo of these glial cell-derived EVs has not yet been well-characterized. In this study, we aimed to characterize signatures of small RNAs in EVs derived from wild-type (WT) SCs and define differentially expressed small RNAs in EVs derived from SCs with genetic deletions of p75NTR (Ngfr-/-) or Sortilin (Sort1-/-). Using RNA sequencing, we identified a total of 366 miRNAs in EVs derived from WT SCs of which the most highly expressed are linked to the regulation of axonogenesis, axon guidance and axon extension, suggesting an involvement of SC EVs in axonal homeostasis. Signaling of SC EVs to non-neuronal cells was also suggested by the presence of several miRNAs important for regulation of the endothelial cell apoptotic process. Ablated p75NTR or sortilin expression in SCs translated into a set of differentially regulated tRNAs and miRNAs, with impact in autophagy and several cellular signaling pathways such as the phosphatidylinositol signaling system. With this work, we identified the global expression profile of small RNAs present in SC-derived EVs and provided evidence for a regulatory function of these vesicles on the homeostasis of other cell types of the PNS. Differentially identified miRNAs can pave the way to a better understanding of p75NTR and sortilin roles regarding PNS homeostasis and disease.
Collapse
Affiliation(s)
- Nádia P. Gonçalves
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (M.U.); (C.B.V.)
| | - Yan Yan
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (M.T.V.); (J.K.)
- Omiics ApS, 8000 Aarhus, Denmark
| | - Maj Ulrichsen
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (M.U.); (C.B.V.)
| | - Morten T. Venø
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (M.T.V.); (J.K.)
- Omiics ApS, 8000 Aarhus, Denmark
| | - Ebbe T. Poulsen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (E.T.P.); (J.J.E.)
| | - Jan J. Enghild
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (E.T.P.); (J.J.E.)
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (M.T.V.); (J.K.)
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (E.T.P.); (J.J.E.)
| | - Christian B. Vægter
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (M.U.); (C.B.V.)
| |
Collapse
|
41
|
Fan B, Chopp M, Zhang ZG, Liu XS. Emerging Roles of microRNAs as Biomarkers and Therapeutic Targets for Diabetic Neuropathy. Front Neurol 2020; 11:558758. [PMID: 33192992 PMCID: PMC7642849 DOI: 10.3389/fneur.2020.558758] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetic neuropathy (DN) is the most prevalent chronic complication of diabetes mellitus. The exact pathophysiological mechanisms of DN are unclear; however, communication network dysfunction among axons, Schwann cells, and the microvascular endothelium likely play an important role in the development of DN. Mounting evidence suggests that microRNAs (miRNAs) act as messengers that facilitate intercellular communication and may contribute to the pathogenesis of DN. Deregulation of miRNAs is among the initial molecular alterations observed in diabetics. As such, miRNAs hold promise as biomarkers and therapeutic targets. In preclinical studies, miRNA-based treatment of DN has shown evidence of therapeutic potential. But this therapy has been hampered by miRNA instability, targeting specificity, and potential toxicities. Recent findings reveal that when packaged within extracellular vesicles, miRNAs are resistant to degradation, and their delivery efficiency and therapeutic potential is markedly enhanced. Here, we review the latest research progress on the roles of miRNAs as biomarkers and as potential clinical therapeutic targets in DN. We also discuss the promise of exosomal miRNAs as therapeutics and provide recommendations for future research on miRNA-based medicine.
Collapse
Affiliation(s)
- Baoyan Fan
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States.,Department of Physics, Oakland University, Rochester, MI, United States
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| | - Xian Shuang Liu
- Department of Neurology, Henry Ford Health System, Detroit, MI, United States
| |
Collapse
|
42
|
The roles of MicroRNAs in neural regenerative medicine. Exp Neurol 2020; 332:113394. [DOI: 10.1016/j.expneurol.2020.113394] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/15/2020] [Accepted: 06/25/2020] [Indexed: 12/22/2022]
|
43
|
Exosome: A New Player in Translational Nanomedicine. J Clin Med 2020; 9:jcm9082380. [PMID: 32722531 PMCID: PMC7463834 DOI: 10.3390/jcm9082380] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
Summary: Exosomes are extracellular vesicles released by the vast majority of cell types both in vivo and ex vivo, upon the fusion of multivesicular bodies (MVBs) with the cellular plasma membrane. Two main functions have been attributed to exosomes: their capacity to transport proteins, lipids and nucleic acids between cells and organs, as well as their potential to act as natural intercellular communicators in normal biological processes and in pathologies. From a clinical perspective, the majority of applications use exosomes as biomarkers of disease. A new approach uses exosomes as biologically active carriers to provide a platform for the enhanced delivery of cargo in vivo. One of the major limitations in developing exosome-based therapies is the difficulty of producing sufficient amounts of safe and efficient exosomes. The identification of potential proteins involved in exosome biogenesis is expected to directly cause a deliberate increase in exosome production. In this review, we summarize the current state of knowledge regarding exosomes, with particular emphasis on their structural features, biosynthesis pathways, production techniques and potential clinical applications.
Collapse
|
44
|
Gonçalves NP, Jager SE, Richner M, Murray SS, Mohseni S, Jensen TS, Vaegter CB. Schwann cell p75 neurotrophin receptor modulates small fiber degeneration in diabetic neuropathy. Glia 2020; 68:2725-2743. [PMID: 32658363 DOI: 10.1002/glia.23881] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/10/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023]
Abstract
Diabetic neuropathy has an incidence as high as 50% of diabetic patients and is characterized by damage to neurons, Schwann cells and blood vessels within the peripheral nervous system. The low-affinity neurotrophin receptor p75 (p75NTR ), particularly expressed by the Schwann cells in the peripheral nerve, has previously been reported to play a role in developmental myelination and cell survival/death. Increased levels of p75NTR , in the endoneurium and plasma from diabetic patients and rodent models of disease, have been observed, proposing that this receptor might be involved in the pathogenesis of diabetic neuropathy. Therefore, in this study, we addressed this hypothesis by utilizing a mouse model of selective nerve growth factor receptor (Ngfr) deletion in Schwann cells (SC-p75NTR -KO). Electron microscopy of sciatic nerves from mice with high fat diet induced obesity demonstrated how loss of Schwann cell-p75NTR aggravated axonal atrophy and loss of C-fibers. RNA sequencing disclosed several pre-clinical signaling alterations in the diabetic peripheral nerves, dependent on Schwann cell p75NTR signaling, specially related with lysosome, phagosome, and immune pathways. Morphological and biochemical analyses identified abundant lysosomes and autophagosomes in the C-fiber axoplasm of the diabetic SC-p75NTR -KO nerves, which together with increased Cathepsin B protein levels corroborates gene upregulation from the phagolysosomal pathways. Altogether, this study demonstrates that Schwann cell p75NTR deficiency amplifies diabetic neuropathy disease by triggering overactivation of immune-related pathways and increased lysosomal stress.
Collapse
Affiliation(s)
- Nádia P Gonçalves
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience-DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus C, Denmark.,International Diabetic Neuropathy Consortium (IDNC), Aarhus University Hospital, Aarhus N, Denmark
| | - Sara E Jager
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience-DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus C, Denmark.,Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Mette Richner
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience-DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus C, Denmark
| | - Simon S Murray
- Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria, Australia
| | - Simin Mohseni
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Troels S Jensen
- International Diabetic Neuropathy Consortium (IDNC), Aarhus University Hospital, Aarhus N, Denmark.,Department of Neurology and Danish Pain Research Center, Aarhus University, Aarhus C, Denmark
| | - Christian B Vaegter
- Department of Biomedicine, Danish Research Institute of Translational Neuroscience-DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus C, Denmark.,International Diabetic Neuropathy Consortium (IDNC), Aarhus University Hospital, Aarhus N, Denmark
| |
Collapse
|
45
|
Wang L, Chopp M, Szalad A, Lu X, Zhang Y, Wang X, Cepparulo P, Lu M, Li C, Zhang ZG. Exosomes Derived From Schwann Cells Ameliorate Peripheral Neuropathy in Type 2 Diabetic Mice. Diabetes 2020; 69:749-759. [PMID: 31915154 PMCID: PMC7085247 DOI: 10.2337/db19-0432] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 12/31/2019] [Indexed: 12/23/2022]
Abstract
Schwann cell-derived exosomes communicate with dorsal root ganglia (DRG) neurons. The current study investigated the therapeutic effect of exosomes derived from healthy Schwann cells (SC-Exos) on diabetic peripheral neuropathy (DPN). We found that intravenous administration of SC-Exos to type 2 diabetic db/db mice with peripheral neuropathy remarkably ameliorated DPN by improving sciatic nerve conduction velocity and increasing thermal and mechanical sensitivity. These functional improvements were associated with the augmentation of epidermal nerve fibers and remyelination of sciatic nerves. Quantitative RT-PCR and Western blot analysis of sciatic nerve tissues showed that SC-Exo treatment reversed diabetes-reduced mature form of miRNA (miR)-21, -27a, and -146a and diabetes-increased semaphorin 6A (SEMA6A); Ras homolog gene family, member A (RhoA); phosphatase and tensin homolog (PTEN); and nuclear factor-κB (NF-κB). In vitro data showed that SC-Exos promoted neurite outgrowth of diabetic DRG neurons and migration of Schwann cells challenged by high glucose. Collectively, these novel data provide evidence that SC-Exos have a therapeutic effect on DPN in mice and suggest that SC-Exo modulation of miRs contributes to this therapy.
Collapse
Affiliation(s)
- Lei Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI
- Department of Physics, Oakland University, Rochester, MI
| | | | - XueRong Lu
- Department of Neurology, Henry Ford Hospital, Detroit, MI
| | - Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI
| | - Xinli Wang
- Department of Neurology, Henry Ford Hospital, Detroit, MI
| | | | - Mei Lu
- Department of Biostatistics and Research Epidemiology, Henry Ford Hospital, Detroit, MI
| | - Chao Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI
| | | |
Collapse
|
46
|
Liu YP, Shao SJ, Guo HD. Schwann cells apoptosis is induced by high glucose in diabetic peripheral neuropathy. Life Sci 2020; 248:117459. [PMID: 32092332 DOI: 10.1016/j.lfs.2020.117459] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 02/14/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
Abstract
Diabetic peripheral neuropathy (DPN) is a common complication of diabetes mellitus that affects approximately half of patients with diabetes. Current treatment regimens cannot treat DPN effectively. Schwann cells (SCs) are very sensitive to glucose concentration and insulin, and closely associated with the occurrence and development of type 1 diabetic mellitus (T1DM) and DPN. Apoptosis of SCs is induced by hyperglycemia and is involved in the pathogenesis of DPN. This review considers the pathological processes of SCs apoptosis under high glucose, which include the following: oxidative stress, inflammatory reactions, endoplasmic reticulum stress, autophagy, nitrification and signaling pathways (PI3K/AKT, ERK, PERK/Nrf2, and Wnt/β-catenin). The clarification of mechanisms underlying SCs apoptosis induced by high glucose will help us to understand and identify more effective strategies for the treatment of T1DM DPN.
Collapse
Affiliation(s)
- Yu-Pu Liu
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shui-Jin Shao
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hai-Dong Guo
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
47
|
Du W, Zhao S, Gao F, Wei M, An J, Jia K, Li F, Zhu L, Hao J. IFN-γ/mTORC1 decreased Rab11 in Schwann cells of diabetic peripheral neuropathy, inhibiting cell proliferation via GLUT1 downregulation. J Cell Physiol 2020; 235:5764-5776. [PMID: 31970777 DOI: 10.1002/jcp.29510] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/09/2020] [Indexed: 02/06/2023]
Abstract
Diabetic peripheral neuropathy (DPN) is the most common complication of diabetes mellitus. Rab11 is conserved gene-regulating vesicle traffic and reported to be involved in the pathogenesis of diabetes mellitus by affecting insulin sensitivity. We aimed to investigate the role of Rab11 in the pathogenesis of DPN. In this study, Rab11 expression decreased in the sciatic nerves of diabetic mice with impaired conduction function versus those of normal mice. In vitro experiment revealed interferon-γ (IFN-γ), not high glucose and interleukin 1β was the main factor to lead to Rab11 downregulation in RSC96 cells. Again, both Rab11 knockdown and IFN-γ treatment caused cell viability inhibition and the decrease in BrdU-positive cells. In contrast, overexpression of Rab11 reversed IFN-γ-reduced cell proliferation. Furthermore, mTORC1 not mTORC2 was proven to be suppressed by IFN-γ treatment in RSC96 cells, indicated in decreased phospho-p70S6K. Inhibition of the mTORC1 pathway resulted in Rab11 expression downregulation in RSC96 cells. Activation of the mTORC1 pathway effectively prevented IFN-γ-reduced Rab11 expression in RSC96 cells. Also, glucose transporter 1 (GLUT1) was found to be downregulated in RSC96 cells with Rab11 silence and overexpression of GLUT1 reversed Rab11 blocking-caused proliferation inhibition. Taken together, our findings suggest that IFN-γ decreases Rab11 expression via the inhibition of the mTORC1 signaling pathway, causing reduced cell proliferation in Schwann cells of DPN by GLUT1 downregulation.
Collapse
Affiliation(s)
- Wei Du
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Song Zhao
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Fan Gao
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Mengyu Wei
- Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiahui An
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Keqi Jia
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Fan Li
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| | - Lin Zhu
- Department of Electromyogram, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jun Hao
- Department of Pathology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
48
|
Simeoli R, Fierabracci A. Insights into the Role of MicroRNAs in the Onset and Development of Diabetic Neuropathy. Int J Mol Sci 2019; 20:ijms20184627. [PMID: 31540445 PMCID: PMC6770207 DOI: 10.3390/ijms20184627] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/30/2019] [Accepted: 09/11/2019] [Indexed: 12/18/2022] Open
Abstract
Diabetic neuropathy is a serious complication of chronic hyperglycemia in diabetes patients. This complication can involve both peripheral sensorimotor and autonomic nervous system. The precise nature of injury to the peripheral nerves mediated by chronic hyperglycemia is unknown; however, several mechanisms have been proposed including polyol pathway activation, enhanced glycation of proteins and lipids, increased oxidative stress, and cytokine release in the site of injury. MicroRNAs (miRNAs) are small non-coding RNAs that mediate RNA interference by post-transcriptionally modulating gene expression and protein synthesis. Therefore, they have been implicated in several developmental, physiological, and pathophysiological processes where they modulate the expression of different proteins. Recently, miRNAs gained an increasing attention also for their role as diagnostic test in many diseases due to their stability in serum and their easy detection. Furthermore, recent studies suggest that miRNAs may be involved in diabetic neuropathy although their role in the onset and the development of this complication is not fully understood. In this review, we discuss the most recent literature providing evidence for miRNAs role in diabetic neuropathy opening new pathways to improve both early diagnosis and treatment of this complication.
Collapse
Affiliation(s)
- Raffaele Simeoli
- Infectivology and Clinical Trials Area, Bambino Gesù Children's Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy.
| | - Alessandra Fierabracci
- Infectivology and Clinical Trials Area, Bambino Gesù Children's Hospital, IRCCS, Viale San Paolo 15, 00146 Rome, Italy.
| |
Collapse
|
49
|
Madison RD, Robinson GA. Muscle-Derived Extracellular Vesicles Influence Motor Neuron Regeneration Accuracy. Neuroscience 2019; 419:46-59. [PMID: 31454553 DOI: 10.1016/j.neuroscience.2019.08.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 01/06/2023]
Abstract
Extracellular vesicles are lipid bilayer-enclosed extracellular structures. Although the term extracellular vesicles is quite inclusive, it generally refers to exosomes (<200 nm), and microvesicles (~100-1000 nm). Such vesicles are resistant to degradation and can contain proteins, lipids, and nucleic acids. Although it was previously thought that the primary purpose of such vesicles was to rid cells of unwanted components, it is now becoming increasingly clear that they can function as intercellular messengers, sometimes operating over long distances. As such, there is now intense interest in extracellular vesicles in fields as diverse as immunology, cell biology, cancer, and more recently, neuroscience. The influence that such extracellular vesicles might exert on peripheral nerve regeneration is just beginning to be investigated. In the current studies we show that muscle-derived extracellular vesicles significantly influence the anatomical accuracy of motor neuron regeneration in the rat femoral nerve. These findings suggest a basic cellular mechanism by which target end-organs could guide their own reinnervation following nerve injury.
Collapse
Affiliation(s)
- Roger D Madison
- Research Service of the Veterans Affairs Medical Center, Durham, NC 27705, USA; Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA.
| | - Grant A Robinson
- Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
50
|
Castillo X, Castro-Obregón S, Gutiérrez-Becker B, Gutiérrez-Ospina G, Karalis N, Khalil AA, Lopez-Noguerola JS, Rodríguez LL, Martínez-Martínez E, Perez-Cruz C, Pérez-Velázquez J, Piña AL, Rubio K, García HPS, Syeda T, Vanoye-Carlo A, Villringer A, Winek K, Zille M. Re-thinking the Etiological Framework of Neurodegeneration. Front Neurosci 2019; 13:728. [PMID: 31396030 PMCID: PMC6667555 DOI: 10.3389/fnins.2019.00728] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 06/28/2019] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases are among the leading causes of disability and death worldwide. The disease-related socioeconomic burden is expected to increase with the steadily increasing life expectancy. In spite of decades of clinical and basic research, most strategies designed to manage degenerative brain diseases are palliative. This is not surprising as neurodegeneration progresses "silently" for decades before symptoms are noticed. Importantly, conceptual models with heuristic value used to study neurodegeneration have been constructed retrospectively, based on signs and symptoms already present in affected patients; a circumstance that may confound causes and consequences. Hence, innovative, paradigm-shifting views of the etiology of these diseases are necessary to enable their timely prevention and treatment. Here, we outline four alternative views, not mutually exclusive, on different etiological paths toward neurodegeneration. First, we propose neurodegeneration as being a secondary outcome of a primary cardiovascular cause with vascular pathology disrupting the vital homeostatic interactions between the vasculature and the brain, resulting in cognitive impairment, dementia, and cerebrovascular events such as stroke. Second, we suggest that the persistence of senescent cells in neuronal circuits may favor, together with systemic metabolic diseases, neurodegeneration to occur. Third, we argue that neurodegeneration may start in response to altered body and brain trophic interactions established via the hardwire that connects peripheral targets with central neuronal structures or by means of extracellular vesicle (EV)-mediated communication. Lastly, we elaborate on how lifespan body dysbiosis may be linked to the origin of neurodegeneration. We highlight the existence of bacterial products that modulate the gut-brain axis causing neuroinflammation and neuronal dysfunction. As a concluding section, we end by recommending research avenues to investigate these etiological paths in the future. We think that this requires an integrated, interdisciplinary conceptual research approach based on the investigation of the multimodal aspects of physiology and pathophysiology. It involves utilizing proper conceptual models, experimental animal units, and identifying currently unused opportunities derived from human data. Overall, the proposed etiological paths and experimental recommendations will be important guidelines for future cross-discipline research to overcome the translational roadblock and to develop causative treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Ximena Castillo
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Institute of Neurobiology, University of Puerto Rico, San Juan, PR, United States
| | - Susana Castro-Obregón
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Benjamin Gutiérrez-Becker
- Artificial Intelligence in Medical Imaging KJP, Ludwig Maximilian University of Munich, Munich, Germany
| | - Gabriel Gutiérrez-Ospina
- Laboratorio de Biología de Sistemas, Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas y Coordinación de Psicobiología y Neurociencias, Facultad de Psicología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Nikolaos Karalis
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Ahmed A. Khalil
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | | | - Liliana Lozano Rodríguez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Eduardo Martínez-Martínez
- Cell Communication & Extracellular Vesicles Laboratory, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Claudia Perez-Cruz
- National Polytechnic Institute, Center of Research in Advanced Studies, Mexico City, Mexico
| | - Judith Pérez-Velázquez
- Departamento de Matemáticas y Mecánica, Instituto de Investigaciones en Matemáticas Aplicadas y Sistemas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Mathematische Modellierung Biologischer Systeme, Fakultät für Mathematik, Technische Universität München, Munich, Germany
| | - Ana Luisa Piña
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Karla Rubio
- Lung Cancer Epigenetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | - Tauqeerunnisa Syeda
- National Polytechnic Institute, Center of Research in Advanced Studies, Mexico City, Mexico
| | - America Vanoye-Carlo
- Laboratorio de Neurociencias, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City, Mexico
| | - Arno Villringer
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin School of Mind and Brain, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Neurology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Katarzyna Winek
- The Shimon Peres Postdoctoral Fellow at the Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marietta Zille
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck, Germany
- Fraunhofer Research Institution for Marine Biotechnology and Cell Technology, Lübeck, Germany
| |
Collapse
|