1
|
Li B, Qi C, Zhang Y, Shi L, Zhang J, Qian H, Ji C. Frontier role of extracellular vesicles in kidney disease. J Nanobiotechnology 2024; 22:583. [PMID: 39304945 DOI: 10.1186/s12951-024-02852-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
Kidney diseases represent a diverse range of conditions that compromise renal function and structure which characterized by a progressive deterioration of kidney function, may ultimately necessitate dialysis or kidney transplantation as end-stage treatment options. This review explores the complex landscape of kidney diseases, highlighting the limitations of existing treatments and the pressing need for innovative strategies. The paper delves into the role of extracellular vesicles (EVs) as emerging biomarkers and therapeutic agents in the context of kidney pathophysiology. Urinary extracellular vesicles (uEVs), in particular, offer a non-invasive means of assessing renal injury and monitoring disease progression. Additionally, mesenchymal stem cell-derived EVs (MSC-EVs) are examined for their immunomodulatory and tissue repair capabilities, presenting a promising avenue for novel therapeutic interventions. And discusses the potential of engineering EVs to enhance their targeting and therapeutic efficacy. This paper systematically integrates the latest research findings and aims to provide a comprehensive overview of the role of EVs in kidney disease, providing cutting-edge insights into their potential as a diagnostic and therapeutic tool.
Collapse
Affiliation(s)
- Bei Li
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Chen Qi
- Department of Clinical Laboratory, Suzhou Municipal Hospital of Anhui Province, Anhui, 234000, China
| | - Yifan Zhang
- College of Medical Imaging, Dalian Medical University, Dalian, Liaoning, 116000, China
| | - Linru Shi
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Jiahui Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| | - Cheng Ji
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
2
|
Liu H, Kuang H, Wang Y, Bao L, Cao W, Yu L, Qi M, Wang R, Yang X, Ye Q, Ding F, Ren L, Liu S, Ma F, Liu S. MSC-derived exosomes protect auditory hair cells from neomycin-induced damage via autophagy regulation. Biol Res 2024; 57:3. [PMID: 38217055 PMCID: PMC10787390 DOI: 10.1186/s40659-023-00475-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/10/2023] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Sensorineural hearing loss (SNHL) poses a major threat to both physical and mental health; however, there is still a lack of effective drugs to treat the disease. Recently, novel biological therapies, such as mesenchymal stem cells (MSCs) and their products, namely, exosomes, are showing promising therapeutic potential due to their low immunogenicity, few ethical concerns, and easy accessibility. Nevertheless, the precise mechanisms underlying the therapeutic effects of MSC-derived exosomes remain unclear. RESULTS Exosomes derived from MSCs reduced hearing and hair cell loss caused by neomycin-induced damage in models in vivo and in vitro. In addition, MSC-derived exosomes modulated autophagy in hair cells to exert a protective effect. Mechanistically, exogenously administered exosomes were internalized by hair cells and subsequently upregulated endocytic gene expression and endosome formation, ultimately leading to autophagy activation. This increased autophagic activity promoted cell survival, decreased the mitochondrial oxidative stress level and the apoptosis rate in hair cells, and ameliorated neomycin-induced ototoxicity. CONCLUSIONS In summary, our findings reveal the otoprotective capacity of exogenous exosome-mediated autophagy activation in hair cells in an endocytosis-dependent manner, suggesting possibilities for deafness treatment.
Collapse
Affiliation(s)
- Huan Liu
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, China
| | - Huijuan Kuang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yiru Wang
- Anesthesiology Department, Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Lili Bao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wanxin Cao
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, China
| | - Lu Yu
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration and Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Meihao Qi
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Air Force Military, Xi'an, Shaanxi, China
| | - Renfeng Wang
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Air Force Military, Xi'an, Shaanxi, China
| | - Xiaoshan Yang
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Qingyuan Ye
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Digital Dentistry Center, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Feng Ding
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lili Ren
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Siying Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Furong Ma
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, China.
| | - Shiyu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
3
|
Abinti M, Favi E, Alfieri CM, Zanoni F, Armelloni S, Ferraresso M, Cantaluppi V, Castellano G. Update on current and potential application of extracellular vesicles in kidney transplantation. Am J Transplant 2023; 23:1673-1693. [PMID: 37517555 DOI: 10.1016/j.ajt.2023.07.010] [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: 05/02/2023] [Revised: 06/28/2023] [Accepted: 07/17/2023] [Indexed: 08/01/2023]
Abstract
Kidney transplantation (KT) is the best treatment for end-stage kidney disease. However, early diagnosis of graft injury remains challenging, mainly because of the lack of accurate and noninvasive diagnostic techniques. Improving graft outcomes is equally demanding, as is the development of innovative therapies. Many research efforts are focusing on extracellular vesicles, cellular particles free in each body fluid that have shown promising results as precise markers of damage and potential therapeutic targets in many diseases, including the renal field. In fact, through their receptors and cargo, they act in damage response and immune modulation. In transplantation, they may be used to determine organ quality and aging, the presence of delayed graft function, rejection, and many other transplant-related pathologies. Moreover, their low immunogenicity and safe profile make them ideal for drug delivery and the development of therapies to improve KT outcomes. In this review, we summarize current evidence about extracellular vesicles in KT, starting with their characteristics and major laboratory techniques for isolation and characterization. Then, we discuss their use as potential markers of damage and as therapeutic targets, discussing their promising use in clinical practice as a form of liquid biopsy.
Collapse
Affiliation(s)
- Matteo Abinti
- Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Evaldo Favi
- Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Carlo Maria Alfieri
- Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Francesca Zanoni
- Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, New York, USA
| | - Silvia Armelloni
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Mariano Ferraresso
- Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Vincenzo Cantaluppi
- Nephrology and Kidney Transplant Unit, Department of Translational Medicine (DIMET), University of Piemonte Orientale (UPO), "Maggiore della Carita" University Hospital, Novara, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
4
|
Dos Santos Bronel BA, Maquigussa E, Boim MA, da Silva Novaes A. Effect of extracellular vesicles derived from induced pluripotent stem cells on mesangial cells underwent a model of fibrosis in vitro. Sci Rep 2023; 13:15749. [PMID: 37735602 PMCID: PMC10514265 DOI: 10.1038/s41598-023-42912-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023] Open
Abstract
The fibrogenic process plays a significant pathophysiological role in the progression of chronic kidney disease. Inhibition of the renin-angiotensin system (RAS) is one strategy to delay disease progression but does not reverse established fibrosis. In this context, induced pluripotent stem cells (iPSCs) have been considered an alternative due to their regenerative potential. iPSCs exert their effects through paracrine signaling, which releases specific biomolecules into the extracellular environment, either directly or within extracellular vesicle (EVs), that can reach target cells. This study aims to evaluate the potential beneficial effects of iPSC-derived EVs (EV-iPSCs) in an in vitro model of fibrosis using mouse mesangial cells (MMCs) stimulated with TGF-β. EV-iPSCs were obtained by differentially ultracentrifuging iPSCs culture medium. MMCs were stimulated with 5 ng/mL of TGF-β and simultaneously treated with or without EV-iPSCs for 24 h. Markers of inflammation, fibrosis, and RAS components were assessed using RT-PCR, western blotting, and immunofluorescence. Under TGF-β stimulus, MMCs exhibited increased expression of inflammation markers, RAS components, and fibrosis. However, these changes were mitigated in the presence of EV-iPSCs. EV-iPSCs effectively reduced inflammation, RAS activation, and fibrogenesis in this fibrosis model involving mesangial cells, suggesting their potential as a strategy to reduce glomerular sclerosis.
Collapse
Affiliation(s)
- Bruno Aristides Dos Santos Bronel
- Renal Division, Department of Medicine, Universidade Federal de São Paulo, 781 Pedro de Toledo St, 13° Floor, São Paulo, SP, 04039-032, Brazil
| | - Edgar Maquigussa
- Renal Division, Department of Medicine, Universidade Federal de São Paulo, 781 Pedro de Toledo St, 13° Floor, São Paulo, SP, 04039-032, Brazil
| | - Mirian Aparecida Boim
- Renal Division, Department of Medicine, Universidade Federal de São Paulo, 781 Pedro de Toledo St, 13° Floor, São Paulo, SP, 04039-032, Brazil
| | - Antônio da Silva Novaes
- Renal Division, Department of Medicine, Universidade Federal de São Paulo, 781 Pedro de Toledo St, 13° Floor, São Paulo, SP, 04039-032, Brazil.
| |
Collapse
|
5
|
Tepus M, Tonoli E, Verderio EAM. Molecular profiling of urinary extracellular vesicles in chronic kidney disease and renal fibrosis. Front Pharmacol 2023; 13:1041327. [PMID: 36712680 PMCID: PMC9877239 DOI: 10.3389/fphar.2022.1041327] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
Chronic kidney disease (CKD) is a long-term kidney damage caused by gradual loss of essential kidney functions. A global health issue, CKD affects up to 16% of the population worldwide. Symptoms are often not apparent in the early stages, and if left untreated, CKD can progress to end-stage kidney disease (ESKD), also known as kidney failure, when the only possible treatments are dialysis and kidney transplantation. The end point of nearly all forms of CKD is kidney fibrosis, a process of unsuccessful wound-healing of kidney tissue. Detection of kidney fibrosis, therefore, often means detection of CKD. Renal biopsy remains the best test for renal scarring, despite being intrinsically limited by its invasiveness and sampling bias. Urine is a desirable source of fibrosis biomarkers as it can be easily obtained in a non-invasive way and in large volumes. Besides, urine contains biomolecules filtered through the glomeruli, mirroring the pathological state. There is, however, a problem of highly abundant urinary proteins that can mask rare disease biomarkers. Urinary extracellular vesicles (uEVs), which originate from renal cells and carry proteins, nucleic acids, and lipids, are an attractive source of potential rare CKD biomarkers. Their cargo consists of low-abundant proteins but highly concentrated in a nanosize-volume, as well as molecules too large to be filtered from plasma. Combining molecular profiling data (protein and miRNAs) of uEVs, isolated from patients affected by various forms of CKD, this review considers the possible diagnostic and prognostic value of uEVs biomarkers and their potential application in the translation of new experimental antifibrotic therapeutics.
Collapse
Affiliation(s)
- Melanie Tepus
- Centre for Health, Ageing and the Understanding of Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Elisa Tonoli
- Centre for Health, Ageing and the Understanding of Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Elisabetta A. M. Verderio
- Centre for Health, Ageing and the Understanding of Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
- Department of Biological, Geological, and Environmental Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
6
|
The Therapeutic Potential and Clinical Significance of Exosomes as Carriers of Drug Delivery System. Pharmaceutics 2022; 15:pharmaceutics15010021. [PMID: 36678650 PMCID: PMC9865231 DOI: 10.3390/pharmaceutics15010021] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Drug delivery system (DDS) realizes the drug delivery process through the drug carrier. As an important part of DDS, the selection of the drug carrier material is extremely critical, which requires the carrier material to possess excellent biocompatibility and targeting and not affect the pharmacological action of the drug. As one of the endogenous extracellular vesicles, exosomes are 30-100 nm in diameter, which are considered a new generation of a natural nanoscale delivery system. Exosomes secreted by different types of cells carry signaling molecules (such as proteins and nucleic acid) playing an important role in cell behaviors. Owing to their ability to specialize in intercellular communication, exosomes provide a distinctive method to deliver therapeutic drugs to target cells. In this concept, exosomes as the natural liposomes carry endogenous biomolecules, have excellent biocompatibility, and could be loaded with cargo both in vivo and in vitro. In addition, modifications by genetic and/or chemical engineering to part of the exosome surface or complement the desired natural effect may enhance the targeting with drug loading capability. Notably, exosomes weakly react with serum proteins prolonging cargo half-life. Overall, exosomes as natural carriers integrate the superiority of synthetic nanocarriers and cellular communication while precluding their limitations, which provides novel and reliable methods for drug delivery and treatment. Our review focuses on the therapeutic potentials and clinical values of exosomes as a carrier of drug delivery system in multiple diseases, including cancer, nervous, immune, and skeletal system diseases.
Collapse
|
7
|
Psaraki A, Ntari L, Karakostas C, Korrou-Karava D, Roubelakis MG. Extracellular vesicles derived from mesenchymal stem/stromal cells: The regenerative impact in liver diseases. Hepatology 2022; 75:1590-1603. [PMID: 34449901 DOI: 10.1002/hep.32129] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/04/2021] [Accepted: 08/24/2021] [Indexed: 02/06/2023]
Abstract
Liver dysfunctions are classified into acute and chronic diseases, which comprise a heterogeneous group of pathological features and a high mortality rate. Liver transplantation remains the gold-standard therapy for most liver diseases, with concomitant limitations related to donor organ shortage and lifelong immunosuppressive therapy. A concept in liver therapy intends to overcome these limitations based on the secreted extracellular vesicles (EVs; microvesicles and exosomes) by mesenchymal stem/stromal cells (MSCs). A significant number of studies have shown that factors released by MSCs could induce liver repair and ameliorate systemic inflammation through paracrine effects. It is well known that this paracrine action is based not only on the secretion of cytokines and growth factors but also on EVs, which regulate pathways associated with inflammation, hepatic fibrosis, integrin-linked protein kinase signaling, and apoptosis. Herein, we extensively discuss the differential effects of MSC-EVs on different liver diseases and on cellular and animal models and address the complex molecular mechanisms involved in the therapeutic potential of EVs. In addition, we cover the crucial information regarding the type of molecules contained in MSC-EVs that can be effective in the context of liver diseases. In conclusion, outcomes on MSC-EV-mediated therapy are expected to lead to an innovative, cell-free, noninvasive, less immunogenic, and nontoxic alternative strategy for liver treatment and to provide important mechanistic information on the reparative function of liver cells.
Collapse
Affiliation(s)
- Adriana Psaraki
- Laboratory of BiologyMedical SchoolNational and Kapodistrian University of AthensAthensGreece
| | - Lydia Ntari
- Laboratory of BiologyMedical SchoolNational and Kapodistrian University of AthensAthensGreece
| | - Christos Karakostas
- Laboratory of BiologyMedical SchoolNational and Kapodistrian University of AthensAthensGreece
| | - Despoina Korrou-Karava
- Laboratory of BiologyMedical SchoolNational and Kapodistrian University of AthensAthensGreece
| | - Maria G Roubelakis
- Laboratory of BiologyMedical SchoolNational and Kapodistrian University of AthensAthensGreece
- Centre of Basic ResearchBiomedical Research Foundation of the Academy of AthensAthensGreece
| |
Collapse
|
8
|
Aparicio-Trejo OE, Aranda-Rivera AK, Osorio-Alonso H, Martínez-Klimova E, Sánchez-Lozada LG, Pedraza-Chaverri J, Tapia E. Extracellular Vesicles in Redox Signaling and Metabolic Regulation in Chronic Kidney Disease. Antioxidants (Basel) 2022; 11:antiox11020356. [PMID: 35204238 PMCID: PMC8868440 DOI: 10.3390/antiox11020356] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/26/2022] [Accepted: 02/03/2022] [Indexed: 12/17/2022] Open
Abstract
Chronic kidney disease (CKD) is a world health problem increasing dramatically. The onset of CKD is driven by several mechanisms; among them, metabolic reprogramming and changes in redox signaling play critical roles in the advancement of inflammation and the subsequent fibrosis, common pathologies observed in all forms of CKD. Extracellular vesicles (EVs) are cell-derived membrane packages strongly associated with cell-cell communication since they transfer several biomolecules that serve as mediators in redox signaling and metabolic reprogramming in the recipient cells. Recent studies suggest that EVs, especially exosomes, the smallest subtype of EVs, play a fundamental role in spreading renal injury in CKD. Therefore, this review summarizes the current information about EVs and their cargos’ participation in metabolic reprogramming and mitochondrial impairment in CKD and their role in redox signaling changes. Finally, we analyze the effects of these EV-induced changes in the amplification of inflammatory and fibrotic processes in the progression of CKD. Furthermore, the data suggest that the identification of the signaling pathways involved in the release of EVs and their cargo under pathological renal conditions can allow the identification of new possible targets of injury spread, with the goal of preventing CKD progression.
Collapse
Affiliation(s)
- Omar Emiliano Aparicio-Trejo
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City 14080, Mexico
| | - Ana Karina Aranda-Rivera
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Horacio Osorio-Alonso
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City 14080, Mexico
| | - Elena Martínez-Klimova
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Laura Gabriela Sánchez-Lozada
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City 14080, Mexico
| | - José Pedraza-Chaverri
- Laboratorio F-315, Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Edilia Tapia
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología "Ignacio Chávez", Mexico City 14080, Mexico
| |
Collapse
|
9
|
Pinson MR, Chung DD, Adams AM, Scopice C, Payne EA, Sivakumar M, Miranda RC. Extracellular Vesicles in Premature Aging and Diseases in Adulthood Due to Developmental Exposures. Aging Dis 2021; 12:1516-1535. [PMID: 34527425 PMCID: PMC8407878 DOI: 10.14336/ad.2021.0322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
The developmental origins of health and disease (DOHaD) is a paradigm that links prenatal and early life exposures that occur during crucial periods of development to health outcome and risk of disease later in life. Maternal exposures to stress, some psychoactive drugs and alcohol, and environmental chemicals, among others, may result in functional changes in developing fetal tissues, creating a predisposition for disease in the individual as they age. Extracellular vesicles (EVs) may be mediators of both the immediate effects of exposure during development and early childhood as well as the long-term consequences of exposure that lead to increased risk and disease severity later in life. Given the prevalence of diseases with developmental origins, such as cardiovascular disease, neurodegenerative disorders, osteoporosis, metabolic dysfunction, and cancer, it is important to identify persistent mediators of disease risk. In this review, we take this approach, viewing diseases typically associated with aging in light of early life exposures and discuss the potential role of EVs as mediators of lasting consequences.
Collapse
Affiliation(s)
- Marisa R Pinson
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Dae D Chung
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Amy M Adams
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Chiara Scopice
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Elizabeth A Payne
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Monisha Sivakumar
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| |
Collapse
|
10
|
Li G, Kidd J, Gehr TWB, Li PL. Podocyte Sphingolipid Signaling in Nephrotic Syndrome. Cell Physiol Biochem 2021; 55:13-34. [PMID: 33861526 PMCID: PMC8193717 DOI: 10.33594/000000356] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2021] [Indexed: 11/25/2022] Open
Abstract
Podocytes play a vital role in the pathogenesis of nephrotic syndrome (NS), which is clinically characterized by heavy proteinuria, hypoalbuminemia, hyperlipidemia, and peripheral edema. The pathogenesis of NS has evolved through several hypotheses ranging from immune dysregulation theory and increased glomerular permeability theory to the current concept of podocytopathy. Podocytopathy is characterized by dysfunction or depletion of podocytes, which may be caused by unknown permeability factor, genetic disorders, drugs, infections, systemic disorders, and hyperfiltration. Over the last two decades, numerous studies have been done to explore the molecular mechanisms of podocyte injuries or NS and to develop the novel therapeutic strategies targeting podocytopathy for treatment of NS. Recent studies have shown that normal sphingolipid metabolism is essential for structural and functional integrity of podocytes. As a basic component of the plasma membrane, sphingolipids not only support the assembly of signaling molecules and interaction of receptors and effectors, but also mediate various cellular activities, such as apoptosis, proliferation, stress responses, necrosis, inflammation, autophagy, senescence, and differentiation. This review briefly summarizes current evidence demonstrating the regulation of sphingolipid metabolism in podocytes and the canonical or noncanonical roles of podocyte sphingolipid signaling in the pathogenesis of NS and associated therapeutic strategies.
Collapse
Affiliation(s)
- Guangbi Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jason Kidd
- Division of Nephrology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Todd W B Gehr
- Division of Nephrology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA,
| |
Collapse
|
11
|
Yin GN, Piao S, Liu Z, Wang L, Ock J, Kwon MH, Kim DK, Gho YS, Suh JK, Ryu JK. RNA-sequencing profiling analysis of pericyte-derived extracellular vesicle-mimetic nanovesicles-regulated genes in primary cultured fibroblasts from normal and Peyronie's disease penile tunica albuginea. BMC Urol 2021; 21:103. [PMID: 34362357 PMCID: PMC8344132 DOI: 10.1186/s12894-021-00872-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 07/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Peyronie's disease (PD) is a severe fibrotic disease of the tunica albuginea that causes penis curvature and leads to penile pain, deformity, and erectile dysfunction. The role of pericytes in the pathogenesis of fibrosis has recently been determined. Extracellular vesicle (EV)-mimetic nanovesicles (NVs) have attracted attention regarding intercellular communication between cells in the field of fibrosis. However, the global gene expression of pericyte-derived EV-mimetic NVs (PC-NVs) in regulating fibrosis remains unknown. Here, we used RNA-sequencing technology to investigate the potential target genes regulated by PC-NVs in primary fibroblasts derived from human PD plaque. METHODS Human primary fibroblasts derived from normal and PD patients was cultured and treated with cavernosum pericytes isolated extracellular vesicle (EV)-mimetic nanovesicles (NVs). A global gene expression RNA-sequencing assay was performed on normal fibroblasts, PD fibroblasts, and PD fibroblasts treated with PC-NVs. Reverse transcription polymerase chain reaction (RT-PCR) was used for sequencing data validation. RESULTS A total of 4135 genes showed significantly differential expression in the normal fibroblasts, PD fibroblasts, and PD fibroblasts treated with PC-NVs. However, only 91 contra-regulated genes were detected among the three libraries. Furthermore, 20 contra-regulated genes were selected and 11 showed consistent changes in the RNA-sequencing assay, which were validated by RT-PCR. CONCLUSION The gene expression profiling results suggested that these validated genes may be good targets for understanding potential mechanisms and conducting molecular studies into PD.
Collapse
Affiliation(s)
- Guo Nan Yin
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd St, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea
| | - Shuguang Piao
- Department of Urology at Changhai Hospital Affiliated with the Naval Medicine University, Shanghai, 200433, People's Republic of China
| | - Zhiyong Liu
- Department of Urology at Changhai Hospital Affiliated with the Naval Medicine University, Shanghai, 200433, People's Republic of China
| | - Lei Wang
- Department of Urology at Changhai Hospital Affiliated with the Naval Medicine University, Shanghai, 200433, People's Republic of China
| | - Jiyeon Ock
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd St, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea
| | - Mi-Hye Kwon
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd St, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea
| | - Do-Kyun Kim
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Jeonbuk, 54531, Korea
| | - Yong Song Gho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Kyeongsangbuk-do, 37673, Korea
| | - Jun-Kyu Suh
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd St, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea.
| | - Ji-Kan Ryu
- Department of Urology and National Research Center for Sexual Medicine, Inha University School of Medicine, 7-206, 3rd St, Shinheung-Dong, Jung-Gu, Incheon, 22332, Republic of Korea.
| |
Collapse
|
12
|
Nowak N, Yamanouchi M, Satake E. The Nephroprotective Properties of Extracellular Vesicles in Experimental Models of Chronic Kidney Disease: a Systematic Review. Stem Cell Rev Rep 2021; 18:902-932. [PMID: 34110587 PMCID: PMC8942930 DOI: 10.1007/s12015-021-10189-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2021] [Indexed: 01/14/2023]
Abstract
Extracellular vesicle (EV)-based therapy was hypothesized as a promising regenerative approach which has led to intensive research of EVs in various pathologies. In this study, we performed a comprehensive systematic review of the current experimental evidence regarding the protective properties of EVs in chronic kidney disease (CKD). We evaluated the EV-based experiments, EV characteristics, and effector molecules with their involvement in CKD pathways. Including all animal records with available creatinine or urea data, we performed a stratified univariable meta-analysis to assess the determinants of EV-based therapy effectiveness. We identified 35 interventional studies that assessed nephroprotective role of EVs and catalogued them according to their involvement in CKD mechanism. Systematic assessment of these studies suggested that EVs had consistently improved glomerulosclerosis, interstitial fibrosis, and cell damage, among different CKD models. Moreover, EV-based therapy reduced the progression of renal decline in CKD. The stratified analyses showed that the disease model, administered dose, and time of therapeutic intervention were potential predictors of therapeutic efficacy. Together, EV therapy is a promising approach for CKD progression in experimental studies. Further standardisation of EV-methods, continuous improvement of the study quality, and better understanding of the determinants of EV effectiveness will facilitate preclinical research, and may help development of clinical trials in people with CKD.
Collapse
Affiliation(s)
- Natalia Nowak
- Faculty of Medicine, Centre for Bioinformatics and Data Analysis, Medical University of Bialystok, Bialystok, Poland.
| | - Masayuki Yamanouchi
- Department of Nephrology and Laboratory Medicine Faculty of Medicine Institute of Medical, Pharmaceutical and Health Sciences Graduate School of Medical Sciences, Kanazawa University, Toranomon Hospital, Nephrology Center, Tokyo, Japan
| | - Eiichiro Satake
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, MA, Boston, USA
| |
Collapse
|
13
|
Granata S, Bruschi M, Deiana M, Petretto A, Lombardi G, Verlato A, Elia R, Candiano G, Malerba G, Gambaro G, Zaza G. Sphingomyelin and Medullary Sponge Kidney Disease: A Biological Link Identified by Omics Approach. Front Med (Lausanne) 2021; 8:671798. [PMID: 34124100 PMCID: PMC8187918 DOI: 10.3389/fmed.2021.671798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/03/2021] [Indexed: 01/07/2023] Open
Abstract
Background: Molecular biology has recently added new insights into the comprehension of the physiopathology of the medullary sponge kidney disease (MSK), a rare kidney malformation featuring nephrocalcinosis and recurrent renal stones. Pathogenesis and metabolic alterations associated to this disorder have been only partially elucidated. Methods: Plasma and urine samples were collected from 15 MSK patients and 15 controls affected by idiopathic calcium nephrolithiasis (ICN). Plasma metabolomic profile of 7 MSK and 8 ICN patients was performed by liquid chromatography combined with electrospray ionization tandem mass spectrometry (UHPLC–ESI-MS/MS). Subsequently, we reinterrogated proteomic raw data previously obtained from urinary microvesicles of MSK and ICN focusing on proteins associated with sphingomyelin metabolism. Omics results were validated by ELISA in the entire patients' cohort. Results: Thirteen metabolites were able to discriminate MSK from ICN (7 increased and 6 decreased in MSK vs. ICN). Sphingomyelin reached the top level of discrimination between the two study groups (FC: −1.8, p < 0.001). Ectonucleotide pyrophophatase phosphodiesterase 6 (ENPP6) and osteopontin (SPP1) resulted the most significant deregulated urinary proteins in MSK vs. ICN (p < 0.001). ENPP6 resulted up-regulated also in plasma of MSK by ELISA. Conclusion: Our data revealed a specific high-throughput metabolomics signature of MSK and indicated a pivotal biological role of sphingomyelin in this disease.
Collapse
Affiliation(s)
- Simona Granata
- Renal Unit, Department of Medicine, University-Hospital of Verona, Verona, Italy
| | - Maurizio Bruschi
- Laboratory of Molecular Nephrology, Istituto Pediatrico di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Michela Deiana
- Section of Biology and Genetics, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Andrea Petretto
- Core Facilities - Clinical Proteomics and Metabolomics, Istituto Pediatrico di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| | - Gianmarco Lombardi
- Renal Unit, Department of Medicine, University-Hospital of Verona, Verona, Italy
| | - Alberto Verlato
- Renal Unit, Department of Medicine, University-Hospital of Verona, Verona, Italy
| | - Rossella Elia
- Renal Unit, Department of Medicine, University-Hospital of Verona, Verona, Italy
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, Istituto Pediatrico di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Giovanni Malerba
- Section of Biology and Genetics, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Giovanni Gambaro
- Renal Unit, Department of Medicine, University-Hospital of Verona, Verona, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona, Verona, Italy
| |
Collapse
|
14
|
Harnessing the Physiological Functions of Cellular Prion Protein in the Kidneys: Applications for Treating Renal Diseases. Biomolecules 2021; 11:biom11060784. [PMID: 34067472 PMCID: PMC8224798 DOI: 10.3390/biom11060784] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022] Open
Abstract
A cellular prion protein (PrPC) is a ubiquitous cell surface glycoprotein, and its physiological functions have been receiving increased attention. Endogenous PrPC is present in various kidney tissues and undergoes glomerular filtration. In prion diseases, abnormal prion proteins are found to accumulate in renal tissues and filtered into urine. Urinary prion protein could serve as a diagnostic biomarker. PrPC plays a role in cellular signaling pathways, reno-protective effects, and kidney iron uptake. PrPC signaling affects mitochondrial function via the ERK pathway and is affected by the regulatory influence of microRNAs, small molecules, and signaling proteins. Targeting PrPC in acute and chronic kidney disease could help improve iron homeostasis, ameliorate damage from ischemia/reperfusion injury, and enhance the efficacy of mesenchymal stem/stromal cell or extracellular vesicle-based therapeutic strategies. PrPC may also be under the influence of BMP/Smad signaling and affect the progression of TGF-β-related renal fibrosis. PrPC conveys TNF-α resistance in some renal cancers, and therefore, the coadministration of anti-PrPC antibodies improves chemotherapy. PrPC can be used to design antibody-drug conjugates, aptamer-drug conjugates, and customized tissue inhibitors of metalloproteinases to suppress cancer. With preclinical studies demonstrating promising results, further research on PrPC in the kidney may lead to innovative PrPC-based therapeutic strategies for renal disease.
Collapse
|
15
|
Alteration of payload in extracellular vesicles by crosstalk with mesenchymal stem cells from different origin. J Nanobiotechnology 2021; 19:148. [PMID: 34016123 PMCID: PMC8139033 DOI: 10.1186/s12951-021-00890-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/08/2021] [Indexed: 02/07/2023] Open
Abstract
Background The application of extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) requires customized materials to target disease or cell damage. We hypothesized that EVs exert different inflammatory effects on one recipient cell, although stem cells of different origins in humans have similar payloads. Results Here, the payload of EVs released by crosstalk between MSCs and human middle ear epithelial cells (HMEECs) extracted from adipose tissue, bone marrow and tonsils significantly increased the level of anti-inflammatory factors. EVs derived from the co-culture medium decreased TNF-, COX-2, IL-1, and IL-6 levels to approximately zero within 3h in HMEECs. Expression of miR-638 and amyloid- A4 precursor protein-binding family A member 2 was analyzed using microarrays and gene ontology analysis, respectively. Conclusions In conclusion, stem cells of different origins have different payloads through crosstalk with recipient-specific cells. Inducing specific factors in EVs by co-culture with MSCs could be valuable in regenerative medicine. Graphical abstract ![]()
Collapse
|
16
|
Franzin R, Stasi A, Ranieri E, Netti GS, Cantaluppi V, Gesualdo L, Stallone G, Castellano G. Targeting Premature Renal Aging: from Molecular Mechanisms of Cellular Senescence to Senolytic Trials. Front Pharmacol 2021; 12:630419. [PMID: 33995028 PMCID: PMC8117359 DOI: 10.3389/fphar.2021.630419] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/11/2021] [Indexed: 01/10/2023] Open
Abstract
The biological process of renal aging is characterized by progressive structural and functional deterioration of the kidney leading to end-stage renal disease, requiring renal replacement therapy. Since the discovery of pivotal mechanisms of senescence such as cell cycle arrest, apoptosis inhibition, and the development of a senescence-associated secretory phenotype (SASP), efforts in the understanding of how senescent cells participate in renal physiological and pathological aging have grown exponentially. This has been encouraged by both preclinical studies in animal models with senescent cell clearance or genetic depletion as well as due to evidence coming from the clinical oncologic experience. This review considers the molecular mechanism and pathways that trigger premature renal aging from mitochondrial dysfunction, epigenetic modifications to autophagy, DNA damage repair (DDR), and the involvement of extracellular vesicles. We also discuss the different pharmaceutical approaches to selectively target senescent cells (namely, senolytics) or the development of systemic SASP (called senomorphics) in basic models of CKD and clinical trials. Finally, an overview will be provided on the potential opportunities for their use in renal transplantation during ex vivo machine perfusion to improve the quality of the graft.
Collapse
Affiliation(s)
- Rossana Franzin
- Department of Emergency and Organ Transplantation, Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Alessandra Stasi
- Department of Emergency and Organ Transplantation, Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Elena Ranieri
- Clinical Pathology, Center of Molecular Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Giuseppe Stefano Netti
- Clinical Pathology, Center of Molecular Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Vincenzo Cantaluppi
- Nephrology and Kidney Transplantation Unit, Department of Translational Medicine and Center for Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy
| | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation, Nephrology, Dialysis and Transplantation Unit, University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, Italy
| |
Collapse
|
17
|
Gang D, Yu CJ, Zhu S, Zhu P, Nasser MI. Application of mesenchymal stem cell-derived exosomes in kidney diseases. Cell Immunol 2021; 364:104358. [PMID: 33839596 DOI: 10.1016/j.cellimm.2021.104358] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 01/08/2023]
Abstract
Kidney injury (KI) has high morbidity and mortality; there has been no ideal practical treatment available in clinical practice until now. Exosomes are formed from fusing multisubunit body membranes and are secreted into the extracellular matrix, intercellular communication membracusses. As a cell-free treatment, it offers a new approach to the treatment of KI. Exosomes are spherical vesicles with or no separator cup that shapes proteins, and RNA acts on the target cells through various means to promote tissue damage and mitigate apoptosis, both inflammation and oxidative stress. Exosomes derived from mesenchymal stem cells (MSC) have a paracrine function in promoting tissue repair and immune regulation. The MSC-Exos provide specific benefits over the MSCs. The urinary exosomes closely follow the functions and diseases of the kidneys. Though much of the research in this field is only at the preliminary stages, previous research has demonstrated that MSC-Exos damaged tissues to offer proteins, mRNAs, and microRNAs as remedies for kidney injury. Although exosomes' role in tissue repair is currently is greatly debated, several key issues remain unaddressed. This is a summarization of the work done concerning MSC in the treatment of KI.
Collapse
Affiliation(s)
- Deng Gang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China; School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Chang Jiang Yu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Shuoji Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| | - M I Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China.
| |
Collapse
|
18
|
Wu Y, Zhang C, Guo R, Wu D, Shi J, Li L, Chu Y, Yuan X, Gao J. Mesenchymal Stem Cells: An Overview of Their Potential in Cell-Based Therapy for Diabetic Nephropathy. Stem Cells Int 2021; 2021:6620811. [PMID: 33815509 PMCID: PMC7990550 DOI: 10.1155/2021/6620811] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/11/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetic nephropathy (DN) is a devastating complication associated with diabetes mellitus, and it is the leading cause of end-stage renal diseases (ESRD). Over the last few decades, numerous studies have reported the beneficial effects of stem cell administration, specifically mesenchymal stem or stromal cells (MSCs), on tissue repair and regeneration. MSC therapy has been considered a promising strategy for ameliorating the progression of DN largely based on results obtained from several preclinical studies and recent Phase I/II clinical trials. This paper will review the recent literature on MSC treatment in DN. In addition, the roles and potential mechanisms involved in MSC treatment of DN will be summarized, which may present much needed new drug targets for this disease. Moreover, the potential benefits and related risks associated with the therapeutic action of MSCs are elucidated and may help in achieving a better understanding of MSCs.
Collapse
Affiliation(s)
- Yan Wu
- Heilongjiang Key Laboratory of Antifibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Chunlei Zhang
- Heilongjiang Key Laboratory of Antifibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Ran Guo
- Department of Physiology, Mudanjiang Medical University, Mudanjiang, China
| | - Dan Wu
- Heilongjiang Key Laboratory of Antifibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Jiayi Shi
- Heilongjiang Key Laboratory of Antifibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Luxin Li
- Heilongjiang Key Laboratory of Antifibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Yanhui Chu
- Heilongjiang Key Laboratory of Antifibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Xiaohuan Yuan
- Heilongjiang Key Laboratory of Antifibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, China
| | - Jie Gao
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
19
|
Sun B, Zhai S, Zhang L, Sun G. The role of extracellular vesicles in podocyte autophagy in kidney disease. J Cell Commun Signal 2021; 15:299-316. [PMID: 33619681 DOI: 10.1007/s12079-020-00594-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
Podocytes are the key cells involved in protein filtration in the glomerulus. Once proteins appear in the urine when podocytes fail, patients will end with renal failure due to the progression of glomerular damage if no proper treatment is applied. The injury and loss of podocytes can be attributed to diverse factors, such as genetic, immunologic, toxic, or metabolic disorders. Recently, autophagy has emerged as a key mechanism to eliminate the unwanted cytoplasmic materials and to prolong the lifespan of podocytes by alleviating cell damage and stress. Typically, the fundamental function of extracellular vesicles (EVs) is to mediate the intercellular communication. Recent studies have suggested that, EVs, especially exosomes, play a certain role in information transfer by communicating proteins, mRNAs, and microRNAs with recipient cells. Under physiological and pathological conditions, EVs assist in the bioinformation interchange between kidneys and other organs. It is suggested that EVs are related to the pathogenesis of acute kidney injury and chronic kidney disease, including glomerular disease, diabetic nephropathy, renal fibrosis and end-stage renal disease. However, the role of EVs in podocyte autophagy remains unclear so far. Here, this study integrated the existing information about the relevancy, diagnostic value and therapeutic potential of EVs in a variety of podocytes-related diseases. The accumulating evidence highlighted that autophagy played a critical role in the homeostasis of podocytes in glomerular disease.
Collapse
Affiliation(s)
- Baichao Sun
- Department of Nephrology, The Second Hospital of Jilin University, 218 ZiQiang Street, Changchun, 130041, Jilin, People's Republic of China.,Department of Pediatric Nephrology, The First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Shubo Zhai
- Department of Pediatric Nephrology, The First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Li Zhang
- Department of Pediatric Nephrology, The First Hospital of Jilin University, Changchun, 130021, Jilin, People's Republic of China
| | - Guangdong Sun
- Department of Nephrology, The Second Hospital of Jilin University, 218 ZiQiang Street, Changchun, 130041, Jilin, People's Republic of China.
| |
Collapse
|
20
|
Li G, Li PL. Lysosomal TRPML1 Channel: Implications in Cardiovascular and Kidney Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:275-301. [PMID: 35138619 PMCID: PMC9899368 DOI: 10.1007/978-981-16-4254-8_13] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lysosomal ion channels mediate ion flux from lysosomes and regulate membrane potential across the lysosomal membrane, which are essential for lysosome biogenesis, nutrient sensing, lysosome trafficking, lysosome enzyme activity, and cell membrane repair. As a cation channel, the transient receptor potential mucolipin 1 (TRPML1) channel is mainly expressed on lysosomes and late endosomes. Recently, the normal function of TRPML1 channels has been demonstrated to be important for the maintenance of cardiovascular and renal glomerular homeostasis and thereby involved in the pathogenesis of some cardiovascular and kidney diseases. In arterial myocytes, it has been found that Nicotinic Acid Adenine Dinucleotide Phosphate (NAADP), an intracellular second messenger, can induce Ca2+ release through the lysosomal TRPML1 channel, leading to a global Ca2+ release response from the sarcoplasmic reticulum (SR). In podocytes, it has been demonstrated that lysosomal TRPML1 channels control lysosome trafficking and exosome release, which contribute to the maintenance of podocyte functional integrity. The defect or functional deficiency of lysosomal TRPML1 channels has been shown to critically contribute to the initiation and development of some chronic degeneration or diseases in the cardiovascular system or kidneys. Here we briefly summarize the current evidence demonstrating the regulation of lysosomal TRPML1 channel activity and related signaling mechanisms. We also provide some insights into the canonical and noncanonical roles of TRPML1 channel dysfunction as a potential pathogenic mechanism for certain cardiovascular and kidney diseases and associated therapeutic strategies.
Collapse
Affiliation(s)
- Guangbi Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
21
|
Li G, Huang D, Bhat OM, Poklis JL, Zhang A, Zou Y, Kidd J, Gehr TWB, Li PL. Abnormal podocyte TRPML1 channel activity and exosome release in mice with podocyte-specific Asah1 gene deletion. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1866:158856. [PMID: 33221496 PMCID: PMC7770122 DOI: 10.1016/j.bbalip.2020.158856] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/16/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022]
Abstract
Podocytopathy and associated nephrotic syndrome (NS) have been reported in a knockout mouse strain (Asah1fl/fl/PodoCre) with a podocyte-specific deletion of α subunit (the main catalytic subunit) of acid ceramidase (Ac). However, the pathogenesis of podocytopathy of these mice remains unknown. The present study tested whether exosome release from podocytes is enhanced due to Asah1 gene knockout, which may serve as a pathogenic mechanism switching on podocytopathy and associated NS in Asah1fl/fl/PodoCre mice. We first demonstrated the remarkable elevation of urinary exosome excretion in Asah1fl/fl/PodoCre mice compared with WT/WT mice, which was accompanied by significant Annexin-II (an exosome marker) accumulation in glomeruli of Asah1fl/fl/PodoCre mice, as detected by immunohistochemistry. In cell studies, we also confirmed that Asah1 gene knockout enhanced exosome release in the primary cultures of podocyte isolated from Asah1fl/fl/PodoCre mice compared to WT/WT mice. In the podocytes from Asah1fl/fl/PodoCre mice, the interactions of lysosome and multivesicular body (MVB) were demonstrated to be decreased in comparison with those from their control littermates, suggesting reduced MVB degradation that may lead to increase in exosome release. Given the critical role of transient receptor potential mucolipin 1 (TRPML1) channel in Ca2+-dependent lysosome trafficking and consequent lysosome-MVB interaction, we tested whether lysosomal Ca2+ release through TRPML1 channels is inhibited in the podocytes of Asah1fl/fl/PodoCre mice. By GCaMP3 Ca2+ imaging, it was found that lysosomal Ca2+ release through TRPML1 channels was substantially suppressed in podocytes with Asah1 gene deletion. As an Ac product, sphingosine was found to rescue TRPML1 channel activity and thereby recover lysosome-MVB interaction and reduce exosome release of podocytes from Asah1fl/fl/PodoCre mice. Combination of N, N-dimethylsphingosine (DMS), a potent sphingosine kinase inhibitor, and sphingosine significantly inhibited urinary exosome excretion of Asah1fl/fl/PodoCre mice. Moreover, rescue of Aash1 gene expression in podocytes of Asah1fl/fl/PodoCre mice showed normal ceramide metabolism and exosome secretion. Based on these results, we conclude that the normal expression of Ac importantly contributes to the control of TRPML1 channel activity, lysosome-MVB interaction, and consequent exosome release from podocytes. Asah1 gene defect inhibits TRPML1 channel activity and thereby enhances exosome release, which may contribute to the development of podocytopathy and associated NS.
Collapse
Affiliation(s)
- Guangbi Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Dandan Huang
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Owais M Bhat
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Justin L Poklis
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Aolin Zhang
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Yao Zou
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jason Kidd
- Division of Nephrology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Todd W B Gehr
- Division of Nephrology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
22
|
Lian J, Lin J, Zakaria N, Yahaya BH. Acute Lung Injury: Disease Modelling and the Therapeutic Potential of Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1298:149-166. [PMID: 32424492 DOI: 10.1007/5584_2020_538] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute lung injury (ALI) is a severe clinical condition with high morbidity and mortality that usually results in the development of multiple organ dysfunction. The complex pathophysiology of ALI seems to provide a wide range of targets that offer numerous therapeutic options. However, despite extensive studies of ALI pathophysiology and treatment, no effective pharmacotherapy is available. Increasing evidence from both preclinical and clinical studies supports the preventive and therapeutic effects of mesenchymal stem cells (MSCs) for treating ALI. As cell-based therapy poses the risk of occlusion in microvasculature or unregulated growth, MSC-derived extracellular vesicles (MSC-EVs) have been extensively studied as a new therapeutic strategy for non-cell based therapy. It is widely accepted that the therapeutic properties of MSCs are derived from soluble factors with paracrine or endocrine effects, and EVs are among the most important paracrine or endocrine vehicles that can deliver various soluble factors with a similar phenotype as the parent cell. Therapeutic effects of MSCs have been reported for various delivery approaches, diverse doses, multiple origins, and different times of administration, and MSC-EVs treatment may include but is not limited to these choices. The mechanisms by which MSCs and MSC-EVs may contribute to ALI treatment remain elusive and need further exploration. This review provides an overview of preclinical studies that support the application of MSC-EVs for treating ALI, and it discusses emerging opportunities and their associated challenges.
Collapse
Affiliation(s)
- Jie Lian
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, SAINS@Bertam, Penang, Malaysia.,Stem Cell and Biotherapy Technology Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Norashikin Zakaria
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, SAINS@Bertam, Penang, Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, SAINS@Bertam, Penang, Malaysia.
| |
Collapse
|
23
|
de Souza W, Barrias ES. Membrane-bound extracellular vesicles secreted by parasitic protozoa: cellular structures involved in the communication between cells. Parasitol Res 2020; 119:2005-2023. [PMID: 32394001 DOI: 10.1007/s00436-020-06691-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
Abstract
The focus of this review is a group of structures/organelles collectively known as extracellular vesicles (EVs) that are secreted by most, if not all, cells, varying from mammalian cells to protozoa and even bacteria. They vary in size: some are small (100-200 nm) and others are larger (> 200 nm). In protozoa, however, most of them are small or medium in size (200-400 nm). These include vesicles from different origins. We briefly review the biogenesis of this distinct group that includes (a) exosome, which originates from the multivesicular bodies, an important component of the endocytic pathway; (b) ectosome, formed from a budding process that takes place in the plasma membrane of the cells; (c) vesicles released from the cell surface following a process of patching and capping of ligand/receptor complexes; (d) other processes where tubules secreted by the parasite subsequently originate exosome-like structures. Here, special emphasis is given to EVs secreted by parasitic protozoa such as Leishmania, Trypanosoma, Plasmodium, Toxoplasma, Cryptosporidium, Trichomonas, and Giardia. Most of them have been characterized as exosomes that were isolated using several approaches and characterized by electron microscopy, proteomic analysis, and RNA sequencing. The results obtained show clearly that they present several proteins and different types of RNAs. From the functional point of view, it is now clear that the secreted exosomes can be incorporated by the parasite itself as well as by mammalian cells with which they interact. As a consequence, there is interference both with the parasite (induction of differentiation, changes in infectivity, etc.) and with the host cell. Therefore, the EVs constitute a new system of transference of signals among cells. On the other hand, there are suggestions that exosomes may constitute potential biotechnology tools and are important players of what has been designated as nanobiotechnology. They may constitute an important delivery system for gene therapy and molecular-displaying cell regulation capabilities when incorporated into other cells and even by interfering with the exosomal membrane during its biogenesis, targeting the vesicles via specific ligands to different cell types. These vesicles may reach the bloodstream, overflow through intercellular junctions, and even pass through the central nervous system blood barrier. There is evidence that it is possible to interfere with the composition of the exosomes by interfering with multivesicular body biogenesis.
Collapse
Affiliation(s)
- Wanderley de Souza
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Centro de Ciências da Saúde, Bloco G, Ilha do Fundão, Rio de Janeiro, RJ, 21941-900, Brazil. .,Instituto Nacional de Ciência e Tecnologia and Núcleo de Biologia Estrutural e Bioimagens, CENABIO, Rio de Janeiro, Brazil.
| | - Emile S Barrias
- Instituto Nacional de Ciência e Tecnologia and Núcleo de Biologia Estrutural e Bioimagens, CENABIO, Rio de Janeiro, Brazil.,Laboratorio de Metrologia Aplicada à Ciências da Vida, Diretoria de Metrologia Aplicada à Ciências da Vida - Instituto Nacional de Metrologia, Qualidade e Tecnologia (Inmetro), Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Carracedo J, Alique M, Vida C, Bodega G, Ceprián N, Morales E, Praga M, de Sequera P, Ramírez R. Mechanisms of Cardiovascular Disorders in Patients With Chronic Kidney Disease: A Process Related to Accelerated Senescence. Front Cell Dev Biol 2020; 8:185. [PMID: 32266265 PMCID: PMC7099607 DOI: 10.3389/fcell.2020.00185] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/05/2020] [Indexed: 01/08/2023] Open
Abstract
Cardiovascular diseases (CVDs), especially those involving a systemic inflammatory process such as atherosclerosis, remain the leading cause of morbidity and mortality in patients with chronic kidney disease (CKD). CKD is a systemic condition affecting approximately 10% of the general population. The prevalence of CKD has increased over the past decades because of the aging of the population worldwide. Indeed, CVDs in patients with CKD constitute a premature form of CVD observed in the general population. Multiple studies indicate that patients with renal disease undergo accelerated aging, which precipitates the appearance of pathologies, including CVDs, usually associated with advanced age. In this review, we discuss several aspects that characterize CKD-associated CVDs, such as etiopathogenic elements that CKD patients share with the general population, changes in the cellular balance of reactive oxygen species (ROS), and the associated process of cellular senescence. Uremia-associated aging is linked with numerous changes at the cellular and molecular level. These changes are similar to those observed in the normal process of physiologic aging. We also discuss new perspectives in the study of CKD-associated CVDs and epigenetic alterations in intercellular signaling, mediated by microRNAs and/or extracellular vesicles (EVs), which promote vascular damage and subsequent development of CVD. Understanding the processes and factors involved in accelerated senescence and other abnormal intercellular signaling will identify new therapeutic targets and lead to improved methods of diagnosis and monitoring for patients with CKD-associated CVDs.
Collapse
Affiliation(s)
- Julia Carracedo
- Departamento de Genética, Fisiología y Microbiología, Universidad Complutense/Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Matilde Alique
- Departamento Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud (IRYCIS), Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - Carmen Vida
- Departamento Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud (IRYCIS), Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - Guillermo Bodega
- Departamento de Biomedicina y Biotecnología, Facultad de Biología, Química y Ciencias Ambientales, Universidad de Alcalá, Alcalá de Henares, Spain
| | - Noemí Ceprián
- Departamento de Genética, Fisiología y Microbiología, Universidad Complutense/Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Enrique Morales
- Departamento de Nefrología, Hospital Universitario 12 de Octubre/Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain.,Departamento de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Praga
- Departamento de Nefrología, Hospital Universitario 12 de Octubre/Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain.,Departamento de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Patricia de Sequera
- Departamento de Medicina, Universidad Complutense de Madrid, Madrid, Spain.,Sección de Nefrología, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Rafael Ramírez
- Departamento Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud (IRYCIS), Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| |
Collapse
|
25
|
Musante L, Bontha SV, La Salvia S, Fernandez-Piñeros A, Lannigan J, Le TH, Mas V, Erdbrügger U. Rigorous characterization of urinary extracellular vesicles (uEVs) in the low centrifugation pellet - a neglected source for uEVs. Sci Rep 2020; 10:3701. [PMID: 32111925 PMCID: PMC7048852 DOI: 10.1038/s41598-020-60619-w] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 01/30/2020] [Indexed: 02/07/2023] Open
Abstract
Urinary extracellular vesicles (uEVs) provide bio-markers for kidney and urogenital diseases. Centrifugation is the most common method used to enrich uEVs. However, a majority of studies to date have focused on the ultracentrifugation pellet, potentially losing a novel source of important biomarkers that could be obtained at lower centrifugation. Thus, the aim of this study is to rigorously characterize for the first time uEVs in the low speed pellet and determine the minimal volume of urine required for proteomic analysis (≥9.0 mL urine) and gene ontology classification identified 75% of the protein as extracellular exosomes. Cryo-Transmission Electron Microscopy (≥3.0 mL urine) provided evidence of a heterogeneous population of EVs for size and morphology independent of uromodulin filaments. Western blot detected several specific uEV kidney and EV markers (≥4.5 mL urine per lane). microRNAs quantification by qPCR was possible with urine volume as low as 0.5 mL. Particle enumeration with tunable resistive pulse sensing, nano particles tracking analysis and single EV high throughput imaging flow cytometry are possible starting from 0.5 and 3.0 mL of urine respectively. This work characterizes a neglected source of uEVs and provides guidance with regard to volume of urine necessary to carry out multi-omic studies and reveals novel aspects of uEV analysis such as autofluorescence of podocyte origin.
Collapse
Affiliation(s)
- Luca Musante
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Sai Vineela Bontha
- Transplant Research Institute, James D. Eason Transplant Institute, School of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sabrina La Salvia
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Angela Fernandez-Piñeros
- Transplant Research Institute, James D. Eason Transplant Institute, School of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Joanne Lannigan
- School of Medicine, Flow Cytometry Core, University of Virginia, Charlottesville, VA, USA
| | - Thu H Le
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY, USA
| | - Valeria Mas
- Transplant Research Institute, James D. Eason Transplant Institute, School of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Uta Erdbrügger
- Division of Nephrology and Center for Immunity, Inflammation and Regenerative Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
26
|
Podocyte Lysosome Dysfunction in Chronic Glomerular Diseases. Int J Mol Sci 2020; 21:ijms21051559. [PMID: 32106480 PMCID: PMC7084483 DOI: 10.3390/ijms21051559] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 02/06/2023] Open
Abstract
Podocytes are visceral epithelial cells covering the outer surface of glomerular capillaries in the kidney. Blood is filtered through the slit diaphragm of podocytes to form urine. The functional and structural integrity of podocytes is essential for the normal function of the kidney. As a membrane-bound organelle, lysosomes are responsible for the degradation of molecules via hydrolytic enzymes. In addition to its degradative properties, recent studies have revealed that lysosomes may serve as a platform mediating cellular signaling in different types of cells. In the last decade, increasing evidence has revealed that the normal function of the lysosome is important for the maintenance of podocyte homeostasis. Podocytes have no ability to proliferate under most pathological conditions; therefore, lysosome-dependent autophagic flux is critical for podocyte survival. In addition, new insights into the pathogenic role of lysosome and associated signaling in podocyte injury and chronic kidney disease have recently emerged. Targeting lysosomal functions or signaling pathways are considered potential therapeutic strategies for some chronic glomerular diseases. This review briefly summarizes current evidence demonstrating the regulation of lysosomal function and signaling mechanisms as well as the canonical and noncanonical roles of podocyte lysosome dysfunction in the development of chronic glomerular diseases and associated therapeutic strategies.
Collapse
|
27
|
Grange C, Papadimitriou E, Dimuccio V, Pastorino C, Molina J, O'Kelly R, Niedernhofer LJ, Robbins PD, Camussi G, Bussolati B. Urinary Extracellular Vesicles Carrying Klotho Improve the Recovery of Renal Function in an Acute Tubular Injury Model. Mol Ther 2020; 28:490-502. [PMID: 31818691 PMCID: PMC7000999 DOI: 10.1016/j.ymthe.2019.11.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 02/08/2023] Open
Abstract
Acute kidney injury, defined by a rapid deterioration of renal function, is a common complication in hospitalized patients. Among the recent therapeutic options, the use of extracellular vesicles (EVs) is considered a promising strategy. Here we propose a possible therapeutic use of renal-derived EVs isolated from normal urine (urine-derived EVs [uEVs]) in a murine model of acute injury generated by glycerol injection. uEVs accelerated renal recovery, stimulating tubular cell proliferation, reducing the expression of inflammatory and injury markers, and restoring endogenous Klotho loss. When intravenously injected, labeled uEVs localized within injured kidneys and transferred their microRNA cargo. Moreover, uEVs contained the reno-protective Klotho molecule. Murine uEVs derived from Klotho null mice lost the reno-protective effect observed using murine EVs from wild-type mice. This was regained when Klotho-negative murine uEVs were reconstituted with recombinant Klotho. Similarly, ineffective fibroblast EVs acquired reno-protection when engineered with human recombinant Klotho. Our results reveal a novel potential use of uEVs as a new therapeutic strategy for acute kidney injury, highlighting the presence and role of the reno-protective factor Klotho.
Collapse
Affiliation(s)
- Cristina Grange
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Elli Papadimitriou
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Veronica Dimuccio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Cecilia Pastorino
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Jordi Molina
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Ryan O'Kelly
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Paul D Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Giovanni Camussi
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy.
| |
Collapse
|
28
|
El Harane N, Kervadec A, Bellamy V, Pidial L, Neametalla HJ, Perier MC, Lima Correa B, Thiébault L, Cagnard N, Duché A, Brunaud C, Lemitre M, Gauthier J, Bourdillon AT, Renault MP, Hovhannisyan Y, Paiva S, Colas AR, Agbulut O, Hagège A, Silvestre JS, Menasché P, Renault NKE. Acellular therapeutic approach for heart failure: in vitro production of extracellular vesicles from human cardiovascular progenitors. Eur Heart J 2019; 39:1835-1847. [PMID: 29420830 DOI: 10.1093/eurheartj/ehy012] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 01/09/2018] [Indexed: 01/15/2023] Open
Abstract
Aims We have shown that extracellular vesicles (EVs) secreted by embryonic stem cell-derived cardiovascular progenitor cells (Pg) recapitulate the therapeutic effects of their parent cells in a mouse model of chronic heart failure (CHF). Our objectives are to investigate whether EV released by more readily available cell sources are therapeutic, whether their effectiveness is influenced by the differentiation state of the secreting cell, and through which mechanisms they act. Methods and results The total EV secreted by human induced pluripotent stem cell-derived cardiovascular progenitors (iPSC-Pg) and human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) were isolated by ultracentrifugation and characterized by Nanoparticle Tracking Analysis, western blot, and cryo-electron microscopy. In vitro bioactivity assays were used to evaluate their cellular effects. Cell and EV microRNA (miRNA) content were assessed by miRNA array. Myocardial infarction was induced in 199 nude mice. Three weeks later, mice with left ventricular ejection fraction (LVEF) ≤ 45% received transcutaneous echo-guided injections of iPSC-CM (1.4 × 106, n = 19), iPSC-Pg (1.4 × 106, n = 17), total EV secreted by 1.4 × 106 iPSC-Pg (n = 19), or phosphate-buffered saline (control, n = 17) into the peri-infarct myocardium. Seven weeks later, hearts were evaluated by echocardiography, histology, and gene expression profiling, blinded to treatment group. In vitro, EV were internalized by target cells, increased cell survival, cell proliferation, and endothelial cell migration in a dose-dependent manner and stimulated tube formation. Extracellular vesicles were rich in miRNAs and most of the 16 highly abundant, evolutionarily conserved miRNAs are associated with tissue-repair pathways. In vivo, EV outperformed cell injections, significantly improving cardiac function through decreased left ventricular volumes (left ventricular end systolic volume: -11%, P < 0.001; left ventricular end diastolic volume: -4%, P = 0.002), and increased LVEF (+14%, P < 0.0001) relative to baseline values. Gene profiling revealed that EV-treated hearts were enriched for tissue reparative pathways. Conclusion Extracellular vesicles secreted by iPSC-Pg are effective in the treatment of CHF, possibly, in part, through their specific miRNA signature and the associated stimulation of distinct cardioprotective pathways. The processing and regulatory advantages of EV could make them effective substitutes for cell transplantation.
Collapse
Affiliation(s)
- Nadia El Harane
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, 56, rue Leblanc, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anaïs Kervadec
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, 56, rue Leblanc, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Valérie Bellamy
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, 56, rue Leblanc, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Laetitia Pidial
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, 56, rue Leblanc, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Hany J Neametalla
- Department of Cardiology, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 20, rue Leblanc, 75015 Paris, France
| | - Marie-Cécile Perier
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, 56, rue Leblanc, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bruna Lima Correa
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, 56, rue Leblanc, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Léa Thiébault
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, 56, rue Leblanc, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Nicolas Cagnard
- Université Paris Descartes, US 024 SFR Necker, 24 Boulevard du Montparnasse, 75015 Paris, France
| | - Angéline Duché
- Institut National de la Santé et de la Recherche Médicale (INSERM), U 1016, Institut Cochin, Université Paris Descartes, Sorbonne Paris Cité, 22 rue Mechain, 75014 Paris, France.,CNRS, Université Paris Descartes, UMR-8104, Paris, France
| | - Camille Brunaud
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Mathilde Lemitre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, 56, rue Leblanc, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jeanne Gauthier
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, 56, rue Leblanc, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Ecole normale supérieure, PSL Research University Paris, 45 rue d'Ulm, 75005 Paris, France
| | | | - Marc P Renault
- IRIF, Université Paris Diderot-Paris 7, UMR 8243, Bâtiment Sophie Germain, 8 place Aurélie Nemours, 75013 Paris, France
| | - Yeranuhi Hovhannisyan
- Sorbonne Universités, UPMC Université de Paris-6, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, 4 Place Jussieu, 75005 Paris, France
| | - Solenne Paiva
- Sorbonne Universités, UPMC Université de Paris-6, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, 4 Place Jussieu, 75005 Paris, France
| | - Alexandre R Colas
- Sanford Burnham Prebys Medical Discovery Institute, Development, Aging and Regeneration Program, 10901 N Torrey Pines Rd, La Jolla, 92037 CA, USA
| | - Onnik Agbulut
- Sorbonne Universités, UPMC Université de Paris-6, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, 4 Place Jussieu, 75005 Paris, France
| | - Albert Hagège
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, 56, rue Leblanc, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Cardiology, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 20, rue Leblanc, 75015 Paris, France
| | - Jean-Sébastien Silvestre
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, 56, rue Leblanc, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Philippe Menasché
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, 56, rue Leblanc, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Department of Cardiovascular Surgery, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 20, rue Leblanc, 75015 Paris, France
| | - Nisa K E Renault
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-970, Paris Centre de Recherche Cardiovasculaire, 56, rue Leblanc, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
29
|
Xu X, Barreiro K, Musante L, Kretz O, Lin H, Zou H, Huber TB, Holthofer H. Management of Tamm-Horsfall Protein for Reliable Urinary Analytics. Proteomics Clin Appl 2019; 13:e1900018. [PMID: 31424164 PMCID: PMC6900072 DOI: 10.1002/prca.201900018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 08/10/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE Urinary extracellular vesicles (uEVs) are a novel source of biomarkers. However, urinary Tamm-Horsfall Protein (THP; uromodulin) interferes with all vesicle isolation attempts, precipitates with normal urinary proteins, thus, representing an unwanted "contaminant" in urinary assays. Thus, the aim is to develop a simple method to manage THP efficiently. EXPERIMENTAL DESIGN The uEVs are isolated by hydrostatic filtration dialysis (HFD) and treated with a defined solution of urea to optimize release of uEVs from sample. Presence of uEVs is confirmed by transmission electron microscopy, Western blotting, and proteomic profiling in MS. RESULTS Using HFD with urea treatment for uEV isolation reduces sample complexity to a great extent. The novel simplified uEV isolation protocol allows comprehensive vesicle proteomics analysis and should be part of any urine analytics to release all sample constituents from THP trap. CONCLUSIONS AND CLINICAL RELEVANCE The method brings a quick and easy protocol for THP management during uEV isolation, providing major benefits for comprehensive sample analytics.
Collapse
Affiliation(s)
- Xiaomeng Xu
- Institute of Nephrology and UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
- Guangdong Shunde Southern Medical University Science Park
| | - Karina Barreiro
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
| | - Luca Musante
- Division of NephrologyUniversity of VirginiaCharlottesvilleUSA
| | - Oliver Kretz
- III. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Hanfei Lin
- Institute of Nephrology and UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Hequn Zou
- Institute of Nephrology and UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Tobias B. Huber
- III. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Harry Holthofer
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
- III. Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Centre for Bioanalytical Sciences (CBAS)Dublin City UniversityDublinIreland
| |
Collapse
|
30
|
Kale VP. Application of "Primed" Mesenchymal Stromal Cells in Hematopoietic Stem Cell Transplantation: Current Status and Future Prospects. Stem Cells Dev 2019; 28:1473-1479. [PMID: 31559908 DOI: 10.1089/scd.2019.0149] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Regenerative potential of mesenchymal stem/stromal cells (MSCs) has led to their application in various cellular therapies. Since in vivo these cells are present in very low numbers, they need expansion in culture to get clinically relevant numbers; however, such long-term ex vivo manipulation leads to loss of their regenerative capacity. Although use of naïve MSCs is still the most common approach used in various therapies, several strategies, both genetic and pharmacological, are being tried out to boost the regenerative capacity of in vitro expanded MSCs. Such manipulations are very commonly reported for regeneration of various tissues like bone, cartilage, kidney, pancreas, and others. Likewise, several efforts have been made to investigate priming of MSCs to enhance their immunoregulatory activity, but such efforts have not been made to the same extent for enhancing the efficacy of hematopoietic stem cell transplantation (HSCT). Development of such approaches for HSCT would not only be useful for enhancing the transplantation efficacy of cord blood cells, which are fewer in numbers, and aged HSCs, which could be functionally compromised, but also for genetically modified HSCs, which are likely to be both, fewer in number and functionally compromised. This review specifically deals with application of "primed" MSCs in the scenario of HSCT.
Collapse
Affiliation(s)
- Vaijayanti P Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis School of Biological Sciences, Symbiosis International University, Pune, India
| |
Collapse
|
31
|
Barry F. MSC Therapy for Osteoarthritis: An Unfinished Story. J Orthop Res 2019; 37:1229-1235. [PMID: 31081558 DOI: 10.1002/jor.24343] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 04/26/2019] [Indexed: 02/04/2023]
Abstract
Mesenchymal stromal cells (MSCs) have firmly occupied the attention of orthopedic clinicians and scientists for most of the last 25 years. Hundreds of laboratories worldwide have carried out research aimed at unraveling the biological characteristics of these cells and probing the manner in which they potentially contribute to cartilage and bone repair. Clinical trials registries indicate that they are also being tested in patient studies for a wide range of conditions such as osteoarthritis, rheumatoid arthritis, fracture repair, regeneration of articular cartilage, tendon repair, and for treatment of degenerative disc disease. Despite these efforts, the effectiveness of MSCs as a treatment modality for these conditions is still uncertain and market authorizations have been limited. In addition, critical and clear phenotypic parameters for defining MSCs are uncertain and a coherent biological framework surrounding the therapeutic mechanism of action is not yet available. Added to this, cell manufacturing protocols are complex and costly and present substantial challenges in terms of regulatory oversight and standardization. Despite these obstacles, MSCs still remain at the forefront of efforts in Regenerative Medicine, based on a conviction that this technology can provide an effective treatment paradigm for major diseases where there is still an unmet need. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1229-1235, 2019.
Collapse
Affiliation(s)
- Frank Barry
- Regenerative Medicine Institute, National University of Ireland Galway, Arthritis Programme, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
32
|
Tseng AM, Chung DD, Pinson MR, Salem NA, Eaves SE, Miranda RC. Ethanol Exposure Increases miR-140 in Extracellular Vesicles: Implications for Fetal Neural Stem Cell Proliferation and Maturation. Alcohol Clin Exp Res 2019; 43:1414-1426. [PMID: 31009095 DOI: 10.1111/acer.14066] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/12/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Neural stem cells (NSCs) generate most of the neurons of the adult brain in humans, during the mid-first through second-trimester period. This critical neurogenic window is particularly vulnerable to prenatal alcohol exposure, which can result in diminished brain growth. Previous studies showed that ethanol (EtOH) exposure does not kill NSCs, but, rather, results in their depletion by influencing cell cycle kinetics and promoting aberrant maturation, in part, by altering NSC expression of key neurogenic miRNAs. NSCs reside in a complex microenvironment rich in extracellular vesicles, shown to traffic miRNA cargo between cells. METHODS We profiled the miRNA content of extracellular vesicles from control and EtOH-exposed ex vivo neurosphere cultures of fetal NSCs. We subsequently examined the effects of one EtOH-sensitive miRNA, miR-140-3p, on NSC growth, survival, and maturation. RESULTS EtOH exposure significantly elevates levels of a subset of miRNAs in secreted extracellular vesicles. Overexpression of one of these elevated miRNAs, miR-140-3p, and its passenger strand relative, miR-140-5p, significantly increased the proportion of S-phase cells while decreasing the proportion of G0 /G1 cells compared to controls. In contrast, while miR-140-3p knockdown had minimal effects on the proportion of cells in each phase of the cell cycle, knockdown of miR-140-5p significantly decreased the proportion of cells in G2 /M phase. Furthermore, miR-140-3p overexpression, during mitogen-withdrawal-induced NSC differentiation, favors astroglial maturation at the expense of neural and oligodendrocyte differentiation. CONCLUSIONS Collectively, the dysregulated miRNA content of extracellular vesicles following EtOH exposure may result in aberrant neural progenitor cell growth and maturation, explaining brain growth deficits associated with prenatal alcohol exposure.
Collapse
Affiliation(s)
- Alexander M Tseng
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas
| | - Dae D Chung
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas
| | - Marisa R Pinson
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas
| | - Nihal A Salem
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas
| | - Sarah E Eaves
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University Health Science Center, Bryan, Texas
| |
Collapse
|
33
|
The role of extracellular vesicles in renal fibrosis. Cell Death Dis 2019; 10:367. [PMID: 31068572 PMCID: PMC6506498 DOI: 10.1038/s41419-019-1605-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/11/2019] [Accepted: 04/16/2019] [Indexed: 12/15/2022]
Abstract
As a particularly important mediator of intercellular communication, extracellular vesicles (EVs) have been proved to be extensively involved in various system diseases over the past two decades, including in renal diseases. As is well-known, renal fibrosis is the common pathological process of any ongoing renal disease or adaptive repair of kidney injury based on current knowledge. Although much work has been performed focusing on EVs in various renal diseases, the role of EVs in renal fibrosis has not been described in detail and summarized. In this review, we provide a brief overview of the definition, classification and biological process of EVs. Then, the potential mechanisms of EVs in renal fibrosis are illustrated. Lastly, recent advances in EVs and the implications of EVs for diagnosis and therapy in renal fibrosis disease are introduced. We look forward to a more comprehensive understanding of EVs in renal fibrosis, which could be a boon to patients with renal fibrosis disease.
Collapse
|
34
|
Influence of high glucose on mesangial cell-derived exosome composition, secretion and cell communication. Sci Rep 2019; 9:6270. [PMID: 31000742 PMCID: PMC6472340 DOI: 10.1038/s41598-019-42746-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 11/12/2018] [Indexed: 01/10/2023] Open
Abstract
Mesangial cells stimulated with high glucose (HG) exhibit increased intracellular angiotensin II (AngII) synthesis that is correlated with the upregulation of AngII target genes, such as profibrotic cytokines. The intracrine effects of AngII can be mediated by several molecules transferred to other cells via exosomes (Exos), which play a key role in cellular communication under many physiological and pathological conditions. The aim of this study was to investigate the effects of exosomes derived from HG-stimulated human mesangial cells (HG-HMCs) on normal unstimulated HMCs. Exosomes from HMCs (C-Exos) and HG-HMCs (HG-Exos) were obtained from cell culture supernatants. HMCs were incubated with C-Exos or HG-Exos. HG stimulus induced a change in the amount but not the size of Exos. Both C-Exos and HG-Exos contained angiotensinogen and renin, but no angiotensin converting enzyme was detected. Compared with HMCs treated with C-Exos, HMCs treated with HG-Exos presented higher levels of fibronectin, angiotensinogen, renin, AT1 and AT2 receptors, indicating that HG-Exos modified the function of normal HMCs. These results suggest that the intercellular communication through Exos may have pathophysiological implications in the diabetic kidney.
Collapse
|
35
|
Hong J, Bhat OM, Li G, Dempsey SK, Zhang Q, Ritter JK, Li W, Li PL. Lysosomal regulation of extracellular vesicle excretion during d-ribose-induced NLRP3 inflammasome activation in podocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:849-860. [PMID: 30771382 PMCID: PMC6800119 DOI: 10.1016/j.bbamcr.2019.02.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/09/2019] [Accepted: 02/12/2019] [Indexed: 12/11/2022]
Abstract
The NLRP3 inflammasome is activated in the cytoplasm of cells and its products such as IL-1β are exported through a non-classical ER-Golgi pathway. Several mechanistically distinct models including exocytosis of secretory lysosomes, microvesicles (MVs) and extracellular vehicles (EVs) have been proposed for their release. In this study, we hypothesized that the NLRP3 inflammasome product, IL-1β in response to exogenously administrated and endogenously produced D-ribose stimulation is released via extracellular vesicles including EVs via a sphingolipid-mediated molecular mechanisms controlling lysosome and multivesicular body (MVB) interaction. First, we demonstrated that both endogenous and exogenous D-ribose induced NLRP3 inflammasome activation to produce IL-1β, which was released via EVs in podocytes. Then, we found that colocalization of marker MVB marker VPS16 with IL-1β within podocytes increased upon D-ribose stimulation, which was accompanied by decreased colocalization of lysosome marker Lamp-1 and VPS16, suggesting decrease in MVB inclusion of IL-1β due to reduced lysosome and MVB interaction. All these changes were mimicked and accelerated by lysosome v-ATPase inhibitor, bafilomycin. Moreover, ceramide in podocytes was found elevated upon D-ribose stimulation, and prior treatments of podocyte with acid sphingomyelinase (Asm) inhibitor, amitriptyline, acid ceramidase (AC) inducer, genistein, or AC CRISPR/cas9 activation plasmids were found to decrease D-ribose-induced ceramide accumulation, EVs release and IL-1β secretion due to reduced interactions of lysosome with MVBs. These results suggest that inflammasome-derived products such as IL-1β during D-ribose stimulation are released via EVs, in which lysosomal sphingolipid-mediated regulation of lysosome function plays an important role.
Collapse
Affiliation(s)
- Jinni Hong
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States of America; Integrated Laboratory of Traditional Chinese Medicine and Western Medicine, Peking University First Hospital, Beijing, People's Republic of China
| | - Owais M Bhat
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Guangbi Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Sara K Dempsey
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Qinghua Zhang
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Joseph K Ritter
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Weiwei Li
- Integrated Laboratory of Traditional Chinese Medicine and Western Medicine, Peking University First Hospital, Beijing, People's Republic of China
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States of America.
| |
Collapse
|
36
|
Farzamfar S, Hasanpour A, Nazeri N, Razavi H, Salehi M, Shafei S, Nooshabadi VT, Vaez A, Ehterami A, Sahrapeyma H, Ai J. Extracellular micro/nanovesicles rescue kidney from ischemia-reperfusion injury. J Cell Physiol 2019; 234:12290-12300. [PMID: 30609022 DOI: 10.1002/jcp.27998] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/03/2018] [Indexed: 12/16/2022]
Abstract
Acute renal failure (ARF) is a clinical challenge that is highly resistant to treatment, and its high rate of mortality is alarming. Ischemia-reperfusion injury (IRI) is the most common cause of ARF. Especially IRI is implicated in kidney transplantation and can determine graft survival. Although the exact pathophysiology of renal IRI is unknown, the role of inflammatory responses has been elucidated. Because mesenchymal stromal cells (MSCs) have strong immunomodulatory properties, they are under extensive investigation as a therapeutic modality for renal IRI. Extracellular vesicles (EVs) play an integral role in cell-to-cell communication. Because the regenerative potential of the MSCs can be recapitulated by their EVs, the therapeutic appeal of MSC-derived EVs has dramatically increased in the past decade. Higher safety profile and ease of preservation without losing function are other advantages of EVs compared with their producing cells. In the current review, the preliminary results and potential of MSC-derived EVs to alleviate kidney IRI are summarized. We might be heading toward a cell-free approach to treat renal IRI.
Collapse
Affiliation(s)
- Saeed Farzamfar
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Akram Hasanpour
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloufar Nazeri
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hengameh Razavi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Salehi
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.,Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Shilan Shafei
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, International Campus of Tehran University of Medical Sciences, Tehran, Iran
| | - Vajiheh T Nooshabadi
- Department of Applied Cell Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arian Ehterami
- Department of Mechanical and Aerospace Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hamed Sahrapeyma
- Department of Biomaterial Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Li H, Rong P, Ma X, Nie W, Chen C, Yang C, Zhang J, Dong Q, Wang W. Paracrine effect of mesenchymal stem cell as a novel therapeutic strategy for diabetic nephropathy. Life Sci 2018; 215:113-118. [PMID: 30399376 DOI: 10.1016/j.lfs.2018.11.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/25/2018] [Accepted: 11/01/2018] [Indexed: 12/14/2022]
Abstract
Diabetic nephropathy (DN) is a microvascular complication of diabetes mellitus (DM) and the main reason for end-stage renal diseases (ESRD). Based on the role of mesenchymal stem cells (MSCs) in regenerative medicine, the MSC therapy has been considered a promising strategy to ameliorate the progression of DN. In this article, we review the therapeutic potential of MSCs in DN, mainly involving MSC paracrine mechanism based on trophic factors and extracellular vesicles. Knowledge of mechanism underlying the therapeutic action of MSCs on DN can provide much needed new drug targets for this disease.
Collapse
Affiliation(s)
- Hongde Li
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Pengfei Rong
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaoqian Ma
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wei Nie
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Cheng Chen
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Cejun Yang
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Juan Zhang
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qiong Dong
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wei Wang
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China; Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
38
|
Kubota K, Nakano M, Kobayashi E, Mizue Y, Chikenji T, Otani M, Nagaishi K, Fujimiya M. An enriched environment prevents diabetes-induced cognitive impairment in rats by enhancing exosomal miR-146a secretion from endogenous bone marrow-derived mesenchymal stem cells. PLoS One 2018; 13:e0204252. [PMID: 30240403 PMCID: PMC6150479 DOI: 10.1371/journal.pone.0204252] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/04/2018] [Indexed: 01/18/2023] Open
Abstract
Increasing evidence suggests that an enriched environment (EE) ameliorates cognitive impairment by promoting repair of brain damage. However, the mechanisms by which this occurs have not been determined. To address this issue, we investigated whether an EE enhanced the capability of endogenous bone marrow-derived mesenchymal stem/stromal cells (BM-MSCs) to prevent hippocampal damage due to diabetes by focusing on miRNA carried in BM-MSC-derived exosomes. In diabetic streptozotocin (STZ) rats housed in an EE (STZ/EE), cognitive impairment was significantly reduced, and both neuronal and astroglial damage in the hippocampus was alleviated compared with STZ rats housed in conventional cages (STZ/CC). BM-MSCs isolated from STZ/CC rats had functional and morphological abnormalities that were not detected in STZ/EE BM-MSCs. The miR-146a levels in exosomes in conditioned medium of cultured BM-MSCs and serum from STZ/CC rats were decreased compared with non-diabetic rats, and the level was restored in STZ/EE rats. Thus, the data suggest that increased levels of miR-146a in sera were derived from endogenous BM-MSCs in STZ/EE rats. To examine the possibility that increased miR-146a in serum may exert anti-inflammatory effects on astrocytes in diabetic rats, astrocytes transfected with miR-146a were stimulated with advanced glycation end products (AGEs) to mimic diabetic conditions. The expression of IRAK1, NF-κB, and tumor necrosis factor-α was significantly higher in AGE-stimulated astrocytes, and these factors were decreased in miR-146a-transfected astrocytes. These results suggested that EEs stimulate up-regulation of exosomal miR-146a secretion by endogenous BM-MSCs, which exerts anti-inflammatory effects on damaged astrocytes and prevents diabetes-induced cognitive impairment.
Collapse
Affiliation(s)
- Kenta Kubota
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
- Department of Physical Therapy, Hokkaido Chitose Rehabilitation University, Chitose, Hokkaido, Japan
| | - Masako Nakano
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
| | - Eiji Kobayashi
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
| | - Yuka Mizue
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
| | - Takako Chikenji
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
| | - Miho Otani
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
| | - Kanna Nagaishi
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
| | - Mineko Fujimiya
- Department of Anatomy, Sapporo Medical University, School of Medicine, Sapporo, Hokkaido, Japan
- * E-mail:
| |
Collapse
|
39
|
Chiriacò MS, Bianco M, Nigro A, Primiceri E, Ferrara F, Romano A, Quattrini A, Furlan R, Arima V, Maruccio G. Lab-on-Chip for Exosomes and Microvesicles Detection and Characterization. SENSORS (BASEL, SWITZERLAND) 2018; 18:E3175. [PMID: 30241303 PMCID: PMC6210978 DOI: 10.3390/s18103175] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/05/2018] [Accepted: 09/16/2018] [Indexed: 12/11/2022]
Abstract
Interest in extracellular vesicles and in particular microvesicles and exosomes, which are constitutively produced by cells, is on the rise for their huge potential as biomarkers in a high number of disorders and pathologies as they are considered as carriers of information among cells, as well as being responsible for the spreading of diseases. Current methods of analysis of microvesicles and exosomes do not fulfill the requirements for their in-depth investigation and the complete exploitation of their diagnostic and prognostic value. Lab-on-chip methods have the potential and capabilities to bridge this gap and the technology is mature enough to provide all the necessary steps for a completely automated analysis of extracellular vesicles in body fluids. In this paper we provide an overview of the biological role of extracellular vesicles, standard biochemical methods of analysis and their limits, and a survey of lab-on-chip methods that are able to meet the needs of a deeper exploitation of these biological entities to drive their use in common clinical practice.
Collapse
Affiliation(s)
| | - Monica Bianco
- CNR NANOTEC Institute of Nanotechnology, via Monteroni, 73100 Lecce, Italy.
| | - Annamaria Nigro
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.
| | | | - Francesco Ferrara
- CNR NANOTEC Institute of Nanotechnology, via Monteroni, 73100 Lecce, Italy.
- STMicroelectronics, Via Monteroni, I-73100 Lecce, Italy.
| | - Alessandro Romano
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.
| | - Angelo Quattrini
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.
| | - Roberto Furlan
- Institute of Experimental Neurology, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.
| | - Valentina Arima
- CNR NANOTEC Institute of Nanotechnology, via Monteroni, 73100 Lecce, Italy.
| | - Giuseppe Maruccio
- CNR NANOTEC Institute of Nanotechnology, via Monteroni, 73100 Lecce, Italy.
- Department of Mathematics and Physics, University of Salento, via Monteroni, 73100 Lecce, Italy.
| |
Collapse
|
40
|
Alberti D, Grange C, Porta S, Aime S, Tei L, Geninatti Crich S. Efficient Route to Label Mesenchymal Stromal Cell-Derived Extracellular Vesicles. ACS OMEGA 2018; 3:8097-8103. [PMID: 30087935 PMCID: PMC6072237 DOI: 10.1021/acsomega.8b00908] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/06/2018] [Indexed: 05/23/2023]
Abstract
Recent research results report that extracellular vesicles (EVs) have a central role in both physiological and pathological processes involving intercellular communication. Herein, a simple EVs labeling procedure based on the metabolic labeling of secreting cells (mesenchymal stroma cells, MSCs) with a fluorescein-containing bio-orthogonal dye is described. This procedure was carried out by incubating cells for 72 h with tetraacetylated N-azidoacetyl-d-mannosamine (Ac4ManNAz), a modified sugar containing an azido group that, upon incorporation on the external surface of the cytoplasmatic cell membrane, is specifically conjugated with cyclooctyne-modified fluorescein isothiocyanate (ADIBO-FITC). MSCs released fluorescent EVs did not need any further purification. Finally, cellular uptake and tracking of the fluorescein-labeled EVs were successfully assessed by targeting experiments with MSCs. The method appears of general applicability and it may be very useful opening new horizon on diagnostic and therapeutic protocols exploiting EVs.
Collapse
Affiliation(s)
- Diego Alberti
- Department
of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, 10126 Torino, Italy
| | - Cristina Grange
- Department
of Medical Sciences, University of Turin, Corso Dogliotti 14, 10126 Torino, Italy
| | - Stefano Porta
- Department
of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, 10126 Torino, Italy
| | - Silvio Aime
- Department
of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, 10126 Torino, Italy
| | - Lorenzo Tei
- Department
of Science and Technological Innovation, University of Piemonte Orientale, Viale T. Michel 11, 15121 Alessandria, Italy
| | - Simonetta Geninatti Crich
- Department
of Molecular Biotechnology and Health Science, University of Turin, Via Nizza 52, 10126 Torino, Italy
| |
Collapse
|
41
|
Fu Y, Karbaat L, Wu L, Leijten J, Both SK, Karperien M. Trophic Effects of Mesenchymal Stem Cells in Tissue Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2018; 23:515-528. [PMID: 28490258 DOI: 10.1089/ten.teb.2016.0365] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) are considered to hold great therapeutic value for cell-based therapy and for tissue regeneration in particular. Recent evidence indicates that the main underlying mechanism for MSCs' beneficial effects in tissue regeneration is based on their capability to produce a large variety of bioactive trophic factors that stimulate neighboring parenchymal cells to start repairing damaged tissues. These new findings could potentially replace the classical paradigm of MSC differentiation and cell replacement. These bioactive factors have diverse actions like modulating the local immune system, enhancing angiogenesis, preventing cell apoptosis, and stimulating survival, proliferation, and differentiation of resident tissue specific cells. Therefore, MSCs are referred to as conductors of tissue repair and regeneration by secreting trophic mediators. In this review article, we have summarized the studies that focused on the trophic effects of MSC within the context of tissue regeneration. We will also highlight the various underlying mechanisms used by MSCs to act as trophic mediators. Besides the secretion of growth factors, we discuss two additional mechanisms that are likely to mediate MSC's beneficial effects in tissue regeneration, namely the production of extracellular vesicles and the formation of membrane nanotubes, which can both connect different cells and transfer a variety of trophic factors varying from proteins to mRNAs and miRNAs. Furthermore, we postulate that apoptosis of the MSCs is an integral part of the trophic effect during tissue repair.
Collapse
Affiliation(s)
- Yao Fu
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| | - Lisanne Karbaat
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| | - Ling Wu
- 2 Center for Craniofacial Molecular Biology, University of Southern California , Los Angeles, Los Angeles, California
| | - Jeroen Leijten
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| | - Sanne K Both
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| | - Marcel Karperien
- 1 Developmental BioEngineering, MIRA Institute for Biomedical Technology & Technical Medicine, University of Twente , Enschede, Netherlands
| |
Collapse
|
42
|
Cunnane EM, Weinbaum JS, O'Brien FJ, Vorp DA. Future Perspectives on the Role of Stem Cells and Extracellular Vesicles in Vascular Tissue Regeneration. Front Cardiovasc Med 2018; 5:86. [PMID: 30018970 PMCID: PMC6037696 DOI: 10.3389/fcvm.2018.00086] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/13/2018] [Indexed: 02/06/2023] Open
Abstract
Vascular tissue engineering is an area of regenerative medicine that attempts to create functional replacement tissue for defective segments of the vascular network. One approach to vascular tissue engineering utilizes seeding of biodegradable tubular scaffolds with stem (and/or progenitor) cells wherein the seeded cells initiate scaffold remodeling and prevent thrombosis through paracrine signaling to endogenous cells. Stem cells have received an abundance of attention in recent literature regarding the mechanism of their paracrine therapeutic effect. However, very little of this mechanistic research has been performed under the aegis of vascular tissue engineering. Therefore, the scope of this review includes the current state of TEVGs generated using the incorporation of stem cells in biodegradable scaffolds and potential cell-free directions for TEVGs based on stem cell secreted products. The current generation of stem cell-seeded vascular scaffolds are based on the premise that cells should be obtained from an autologous source. However, the reduced regenerative capacity of stem cells from certain patient groups limits the therapeutic potential of an autologous approach. This limitation prompts the need to investigate allogeneic stem cells or stem cell secreted products as therapeutic bases for TEVGs. The role of stem cell derived products, particularly extracellular vesicles (EVs), in vascular tissue engineering is exciting due to their potential use as a cell-free therapeutic base. EVs offer many benefits as a therapeutic base for functionalizing vascular scaffolds such as cell specific targeting, physiological delivery of cargo to target cells, reduced immunogenicity, and stability under physiological conditions. However, a number of points must be addressed prior to the effective translation of TEVG technologies that incorporate stem cell derived EVs such as standardizing stem cell culture conditions, EV isolation, scaffold functionalization with EVs, and establishing the therapeutic benefit of this combination treatment.
Collapse
Affiliation(s)
- Eoghan M Cunnane
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Justin S Weinbaum
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - David A Vorp
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
43
|
Torres Crigna A, Daniele C, Gamez C, Medina Balbuena S, Pastene DO, Nardozi D, Brenna C, Yard B, Gretz N, Bieback K. Stem/Stromal Cells for Treatment of Kidney Injuries With Focus on Preclinical Models. Front Med (Lausanne) 2018; 5:179. [PMID: 29963554 PMCID: PMC6013716 DOI: 10.3389/fmed.2018.00179] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/24/2018] [Indexed: 12/18/2022] Open
Abstract
Within the last years, the use of stem cells (embryonic, induced pluripotent stem cells, or hematopoietic stem cells), Progenitor cells (e.g., endothelial progenitor cells), and most intensely mesenchymal stromal cells (MSC) has emerged as a promising cell-based therapy for several diseases including nephropathy. For patients with end-stage renal disease (ESRD), dialysis or finally organ transplantation are the only therapeutic modalities available. Since ESRD is associated with a high healthcare expenditure, MSC therapy represents an innovative approach. In a variety of preclinical and clinical studies, MSC have shown to exert renoprotective properties, mediated mainly by paracrine effects, immunomodulation, regulation of inflammation, secretion of several trophic factors, and possibly differentiation to renal precursors. However, studies are highly diverse; thus, knowledge is still limited regarding the exact mode of action, source of MSC in comparison to other stem cell types, administration route and dose, tracking of cells and documentation of therapeutic efficacy by new imaging techniques and tissue visualization. The aim of this review is to provide a summary of published studies of stem cell therapy in acute and chronic kidney injury, diabetic nephropathy, polycystic kidney disease, and kidney transplantation. Preclinical studies with allogeneic or xenogeneic cell therapy were first addressed, followed by a summary of clinical trials carried out with autologous or allogeneic hMSC. Studies were analyzed with respect to source of cell type, mechanism of action etc.
Collapse
Affiliation(s)
- Adriana Torres Crigna
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Cristina Daniele
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Carolina Gamez
- Department for Experimental Orthopaedics and Trauma Surgery, Medical Faculty Mannheim, Orthopaedic and Trauma Surgery Centre (OUZ), Heidelberg University, Mannheim, Germany
| | - Sara Medina Balbuena
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Diego O. Pastene
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Daniela Nardozi
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Cinzia Brenna
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Benito Yard
- Department of Medicine (Nephrology/Endrocrinology/Rheumathology), University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Norbert Gretz
- Medical Faculty Mannheim, Medical Research Centre, University of Heidelberg, Mannheim, Germany
| | - Karen Bieback
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, University of Heidelberg, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| |
Collapse
|
44
|
Perco P, Mayer G. Endogenous factors and mechanisms of renoprotection and renal repair. Eur J Clin Invest 2018; 48:e12914. [PMID: 29460289 DOI: 10.1111/eci.12914] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/14/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND An imbalance between renal damaging molecules and nephroprotective factors contributes to the development and progression of kidney diseases. Molecules with renoprotective properties might serve as biomarkers, drug targets as well as therapeutic options themselves. MATERIALS AND METHODS For this review, we generated a set of renoprotective factors based on GeneRIF (Gene Reference Into Function) information available at NCBI's PubMed. The final set of manually curated renoprotective factors was investigated with respect to tissue-specific expression, subcellular location distribution and involvement in biological processes using information from gene ontology as well as information from protein-protein interaction databases. We furthermore investigated the factors in the context of clinical trials of renal disease and diabetes. RESULTS One hundred and ninety-three factors could be retrieved from the set of GeneRIFs on nephroprotection and renal repair. A large number of factors were either secretory molecules or plasma membrane receptors. Next to the elevated expression in renal tissue, also higher expression in connective tissue and pancreas was observed. The proteins could be assigned to the broad functional categories of cell proliferation and signalling, inflammatory response, apoptosis, blood pressure regulation as well as cellular response to different kinds of insults such as hypoxia, heat or mechanical stimulus. Eight factors are studied in clinical trials with additional ones being targeted by compounds. CONCLUSIONS We have generated a set of renoprotective factors based on the literature information, which was functionally annotated and evaluated with respect to tested compounds in kidney disease and diabetes clinical trials.
Collapse
Affiliation(s)
- Paul Perco
- Department of Internal Medicine IV, Medical University of Innsbruck, Innsbruck, Austria
| | - Gert Mayer
- Department of Internal Medicine IV, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
45
|
Lv LL, Feng Y, Wen Y, Wu WJ, Ni HF, Li ZL, Zhou LT, Wang B, Zhang JD, Crowley SD, Liu BC. Exosomal CCL2 from Tubular Epithelial Cells Is Critical for Albumin-Induced Tubulointerstitial Inflammation. J Am Soc Nephrol 2018; 29:919-935. [PMID: 29295871 DOI: 10.1681/asn.2017050523] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 11/14/2017] [Indexed: 11/03/2022] Open
Abstract
Albuminuria is a key instigator of tubulointerstitial inflammation associated with CKD, but the mechanism through which filtered albumin propagates renal injury remains unclear. In this study, we explored the role in this process of exosome mRNA released from tubular epithelial cells (TECs). Compared with control mice, acute and chronic kidney injury models had more exosomes containing inflammatory cytokine mRNA, particularly the chemokine CCL2, in kidneys and urine. In vitro stimulation of TECs with BSA recapitulated this finding. Notably, the internalization of purified TEC exosomes by cultured macrophages increased if TECs were exposed to BSA. Macrophage internalization of exosomes from BSA-treated TECs led to an enhanced inflammatory response and macrophage migration, but CCL2 silencing in TECs prevented these effects. Using a GFP-CCL2 fusion mRNA construct, we observed direct transfer of CCL2 mRNA from TEC exosomes to macrophages. Mice subjected to tail vein injection of purified BSA-treated TEC exosomes developed tubular injury with renal inflammatory cell infiltration. However, injection of exosomes from BSA-treated CCL2-deficient TECs induced less severe kidney inflammation. Finally, in patients with IgA nephropathy, the increase of proteinuria correlated with augmented urinary excretion of exosomes with exaggerated expression of CCL2 mRNA. Moreover, the level of CCL2 mRNA in urinary exosomes correlated closely with levels of renal interstitial macrophage infiltration in these patients. Our studies demonstrate that the increasing release of exosomes that transfer CCL2 mRNA from TECs to macrophages constitutes a critical mechanism of albumin-induced tubulointerstitial inflammation.
Collapse
Affiliation(s)
- Lin-Li Lv
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Ye Feng
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Yi Wen
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Wei-Jun Wu
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Hai-Feng Ni
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Zuo-Lin Li
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Le-Ting Zhou
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Bin Wang
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Jian-Dong Zhang
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University and Durham Veterans Affairs Medical Centers, Durham, North Carolina
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhongda Hospitial, Southeast University School of Medicine, Nanjing, China; and
| |
Collapse
|
46
|
Extracellular vesicles from human-induced pluripotent stem cell-derived mesenchymal stromal cells (hiPSC-MSCs) protect against renal ischemia/reperfusion injury via delivering specificity protein (SP1) and transcriptional activating of sphingosine kinase 1 and inhibiting necroptosis. Cell Death Dis 2017; 8:3200. [PMID: 29233979 PMCID: PMC5870585 DOI: 10.1038/s41419-017-0041-4] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 02/07/2023]
Abstract
Renal ischemia-reperfusion is a main cause of acute kidney injury (AKI), which is associated with high mortality. Here we show that extracellular vesicles (EVs) secreted from hiPSC-MSCs play a critical role in protection against renal I/R injury. hiPSC-MSCs-EVs can fuse with renal cells and deliver SP1 into target cells, subsequently active SK1 expression and increase S1P formation. Chromatin immunoprecipitation (ChIP) analyses and luciferase assay were used to confirm SP1 binds directly to the SK1 promoter region and promote promoter activity. Moreover, SP1 inhibition (MIT) or SK1 inhibition (SKI-II) completely abolished the renal protective effect of hiPSC-MSCs-EVs in rat I/R injury mode. However, pre-treatment of necroptosis inhibitor Nec-1 showed no difference with the administration of hiPSC-MSCs-EVs only. We then generated an SP1 knockout hiPSC-MSC cell line by CRISPR/Cas9 system and found that SP1 knockout failed to show the protective effect of hiPSC-MSCs-EVs unless restoring the level of SP1 by Ad-SP1 in vitro and in vivo. In conclusion, this study describes an anti-necroptosis effect of hiPSC-MSCs-EVs against renal I/R injury via delivering SP1 into target renal cells and intracellular activating the expression of SK1 and the generation of S1P. These findings suggest a novel mechanism for renal protection against I/R injury, and indicate a potential therapeutic approach for a variety of renal diseases and renal transplantation.
Collapse
|
47
|
Minuth WW. Concepts for a therapeutic prolongation of nephrogenesis in preterm and low-birth-weight babies must correspond to structural-functional properties in the nephrogenic zone. Mol Cell Pediatr 2017; 4:12. [PMID: 29218481 PMCID: PMC5721096 DOI: 10.1186/s40348-017-0078-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/20/2017] [Indexed: 12/30/2022] Open
Abstract
Numerous investigations are dealing with anlage of the mammalian kidney and primary development of nephrons. However, only few information is available about the last steps in kidney development leading at birth to a downregulation of morphogen activity in the nephrogenic zone and to a loss of stem cell niches aligned beyond the organ capsule. Surprisingly, these natural changes in the developmental program display similarities to processes occurring in the kidneys of preterm and low-birth-weight babies. Although those babies are born at a time with a principally intact nephrogenic zone and active niches, a high proportion of them suffers on impairment of nephrogenesis resulting in oligonephropathy, formation of atypical glomeruli, and immaturity of parenchyma. The setting points out that up to date not identified noxae in the nephrogenic zone hamper primary steps of parenchyma development. In this situation, a possible therapeutic aim is to prolong nephrogenesis by medications. However, actual data provide information that administration of drugs is problematic due to an unexpectedly complex microanatomy of the nephrogenic zone, in niches so far not considered textured extracellular matrix and peculiar contacts between mesenchymal cell projections and epithelial stem cells via tunneling nanotubes. Thus, it remains to be figured out whether disturbance of morphogen signaling altered synthesis of extracellular matrix, disturbed cell-to-cell contacts, or modified interstitial fluid impair nephrogenic activity. Due to most unanswered questions, search for eligible drugs prolonging nephrogenesis and their reliable administration is a special challenge for the future.
Collapse
Affiliation(s)
- Will W Minuth
- Institute of Anatomy, University of Regensburg, 93053, Regensburg, Germany.
| |
Collapse
|
48
|
Amniotic fluid stem cell-derived vesicles protect from VEGF-induced endothelial damage. Sci Rep 2017; 7:16875. [PMID: 29203902 PMCID: PMC5715019 DOI: 10.1038/s41598-017-17061-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/21/2017] [Indexed: 11/14/2022] Open
Abstract
Injection of amniotic fluid stem cells (AFSC) delays the course of progression of renal fibrosis in animals with Alport Syndrome, enhancing kidney function and improving survival. The mechanisms responsible for these protective outcomes are still largely unknown. Here, we showed that vascular endothelial growth factor (VEGF) signaling within the glomeruli of Alport mice is strongly elevated early on in the disease, causing glomerular endothelial cell damage. Intraventricular injected AFSC that homed within the glomeruli showed strong modulation of the VEGF activity, particularly in glomerular endothelial cells. To investigate this phenomenon we hypothesized that extracellular vesicles (EVs) produced by the AFSC could be responsible for the observed renoprotection. AFSC derived EVs presented exosomal and stem cell markers on their surface membrane, including VEGFR1 and VEGFR2. EVs were able to modulate VEGF in glomerular endothelial cells by effectively trapping the excess VEGF through VEGFR1-binding preventing cellular damage. In contrast, VEGFR1/sVEGFR1 knockout EVs failed to show similar protection, thus indicating that VEGF trapping is a potentially viable mechanism for AFSC-EV mediated renoprotection. Taken together, our findings establish that EVs secreted by AFSC could target a specific signaling pathway within the glomerulus, thus representing a new potential glomerulus-specific targeted intervention.
Collapse
|
49
|
Grange C, Iampietro C, Bussolati B. Stem cell extracellular vesicles and kidney injury. Stem Cell Investig 2017; 4:90. [PMID: 29270416 DOI: 10.21037/sci.2017.11.02] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 10/17/2017] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs) appear as a new promising cell-free therapy for acute and chronic renal diseases. EVs retain characteristics of the cell of origin and those derived from stem cells may mimic their regenerative properties per se. In fact, EVs contain many active molecules such as proteins and RNA species that act on target cells through different mechanisms, stimulating proliferation and angiogenesis and reducing apoptosis and inflammation. There are several reports that demonstrate a general regenerative potential of EVs derived from mesenchymal stromal cells (MSCs) of different sources in kidney injury models. In addition, a promising new approach is the use of EVs in the graft perfusion solution for kidney conditioning before transplant. Here we summarize the application of EVs released by stem cells in preclinical models of acute and chronic renal damage, comparing animal models, use of EVs of different cell origin and of their sub-fractions, doses, route of administration and efficacy of treatment.
Collapse
Affiliation(s)
- Cristina Grange
- Department of Medical Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Corinne Iampietro
- Department of Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Benedetta Bussolati
- Department of Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| |
Collapse
|
50
|
Pillay P, Moodley K, Moodley J, Mackraj I. Placenta-derived exosomes: potential biomarkers of preeclampsia. Int J Nanomedicine 2017; 12:8009-8023. [PMID: 29184401 PMCID: PMC5673050 DOI: 10.2147/ijn.s142732] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Preeclampsia remains a leading cause of maternal and fetal mortality, due to ineffective treatment and diagnostic strategies, compounded by the lack of clarity on the etiology of the disorder. Although several clinical and biological markers of preeclampsia have been evaluated, they have proven to be ineffective in providing a definitive diagnosis during the various stages of the disorder. Exosomes have emerged as ideal biomarkers of pathological states, such as cancer, and have more recently gained interest in pregnancy-related complications, due to their role in cellular communication in normal and complicated pregnancies. This occurs as a result of the specific placenta-derived exosomal molecular cargo, which may be involved in normal pregnancy-associated immunological events, such as the maintenance of maternal-fetal tolerance. This review provides perspectives on placenta-derived exosomes as possible biomarkers for the diagnosis/prognosis of preeclampsia. Using keywords, online databases were searched to identify relevant publications to review the potential use of placenta-derived exosomes as biomarkers of preeclampsia.
Collapse
Affiliation(s)
- Preenan Pillay
- Discipline of Human Physiology, Nelson R Mandela School of Medicine, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Pearson Institute of Higher Education, Midrand, South Africa
| | - Kogi Moodley
- Discipline of Human Physiology, Nelson R Mandela School of Medicine, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Jagidesa Moodley
- Women’s Health and HIV Research Group, Nelson R Mandela School of Medicine, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Irene Mackraj
- Women’s Health and HIV Research Group, Nelson R Mandela School of Medicine, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|