1
|
Roseti L, Cavallo C, Desando G, D’Alessandro M, Grigolo B. Forty Years of the Use of Cells for Cartilage Regeneration: The Research Side. Pharmaceutics 2024; 16:1622. [PMID: 39771600 PMCID: PMC11677864 DOI: 10.3390/pharmaceutics16121622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The treatment of articular cartilage damage has always represented a problem of considerable practical interest for orthopedics. Over the years, many surgical techniques have been proposed to induce the growth of repairing tissue and limit degeneration. In 1994, the turning point occurred: implanted autologous cells paved the way for a new treatment option based more on regeneration than repair. Objectives: This review aims to outline biological and clinical advances, from the use of mature adult chondrocytes to cell-derived products, going through progenitor cells derived from bone marrow or adipose tissue and their concentrates for articular cartilage repair. Moreover, it highlights the relevance of gene therapy as a valuable tool for successfully implementing current regenerative treatments, and overcoming the limitations of the local delivery of growth factors. Conclusions: Finally, this review concludes with an outlook on the importance of understanding the role and mechanisms of action of the different cell compounds with a view to implementing personalized treatments.
Collapse
Affiliation(s)
| | - Carola Cavallo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.); (G.D.); (M.D.); (B.G.)
| | | | | | | |
Collapse
|
2
|
de Oliveira AT, Braga ARF, Miranda JRF, Fantinato-Neto P, Ambrósio CE. Mesenchymal stem cells in animal reproduction: sources, uses and scenario. BRAZILIAN JOURNAL OF VETERINARY MEDICINE 2024; 46:e002524. [PMID: 38737577 PMCID: PMC11087005 DOI: 10.29374/2527-2179.bjvm002524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 05/14/2024] Open
Abstract
Studies regarding mesenchymal stem cells turned up in the 1960's and this cell type created a great number of questions about its functions and applicability in science and medicine. When used with therapeutic intent, these cells present an inclination to migrate to sites of injury, inflammation or disease, where they secrete bioactive factors that stimulates the synthesis of new tissue. In this context, studies using rodents reported that MSCs promoted positive effects in the ovarian function in mice with premature aging of follicular reserve. In female bovines, experimental stem cell-based therapies have been used to either generate new oocytes with in vitro quality or stimulate such action in vivo. It is also reported, that the intraovarian application of mesenchymal stem cells generates a greater production of embryos in vitro and the production of early and expanded blastocysts. Additionally, analysis of ovarian tissue in animal subjected to treatment showed an increase in the number of developing follicles. Nevertheless, the treatments involving stem cells with different modes of application, different sources and different species were able to act on the hormonal, tissue, cellular and metabolic levels, generating positive results in the recovery and improvement of ovarian functions.
Collapse
Affiliation(s)
- Andrei Takeshita de Oliveira
- Undergraduate in Veterinary Medicine, Faculdade de Zootecnia e Engenharia de Alimentos (FZEA), Universidade de São Paulo (USP). Pirassununga, SP, Brazil.
| | - Antonio Rodrigues Ferreira Braga
- Undergraduate in Veterinary Medicine, Faculdade de Zootecnia e Engenharia de Alimentos (FZEA), Universidade de São Paulo (USP). Pirassununga, SP, Brazil.
| | - José Ricardo Fonseca Miranda
- Undergraduate in Veterinary Medicine, Faculdade de Zootecnia e Engenharia de Alimentos (FZEA), Universidade de São Paulo (USP). Pirassununga, SP, Brazil.
| | - Paulo Fantinato-Neto
- Veterinarian, DSc., Programa de Pós-Graduação em Biociência Animal, FZEA, USP, Pirassununga, SP, Brazil
| | - Carlos Eduardo Ambrósio
- Veterinarian, DSc., Departamento de Medicina Veterinária, FZEA, USP, Pirassununga, SP, Brazil
| |
Collapse
|
3
|
Nakamura A, Towheed T. Pathogenesis, assessment, and management of bone loss in axial spondyloarthritis. Semin Arthritis Rheum 2024; 64:152345. [PMID: 38103486 DOI: 10.1016/j.semarthrit.2023.152345] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION Axial spondyloarthritis (axSpA) presents a complex scenario where both new bone formation in entheseal tissues and significant trabecular bone loss coexist, emphasizing the intricate nature of bone dynamics in this context. METHODS A search of the literature was conducted to compose a narrative review exploring the pathogenesis, possible assessment methods, and potential management options for axSpA. RESULTS While chronic systemic and local inflammation contribute to bone loss, the mechanisms behind axSpA-associated bone loss exhibit distinct characteristics influenced by factors like mechanical stress and the gut microbiome. These factors directly or indirectly stimulate osteoclast differentiation and activation through the RANK-RANKL axis, while simultaneously impeding osteoblast differentiation via negative regulation of bone anabolic pathways, including the Wnt signaling pathway. This disruption in the balance between bone-resorbing osteoclasts and bone-forming osteoblasts contributes to overall bone loss in axSpA. Early evaluation at diagnosis is prudent for detecting bone changes. While traditional dual x-ray absorptiometry (DXA) has limitations due to potential overestimation from spinal new bone formation, alternative methods like trabecular bone score (TBS), quantitative CT (QCT), and quantitative ultrasound (QUS) show promise. However, their integration into routine clinical practice remains limited. In addition to approved anti-inflammatory drugs, lifestyle adjustments like regular exercise play a key role in preserving bone health. Tailoring interventions based on individual risk profiles holds potential for mitigating bone loss progression. CONCLUSION Recognizing the pivotal role of bone loss in axSpA underscores the importance of integrating regular assessments and effective management strategies into clinical practice. Given the multifaceted contributors to bone loss in axSpA, a multidisciplinary approach is essential.
Collapse
Affiliation(s)
- Akihiro Nakamura
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, Ontario, Canada; Translational Institute of Medicine, School of Medicine, Queen's University, Ontario, Canada; Kingston Health Science Centre, Kingston, Ontario, Canada.
| | - Tanveer Towheed
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, Ontario, Canada; Translational Institute of Medicine, School of Medicine, Queen's University, Ontario, Canada; Kingston Health Science Centre, Kingston, Ontario, Canada.
| |
Collapse
|
4
|
Rosen CJ, Horowitz MC. Nutrient regulation of bone marrow adipose tissue: skeletal implications of weight loss. Nat Rev Endocrinol 2023; 19:626-638. [PMID: 37587198 PMCID: PMC10592027 DOI: 10.1038/s41574-023-00879-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/19/2023] [Indexed: 08/18/2023]
Abstract
Adipose tissue is a dynamic component of the bone marrow, regulating skeletal remodelling and secreting paracrine and endocrine factors that can affect haematopoiesis, as well as potentially nourishing the bone marrow during periods of stress. Bone marrow adipose tissue is regulated by multiple factors, but particularly nutrient status. In this Review, we examine how bone marrow adipocytes originate, their function in normal and pathological states and how bone marrow adipose tissue modulates whole-body homoeostasis through actions on bone cells, haematopoietic stem cells and extra-medullary adipocytes during nutritional challenges. We focus on both rodent models and human studies to help understand the unique marrow adipocyte, its response to the external nutrient environment and its effects on the skeleton. We finish by addressing some critical questions that to date remain unanswered.
Collapse
Affiliation(s)
| | - Mark C Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
5
|
Lee E, Moon JY, Ko JY, Park SY, Im GI. GSTT1 as a Predictive Marker and Enhancer for Osteogenic Potential of Human Adipose-Derived Stromal/Stem Cells. J Bone Miner Res 2023; 38:1480-1496. [PMID: 37537994 DOI: 10.1002/jbmr.4893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 07/23/2023] [Accepted: 08/01/2023] [Indexed: 08/05/2023]
Abstract
Adipose-derived stromal/stem cells (ASCs) have been extensively studied as cell sources for regenerative medicine for bone because of their excellent proliferative capacity and the ability to obtain a large number of cells with minimal donor morbidity. On the other hand, the differentiation potential of ASCs is generally lower than that of bone marrow-derived stromal/stem cells and varies greatly depending on donors. In this study, we mined a marker that can predict the osteogenic potential of ASC clones and also investigated the usefulness of the molecule as the enhancer of osteogenic differentiation of ASCs as well as its mechanism of action. Through RNA-seq gene analysis, we discovered that GSTT1 (Glutathione S-transferase theta-1) was the most distinguished gene marker between highly osteogenic and poorly osteogenic ASC clones. Knockdown of GSTT1 in high osteogenic ASCs by siGSTT1 treatment reduced mineralized matrix formation. On the other hand, GSTT1 overexpression by GSTT1 transfection or GSTT1 recombinant protein treatment enhanced osteogenic differentiation of low osteogenic ASCs. Metabolomic analysis confirmed significant changes of metabolites related to bone differentiation in ASCs transfected with GSTT1. A high total antioxidant capacity, low levels of cellular reactive oxygen species, and increased GSH/GSSG ratios were also detected in GSTT1-transfected ASCs. When the in vivo effect of GSTT1-transfected ASCs on bone regeneration was investigated with segmental long-bone defect model in rats, bone regeneration was significantly better after implantation of GSTT1-transfected ASCs compared with that of control vector-transfected ASCs. In conclusion, GSTT1 can be a useful marker to screen the highly osteogenic ASC clones and also a therapeutic factor to enhance the osteogenic differentiation of poorly osteogenic ASC clones. © 2023 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Eugene Lee
- Research Institute for Integrative Regenerative Biomedical Engineering, Dongguk University, Goyang, Republic of Korea
| | - Jae-Yeon Moon
- Research Institute for Integrative Regenerative Biomedical Engineering, Dongguk University, Goyang, Republic of Korea
| | - Ji-Yun Ko
- Research Institute for Integrative Regenerative Biomedical Engineering, Dongguk University, Goyang, Republic of Korea
| | - Seo-Young Park
- Research Institute for Integrative Regenerative Biomedical Engineering, Dongguk University, Goyang, Republic of Korea
| | - Gun-Il Im
- Research Institute for Integrative Regenerative Biomedical Engineering, Dongguk University, Goyang, Republic of Korea
- Department of Orthopedics, Dongguk University Ilsan Hospital, Goyang, Republic of Korea
| |
Collapse
|
6
|
Liu Y, Zhang X, Xiao C, Liu B. Engineered hydrogels for peripheral nerve repair. Mater Today Bio 2023; 20:100668. [PMID: 37273791 PMCID: PMC10232914 DOI: 10.1016/j.mtbio.2023.100668] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/06/2023] [Accepted: 05/16/2023] [Indexed: 06/06/2023] Open
Abstract
Peripheral nerve injury (PNI) is a complex disease that often appears in young adults. It is characterized by a high incidence, limited treatment options, and poor clinical outcomes. This disease not only causes dysfunction and psychological disorders in patients but also brings a heavy burden to the society. Currently, autologous nerve grafting is the gold standard in clinical treatment, but complications, such as the limited source of donor tissue and scar tissue formation, often further limit the therapeutic effect. Recently, a growing number of studies have used tissue-engineered materials to create a natural microenvironment similar to the nervous system and thus promote the regeneration of neural tissue and the recovery of impaired neural function with promising results. Hydrogels are often used as materials for the culture and differentiation of neurogenic cells due to their unique physical and chemical properties. Hydrogels can provide three-dimensional hydration networks that can be integrated into a variety of sizes and shapes to suit the morphology of neural tissues. In this review, we discuss the recent advances of engineered hydrogels for peripheral nerve repair and analyze the role of several different therapeutic strategies of hydrogels in PNI through the application characteristics of hydrogels in nerve tissue engineering (NTE). Furthermore, the prospects and challenges of the application of hydrogels in the treatment of PNI are also discussed.
Collapse
Affiliation(s)
- Yao Liu
- Hand and Foot Surgery Department, First Hospital of Jilin University, Xinmin Street, Changchun, 130061, PR China
| | - Xiaonong Zhang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Bin Liu
- Hand and Foot Surgery Department, First Hospital of Jilin University, Xinmin Street, Changchun, 130061, PR China
| |
Collapse
|
7
|
Cicione C, Vadalà G, Di Giacomo G, Tilotta V, Ambrosio L, Russo F, Zampogna B, Cannata F, Papalia R, Denaro V. Micro-fragmented and nanofat adipose tissue derivatives: In vitro qualitative and quantitative analysis. Front Bioeng Biotechnol 2023; 11:911600. [PMID: 36733959 PMCID: PMC9887143 DOI: 10.3389/fbioe.2023.911600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Introduction: Adipose tissue is widely exploited in regenerative medicine thanks to its trophic properties, mainly based on the presence of adipose-derived stromal cells. Numerous devices have been developed to promote its clinical use, leading to the introduction of one-step surgical procedures to obtain minimally manipulated adipose tissue derivatives. However, only a few studies compared their biological properties. This study aimed to characterize micro-fragmented (MAT) and nanofat adipose tissue (NAT) obtained with two different techniques. Methods: MAT, NAT and unprocessed lipoaspirate were collected from surgical specimens. RNA extraction and collagenase isolation of stromal vascular fraction (SVF) were performed. Tissue sections were analysed by histological and immunohistochemical (collagen type I, CD31, CD34 and PCNA) staining to assess tissue morphology and cell content. qPCR was performed to evaluate the expression of stemness-related (SOX2, NANOG and OCT3/4), extracellular matrix (COL1A1) and inflammatory genes (IL1β, IL6 and iNOS). Furthermore, multilineage differentiation was assessed following culture in adipogenic and osteogenic media and staining with Oil Red O and Alizarin red. ASC immunophenotype was assessed by flow cytometric analysis of CD90, CD105, CD73 and CD45. Results: Histological and immunohistochemical results showed an increased amount of stroma and a reduction of adipocytes in MAT and NAT, with the latter displaying the highest content of collagen type I, CD31, CD34 and PCNA. From LA to MAT and NAT, an increasing expression of NANOG, SOX2, OCT3/4, COL1A1 and IL6 was noted, while no significant differences in terms of IL1β and iNOS emerged. No statistically significant differences were noted between NAT and SVF in terms of stemness-related genes, while the latter demonstrated a significantly higher expression of stress-related markers. SVF cells derived from all three samples (LA, MAT, and NAT) showed a similar ASC immunoprofile as well as osteogenic and adipogenic differentiation. Discussion: Our results showed that both MAT and NAT techniques allowed the rapid isolation of ASC-rich grafts with a high anabolic and proliferative potential. However, NAT showed the highest levels of extracellular matrix content, replicating cells, and stemness gene expression. These results may provide precious clues for the use of adipose tissue derivatives in the clinical setting.
Collapse
Affiliation(s)
- Claudia Cicione
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Gianluca Vadalà
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy,Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy,*Correspondence: Gianluca Vadalà,
| | - Giuseppina Di Giacomo
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Veronica Tilotta
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Luca Ambrosio
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy,Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Fabrizio Russo
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy,Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Biagio Zampogna
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy,Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Francesca Cannata
- Operative Research Unit of Endocrinology and Diabetes, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Rocco Papalia
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy,Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Vincenzo Denaro
- Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| |
Collapse
|
8
|
Early Transcriptional Changes of Adipose-Derived Stem Cells (ADSCs) in Cell Culture. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58091249. [PMID: 36143926 PMCID: PMC9501538 DOI: 10.3390/medicina58091249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/27/2022] [Accepted: 08/28/2022] [Indexed: 11/22/2022]
Abstract
Background and Objectives: While autologous fat grafting has been carried out in the clinical field for many years, the utilization of isolated and cultured adipose-derived stem cells (ADSCs) is highly restricted in many countries. However, ADSCs are under investigation currently and heavily researched in many cell-based therapy approaches in the field of regenerative medicine. Objective: For the utilization of future cell-based therapies with ADSCs, in vitro cell expansion might be necessary in many cases. Thus, the cellular characteristics of ADSCs may be altered though the process of being cultured. The aim of this study was to assess changes in the gene expression profile of ADSCs after cell expansion for 48 h. Materials and Methods: Isolated ADSCs from five different donors were used for in vitro expansion. For the evaluation of the gene expression profile, mRNA deep Next-Generation Sequencing was performed to evaluate the differences between cultured and freshly isolated cells. Results: Our study gives insight into transcriptional changes in ADSCs after a short cell cultivation period. This includes the most prominent upregulated genes such as PPL, PRR15, CCL11 and ABCA9, as well the most downregulated genes, which are FOSB, FOS, EGR1 and DUSP6. Furthermore, we showed different biological processes that changed during short-term cell expansion, which led to downregulation of fat-associated metabolism hormone processes and to an upregulation of extracellular matrix-associated genes. Conclusion: In conclusion, our study reveals a detailed insight into early changes in the gene expression profile of cultured ADSCs. Our results can be utilized in future experiments.
Collapse
|
9
|
Mantovani M, Gennai A, Russo PR. A new approach to regenerative medicine in gynecology. Int J Gynaecol Obstet 2022; 157:536-543. [PMID: 34463351 DOI: 10.1002/ijgo.13906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/05/1901] [Accepted: 08/26/1901] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To evaluate the efficacy of the injection of microfragmented adipose tissue in the treatment of women with genitourinary syndrome of menopause (GSM). METHODS This observational cohort study included 12 women who received one session of multiple injections of microfragmented adipose tissue using the SEFFIGYN™ medical device. Symptoms such as burning, itching, dryness, pain on penetration, pain during deep intercourse, and pain on urination were assessed before the patient's treatment (T0), after 15 days (T15), and after 5 months (5Mo) using the Numerical Rating Scale (NRS). RESULTS An improvement of vulvar trophism was clinically evident already 2 weeks after treatment; all symptoms were notably attenuated compared with the initial visit, as demonstrated by statistically significant reductions of the NRS scores (P = 0.003 for itching, P = 0.008 for pain on urination, and P < 0.001 for the other symptoms, Sign test). Moreover, all symptoms continued to improve over time. All patients reported a positive change in their quality of life and a resumption of sexual life. CONCLUSION The use of microfragmented adipose tissue in GSM is promising. Nevertheless, more studies will be fundamental to exclude a potential placebo effect and better understand the underlying molecular mechanism of action.
Collapse
Affiliation(s)
| | - Alessandro Gennai
- Plastic Reconstructive and Aesthetic Surgeon Medical Director STUDIO GENNAI, Bologna, Italy
| | | |
Collapse
|
10
|
Tevlin R, desJardins-Park H, Huber J, DiIorio S, Longaker M, Wan D. Musculoskeletal tissue engineering: Adipose derived stromal cell implementation for the treatment of osteoarthritis. Biomaterials 2022; 286:121544. [DOI: 10.1016/j.biomaterials.2022.121544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 06/23/2021] [Accepted: 09/13/2021] [Indexed: 11/02/2022]
|
11
|
Bond ST, Calkin AC, Drew BG. Adipose-Derived Extracellular Vesicles: Systemic Messengers and Metabolic Regulators in Health and Disease. Front Physiol 2022; 13:837001. [PMID: 35283789 PMCID: PMC8905439 DOI: 10.3389/fphys.2022.837001] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Adipose tissue is comprised of a heterogeneous population of cells that co-operate to perform diverse physiological roles including endocrine-related functions. The endocrine role of adipose tissue enables it to communicate nutritional and health cues to other organs, such as the liver, muscle, and brain, in order to regulate appetite and whole body metabolism. Adipose tissue dysfunction, which is often observed in obesity, is associated with changes in the adipose secretome, which can subsequently contribute to disease pathology. Indeed, secreted bioactive factors released from adipose tissue contribute to metabolic homeostasis and likely play a causal role in disease; however, what constitutes the entirety of the adipose tissue secretome is still poorly understood. Recent advances in nanotechnology have advanced this field substantially and have led to the identification of small, secreted particles known as extracellular vesicles (EVs). These small nano-sized lipid envelopes are released by most cell types and are capable of systemically delivering bioactive molecules, such as nucleic acids, proteins, and lipids. EVs interact with target cells to deliver specific cargo that can then elicit effects in various tissues throughout the body. Adipose tissue has recently been shown to secrete EVs that can communicate with the periphery to maintain metabolic homeostasis, or under certain pathological conditions, drive disease. In this review, we discuss the current landscape of adipose tissue-derived EVs, with a focus on their role in the regulation of metabolic homeostasis and disease pathology.
Collapse
Affiliation(s)
- Simon T Bond
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Melbourne, VIC, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - Anna C Calkin
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Melbourne, VIC, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - Brian G Drew
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Monash University, Melbourne, VIC, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
12
|
Current strategies to improve erectile function in patients undergoing radical prostatectomy - postoperative scenario. Urol Oncol 2022; 40:87-94. [DOI: 10.1016/j.urolonc.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/01/2021] [Accepted: 12/07/2021] [Indexed: 11/15/2022]
|
13
|
Krawczenko A, Klimczak A. Adipose Tissue-Derived Mesenchymal Stem/Stromal Cells and Their Contribution to Angiogenic Processes in Tissue Regeneration. Int J Mol Sci 2022; 23:ijms23052425. [PMID: 35269568 PMCID: PMC8910401 DOI: 10.3390/ijms23052425] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are widely described in the context of their regenerative and immunomodulatory activity. MSCs are isolated from various tissues and organs. The most frequently described sources are bone marrow and adipose tissue. As stem cells, MSCs are able to differentiate into other cell lineages, but they are usually reported with respect to their paracrine potential. In this review, we focus on MSCs derived from adipose tissue (AT-MSCs) and their secretome in regeneration processes. Special attention is given to the contribution of AT-MSCs and their derivatives to angiogenic processes described mainly in the context of angiogenic dysfunction. Finally, we present clinical trials registered to date that concern the application of AT-MSCs and their secretome in various medical conditions.
Collapse
|
14
|
Zanata F, Curley L, Martin E, Bowles A, Bunnell BA, Wu X, Ferreira LM, Gimble JM. Comparative Analysis of Human Adipose-Derived Stromal/Stem Cells and Dermal Fibroblasts. Stem Cells Dev 2021; 30:1171-1178. [PMID: 34486404 DOI: 10.1089/scd.2021.0164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Dermal fibroblasts (DFs) share several qualities with mesenchymal stem cell/multipotent stromal cells (MSCs) derived from various tissues, including adipose-derived stromal/stem cells (ASCs). ASCs and DFs are morphologically comparable and both cell types can be culture expanded through the utilization of their plastic-adherence properties. Despite these similar characteristics, numerous studies indicate that ASC and DF display distinct therapeutic benefits in clinical applications. To more accurately distinguish between these cell types, human DFs and ASCs isolated from three individual donors were analyzed for multipotency and cell surface marker expressions. The detection of cell surface markers, CD29, CD34, CD44, CD73, CD90, and CD105, were used for phenotypic characterization of the DFs and ASCs. Furthermore, both cell types underwent lineage differentiation based on histochemical staining and the expression of adipogenic related genes, CCAAT/Enhancer-Binding Protein alpha (CEBPα), Peroxisome proliferator-activated receptor gamma (PPARγ), UCP1, Leptin (LEP), and Adiponectin (ADIPOQ); and osteogenic related genes, Runt related transcription factor 2 (Runx2), Alkaline phosphatase (ALPL), Osteocalcin (OCN), and Osteopontin (OPN). Evidence provided by this study demonstrates similarities between donor-matched ASC and DF with respect to morphology, surface marker expression, differentiation potential, and gene expression, although appearance of enhanced adipogenesis in the ASC based solely on spectrophotometric analyses with no significant difference in real-time polymerase chain reaction detection of adipogenic biomarkers. Thus, there is substantial overlap between the ASC and DF phenotypes based on biochemical and differentiation metrics.
Collapse
Affiliation(s)
- Fabiana Zanata
- Plastic Surgery Division, Universidade Federal de Sao Paulo UNIFESP/EPM, Sao Paulo, Brazil
| | | | - Elizabeth Martin
- Center for Stem Cell Research and Regenerative Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Annie Bowles
- Center for Stem Cell Research and Regenerative Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Bruce A Bunnell
- Department of Microbiology, Immunology, and Genetics, The University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - Xiying Wu
- La Cell LLC, New Orleans, Louisiana, USA
| | - Lydia Masako Ferreira
- Plastic Surgery Division, Universidade Federal de Sao Paulo UNIFESP/EPM, Sao Paulo, Brazil
| | - Jeffrey M Gimble
- La Cell LLC, New Orleans, Louisiana, USA.,Center for Stem Cell Research and Regenerative Medicine, Tulane University, New Orleans, Louisiana, USA
| |
Collapse
|
15
|
El-Gendy H, Hawass SED, Awad M, Mohsen MA, Amin M, Abdalla HA, Fouad S, Lotfy A. Comparative study between human mesenchymal stem cells and etanercept as immunomodulatory agents in rat model of rheumatoid arthritis. Immunol Res 2021; 68:255-268. [PMID: 32734446 DOI: 10.1007/s12026-020-09132-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
To compare human adipose tissue mesenchymal stem cells (AT-MSCs) and etanercept as immunomodulatory agents for collagen-induced arthritis (CIA). CIA was induced by rats' immunization with collagen type II (CII) in complete Freund's adjuvant in days 0 and 7. Before the onset of CIA, prevention group received five doses of AT-MSCS intraperitoneally. After establishment of arthritis, rats received either five doses of AT-MSCs or phosphate-buffered saline (PBS) intraperitoneally or six doses of etanercept subcutaneously. Clinical and histopathological evaluation were performed in all groups; serum levels of tumor necrosis factor-α (TNF-α), interleukin-10 (IL-10), and anti-collagen II were assessed by enzyme-linked immunosorbent assay (ELISA). A total percent of autoreactive T and regulatory T (Treg) cells were quantified using spleen immune histochemical analysis. AT-MSCs were able to delay the onset of CIA, suppress the ongoing clinical and histopathological signs, decrease serum levels of TNF-α and anti-collagen type II, and downregulate the autoreactive T cells as etanercept. AT-MSCs were more potent in Treg cells upregulation, producing high serum levels of IL10. AT-MSCs might have a therapeutic effect in CIA via their potency in immune cell education, representing an effective new promising approach in rheumatoid arthritis in human.
Collapse
Affiliation(s)
- Heba El-Gendy
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Mansoura University, Mansoura, Egypt.
| | - Salah El-Deen Hawass
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Manal Awad
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mona Ahmad Mohsen
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Maha Amin
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | | - Samah Fouad
- Medical Experimental Research Centre , Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed Lotfy
- eBiotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
16
|
Rohani Ivari J, Mahdipour E. Adipose tissue versus stem cell-derived small extracellular vesicles to enhance the healing of acute burns. Regen Med 2021; 16:629-641. [PMID: 34259030 DOI: 10.2217/rme-2020-0199] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Aim: Proper healing of extensive burns remains a healthcare challenge. In the present study, we proposed a distinct therapeutic application of adipose tissue and small extracellular vesicles isolated from human menstrual blood-derived mesenchymal stem cells (MenSC) small extracellular vesicles (sEVs) to enhance the repair of third-degree burn injury. Materials & methods: Mouse model of third-degree burn was used. Adipose tissue in the form of nano-fat (NF) and MenSC-sEVs was injected subcutaneously at the site of injuries. Results: NF and sEVs were capable of enhancing wound closure and increasing neoangiogenesis. NF was also effective in accelerating the formation of granulation tissue and boosting the thickness of the new epithelial layer. Conclusion: This study demonstrates the effectiveness of NF and MenSC-sEVs as promising therapeutic approaches to facilitate the repair of skin burns.
Collapse
Affiliation(s)
- Jalil Rohani Ivari
- Department of Medical Biotechnology & Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, 1696700, Iran
| | - Elahe Mahdipour
- Department of Medical Biotechnology & Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, 1696700, Iran
| |
Collapse
|
17
|
Adipose-Derived Exosomes as Possible Players in the Development of Insulin Resistance. Int J Mol Sci 2021; 22:ijms22147427. [PMID: 34299048 PMCID: PMC8304687 DOI: 10.3390/ijms22147427] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/25/2022] Open
Abstract
Adipose tissue (AT) is an endocrine organ involved in the management of energy metabolism via secretion of adipokines, hormones, and recently described secretory microvesicles, i.e., exosomes. Exosomes are rich in possible biologically active factors such as proteins, lipids, and RNA. The secretory function of adipose tissue is affected by pathological processes. One of the most important of these is obesity, which triggers adipose tissue inflammation and adversely affects the release of beneficial adipokines. Both processes may lead to further AT dysfunction, contributing to changes in whole-body metabolism and, subsequently, to insulin resistance. According to recent data, changes within the production, release, and content of exosomes produced by AT may be essential to understand the role of adipose tissue in the development of metabolic disorders. In this review, we summarize actual knowledge about the possible role of AT-derived exosomes in the development of insulin resistance, highlighting methodological challenges and potential gains resulting from exosome studies.
Collapse
|
18
|
Lopez-Santalla M, Bueren JA, Garin MI. Mesenchymal stem/stromal cell-based therapy for the treatment of rheumatoid arthritis: An update on preclinical studies. EBioMedicine 2021; 69:103427. [PMID: 34161884 PMCID: PMC8237294 DOI: 10.1016/j.ebiom.2021.103427] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 12/21/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovial inflammation and progressive joint destruction and is a primary cause of disability worldwide. Despite the existence of numerous anti-rheumatic drugs, a significant number of patients with RA do not respond or are intolerant to current treatments. Mesenchymal stem/stromal cell (MSCs) therapy represents a promising therapeutic tool to treat RA, mainly attributable to the immunomodulatory effects of these cells. This review comprises a comprehensive analysis of the scientific literature related to preclinical studies of MSC-based therapy in RA to analyse key aspects of current protocols as well as novel approaches which aim to improve the efficacy of MSC-based therapy.
Collapse
Affiliation(s)
- Mercedes Lopez-Santalla
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid; Spain; Advanced Therapy Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM).
| | - Juan A Bueren
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid; Spain; Advanced Therapy Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM)
| | - Marina I Garin
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid; Spain; Advanced Therapy Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM).
| |
Collapse
|
19
|
Moccia C, Haase K. Engineering Breast Cancer On-chip-Moving Toward Subtype Specific Models. Front Bioeng Biotechnol 2021; 9:694218. [PMID: 34249889 PMCID: PMC8261144 DOI: 10.3389/fbioe.2021.694218] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the second leading cause of death among women worldwide, and while hormone receptor positive subtypes have a clear and effective treatment strategy, other subtypes, such as triple negative breast cancers, do not. Development of new drugs, antibodies, or immune targets requires significant re-consideration of current preclinical models, which frequently fail to mimic the nuances of patient-specific breast cancer subtypes. Each subtype, together with the expression of different markers, genetic and epigenetic profiles, presents a unique tumor microenvironment, which promotes tumor development and progression. For this reason, personalized treatments targeting components of the tumor microenvironment have been proposed to mitigate breast cancer progression, particularly for aggressive triple negative subtypes. To-date, animal models remain the gold standard for examining new therapeutic targets; however, there is room for in vitro tools to bridge the biological gap with humans. Tumor-on-chip technologies allow for precise control and examination of the tumor microenvironment and may add to the toolbox of current preclinical models. These new models include key aspects of the tumor microenvironment (stroma, vasculature and immune cells) which have been employed to understand metastases, multi-organ interactions, and, importantly, to evaluate drug efficacy and toxicity in humanized physiologic systems. This review provides insight into advanced in vitro tumor models specific to breast cancer, and discusses their potential and limitations for use as future preclinical patient-specific tools.
Collapse
Affiliation(s)
| | - Kristina Haase
- European Molecular Biology Laboratory, European Molecular Biology Laboratory Barcelona, Barcelona, Spain
| |
Collapse
|
20
|
Crohn's Disease Increases the Mesothelial Properties of Adipocyte Progenitors in the Creeping Fat. Int J Mol Sci 2021; 22:ijms22084292. [PMID: 33924264 PMCID: PMC8074767 DOI: 10.3390/ijms22084292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022] Open
Abstract
Our understanding of the interplay between human adipose tissue and the immune system is limited. The mesothelium, an immunologically active structure, emerged as a source of visceral adipose tissue. After investigating the mesothelial properties of human visceral and subcutaneous adipose tissue and their progenitors, we explored whether the dysfunctional obese and Crohn's disease environments influence the mesothelial/mesenchymal properties of their adipocyte precursors, as well as their ability to mount an immune response. Using a tandem transcriptomic/proteomic approach, we evaluated the mesothelial and mesenchymal expression profiles in adipose tissue, both in subjects covering a wide range of body-mass indexes and in Crohn's disease patients. We also isolated adipose tissue precursors (adipose-derived stem cells, ASCs) to assess their mesothelial/mesenchymal properties, as well as their antigen-presenting features. Human visceral tissue presented a mesothelial phenotype not detected in the subcutaneous fat. Only ASCs from mesenteric adipose tissue, named creeping fat, had a significantly higher expression of the hallmark mesothelial genes mesothelin (MSLN) and Wilms' tumor suppressor gene 1 (WT1), supporting a mesothelial nature of these cells. Both lean and Crohn's disease visceral ASCs expressed equivalent surface percentages of the antigen-presenting molecules human leucocyte antigen-DR isotype (HLA-DR) and CD86. However, lean-derived ASCs were predominantly HLA-DR dim, whereas in Crohn's disease, the HLA-DR bright subpopulation was increased 3.2-fold. Importantly, the mesothelial-enriched Crohn's disease precursors activated CD4+ T-lymphocytes. Our study evidences a mesothelial signature in the creeping fat of Crohn's disease patients and its progenitor cells, the latter being able to present antigens and orchestrate an immune response.
Collapse
|
21
|
Llewellyn SH, Faroni A, Iliut M, Bartlam C, Vijayaraghavan A, Reid AJ. Graphene Oxide Substrate Promotes Neurotrophic Factor Secretion and Survival of Human Schwann-Like Adipose Mesenchymal Stromal Cells. Adv Biol (Weinh) 2021; 5:e2000271. [PMID: 33852181 DOI: 10.1002/adbi.202000271] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 02/04/2021] [Indexed: 11/11/2022]
Abstract
Mesenchymal stromal cells from adipose tissue (AD-MSCs) exhibit favorable clinical traits for autologous transplantation and can develop 'Schwann-like' phenotypes (sAD-MSCs) to improve peripheral nerve regeneration, where severe injuries yield insufficient recovery. However, sAD-MSCs regress without biochemical stimulation and detach from conduits under unfavorable transplant conditions, negating their paracrine effects. Graphene-derived materials support AD-MSC attachment, regulating cell adhesion and function through physiochemistry and topography. Graphene oxide (GO) is a suitable substrate for human sAD-MSCs incubation toward severe peripheral nerve injuries by evaluating transcriptome changes, neurotrophic factor expression over a 7-days period, and cell viability in apoptotic conditions is reported. Transcriptome changes from GO incubation across four patients are minor compared to biological variance. Nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and glial-derived neurotrophic factor (GDNF) gene expression is unchanged from sAD-MSCs on GO substrates, but NGF and GDNF protein secretion increase at day 3 and 7. Secretome changes do not improve dorsal root ganglia neuron axon regeneration in conditioned media culture models. Fewer sAD-MSCs detach from GO substrates compared to glass following phosphate buffer saline exposure, which simulates apoptotic conditions. Overall, GO substrates are compatible with sAD-MSC primed for peripheral nerve regeneration strategies and protect the cell population in harsh environments.
Collapse
Affiliation(s)
- Steffan H Llewellyn
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL, UK.,Department of Materials and National Graphene Institute, The University of Manchester, Manchester, M13 9PL, UK
| | - Alessandro Faroni
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL, UK
| | - Maria Iliut
- Department of Materials and National Graphene Institute, The University of Manchester, Manchester, M13 9PL, UK
| | - Cian Bartlam
- Department of Materials and National Graphene Institute, The University of Manchester, Manchester, M13 9PL, UK.,Institute of Physics, EIT 2, Bundeswehr University Munich, Neubiberg, 85577, Germany
| | - Aravind Vijayaraghavan
- Department of Materials and National Graphene Institute, The University of Manchester, Manchester, M13 9PL, UK
| | - Adam J Reid
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PL, UK.,Department of Plastic Surgery & Burns, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M23 9LT, UK
| |
Collapse
|
22
|
Al-Ghadban S, Bunnell BA. Adipose Tissue-Derived Stem Cells: Immunomodulatory Effects and Therapeutic Potential. Physiology (Bethesda) 2021; 35:125-133. [PMID: 32027561 DOI: 10.1152/physiol.00021.2019] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Adipose-derived stem cells (ASCs) can self-renew and differentiate along multiple cell lineages. ASCs are also potently anti-inflammatory due to their inherent ability to regulate the immune system by secreting anti-inflammatory cytokines and growth factors that play a crucial role in the pathology of many diseases, including multiple sclerosis, diabetes mellitus, Crohn's, SLE, and graft-versus-host disease. The immunomodulatory effects and mechanisms of action of ASCs on pathological conditions are reviewed here.
Collapse
Affiliation(s)
- Sara Al-Ghadban
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Bruce A Bunnell
- Center for Stem Cell Research & Regenerative Medicine, Tulane University School of Medicine, New Orleans, Louisiana.,Department of Pharmacology, School of Medicine, Tulane University, New Orleans, Louisiana
| |
Collapse
|
23
|
Hillers-Ziemer LE, Arendt LM. Weighing the Risk: effects of Obesity on the Mammary Gland and Breast Cancer Risk. J Mammary Gland Biol Neoplasia 2020; 25:115-131. [PMID: 32519090 PMCID: PMC7933979 DOI: 10.1007/s10911-020-09452-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022] Open
Abstract
Obesity is a preventable risk factor for breast cancer following menopause. Regardless of menopausal status, obese women who develop breast cancer have a worsened prognosis. Breast tissue is comprised of mammary epithelial cells organized into ducts and lobules and surrounded by adipose-rich connective tissue. Studies utilizing multiple in vivo models of obesity as well as human breast tissue have contributed to our understanding of how obesity alters mammary tissue. Localized changes in mammary epithelial cell populations, elevated secretion of adipokines and angiogenic mediators, inflammation within mammary adipose tissue, and remodeling of the extracellular matrix may result in an environment conducive to breast cancer growth. Despite these significant alterations caused by obesity within breast tissue, studies have suggested that some, but not all, obesity-induced changes may be mitigated with weight loss. Here, we review our current understanding regarding the impact of obesity on the breast microenvironment, how obesity-induced changes may contribute to breast tumor progression, and the impact of weight loss on the breast microenvironment.
Collapse
Affiliation(s)
- Lauren E Hillers-Ziemer
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI, 53706, USA
| | - Lisa M Arendt
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI, 53706, USA.
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA.
| |
Collapse
|
24
|
Hoefner C, Muhr C, Horder H, Wiesner M, Wittmann K, Lukaszyk D, Radeloff K, Winnefeld M, Becker M, Blunk T, Bauer-Kreisel P. Human Adipose-Derived Mesenchymal Stromal/Stem Cell Spheroids Possess High Adipogenic Capacity and Acquire an Adipose Tissue-like Extracellular Matrix Pattern. Tissue Eng Part A 2020; 26:915-926. [PMID: 32070231 DOI: 10.1089/ten.tea.2019.0206] [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] [Indexed: 12/14/2022] Open
Abstract
Adipose-derived mesenchymal stromal/stem cells (ASCs) represent a commonly used cell source for adipose tissue engineering. In this context, ASCs have routinely been cultured in conventional 2D culture and applied as single cell suspension for seeding onto scaffold materials or direct injection. However, this approach is associated with the loss of their intrinsic 3D microenvironment and leads to impaired regenerative capacity of the cells. Thus, the application of ASCs as self-assembled 3D spheroids with cells residing in their own matrix is an attractive alternative. However, characterization of the structural features and differentiation capacity of the spheroids is necessary to effectively apply them as building blocks in adipose tissue engineering. In this study, we focus on extracellular matrix (ECM) development in ASC spheroids, as well as adipogenic differentiation in comparison to conventional 2D culture using different induction protocols. Reproducible assembly of ASCs into spheroids was achieved within 24 h using the liquid overlay technique. Undifferentiated spheroids displayed a stromal ECM pattern, with fibronectin, collagen V, and VI as the main components. In the course of adipogenesis, a dynamic shift in the ECM composition toward an adipogenic phenotype was observed, associated with enhanced expression of laminin, collagen I, IV, V, and VI, similar to native fat. Furthermore, adipogenic differentiation was enhanced in spheroids as compared with 2D cultured cells, with the spheroids needing a distinctly shorter adipogenic stimulus to sustain adipogenesis, which was demonstrated based on analysis of triglyceride content and adipogenic marker gene expression. In summary, culturing ASCs as spheroids can enhance their adipogenic capacity and generate adipose-like microtissues, which may be a promising cell delivery strategy for adipose tissue engineering approaches. Impact statement Adipose-derived mesenchymal stromal/stem cells (ASCs) as a widely used cell source for adipose tissue engineering have been shown to be limited in their regenerative capacity when applied as single cells. As an alternative approach, the delivery as spheroids, consisting of cells in a 3D context, may be favorable. However, insights into extracellular matrix (ECM) development and efficient adipogenic differentiation are required for their effective application. In this study, we show that differentiated ASC spheroids develop an ECM, resembling native adipose tissue. Furthermore, the ASC spheroids exhibited a superior differentiation capacity as compared with conventional 2D culture, and required only a short adipogenic induction stimulus. Our results identify ASC-derived spheroids as an attractive cell delivery method for adipose tissue engineering approaches.
Collapse
Affiliation(s)
- Christiane Hoefner
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Christian Muhr
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Hannes Horder
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Miriam Wiesner
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Katharina Wittmann
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Daniel Lukaszyk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Katrin Radeloff
- Department of Otorhinolaryngology, University of Würzburg, Würzburg, Germany
| | | | - Matthias Becker
- Institute for Medical Radiation and Cell Research, University of Würzburg, Würzburg, Germany
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| | - Petra Bauer-Kreisel
- Department of Trauma, Hand, Plastic and Reconstructive Surgery and University of Würzburg, Würzburg, Germany
| |
Collapse
|
25
|
Zhong YC, Wang SC, Han YH, Wen Y. Recent Advance in Source, Property, Differentiation, and Applications of Infrapatellar Fat Pad Adipose-Derived Stem Cells. Stem Cells Int 2020; 2020:2560174. [PMID: 32215015 PMCID: PMC7081037 DOI: 10.1155/2020/2560174] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/12/2020] [Accepted: 02/20/2020] [Indexed: 12/18/2022] Open
Abstract
Infrapatellar fat pad (IPFP) can be easily obtained during knee surgery, which avoids the damage to patients for obtaining IPFP. Infrapatellar fat pad adipose-derived stem cells (IPFP-ASCs) are also called infrapatellar fat pad mesenchymal stem cells (IPFP-MSCs) because the morphology of IPFP-ASCs is similar to that of bone marrow mesenchymal stem cells (BM-MSCs). IPFP-ASCs are attracting more and more attention due to their characteristics suitable to regenerative medicine such as strong proliferation and differentiation, anti-inflammation, antiaging, secreting cytokines, multipotential capacity, and 3D culture. IPFP-ASCs can repair articular cartilage and relieve the pain caused by osteoarthritis, so most of IPFP-related review articles focus on osteoarthritis. This article reviews the anatomy and function of IPFP, as well as the discovery, amplification, multipotential capacity, and application of IPFP-ASCs in order to explain why IPFP-ASC is a superior stem cell source in regenerative medicine.
Collapse
Affiliation(s)
- Yu-chen Zhong
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
- Class 4, Phase 102, China Medical University, Shenyang 110122, China
| | - Shi-chun Wang
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
- Class 4, Phase 102, China Medical University, Shenyang 110122, China
| | - Yin-he Han
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
| | - Yu Wen
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
| |
Collapse
|
26
|
Human Adipose-Derived Hydrogel Characterization Based on In Vitro ASC Biocompatibility and Differentiation. Stem Cells Int 2019; 2019:9276398. [PMID: 32082388 PMCID: PMC7012213 DOI: 10.1155/2019/9276398] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/12/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022] Open
Abstract
Hydrogels serve as three-dimensional scaffolds whose composition can be customized to allow attachment and proliferation of several different cell types. Extracellular matrix-derived hydrogels are considered close replicates of the tissue microenvironment. They can serve as scaffolds for in vitro tissue engineering and are a useful tool to study cell-scaffold interaction. The aim of the present study was to analyze the effect of adipose-derived stromal/stem cells (ASCs) and decellularized adipose tissue-derived (DAT) hydrogel interaction on ASC morphology, proliferation, differentiation, and DAT hydrogel microstructure. First, the ASCs were characterized using flow cytometry, adipogenic/osteogenic differentiation, colony-forming unit fibroblast assay and doubling time. The viability and proliferation assays showed that ASCs seeded in DAT hydrogel at different concentrations and cultured for 21 days remained viable and displayed proliferation. ASCs were seeded on DAT hydrogel and cultured in stromal, adipogenic, or osteogenic media for 14 or 28 days. The analysis of adipogenic differentiation demonstrated the upregulation of adipogenic marker genes and accumulation of oil droplets in the cells. Osteogenic differentiation demonstrated the upregulation of osteogenic marker genes and mineral deposition in the DAT hydrogel. The analysis of DAT hydrogel fiber metrics revealed that ASC seeding, and differentiation altered both the diameter and arrangement of fibers in the matrix. Matrix metalloproteinase-2 (MMP-2) activity was assessed to determine the possible mechanism for DAT hydrogel remodeling. MMP-2 activity was observed in all ASC seeded samples, with the osteogenic samples displaying the highest MMP-2 activity. These findings indicate that DAT hydrogel is a cytocompatible scaffold that supports the adipogenic and osteogenic differentiation of ASCs. Furthermore, the attachment of ASCs and differentiation along adipogenic and osteogenic lineages remodels the microstructure of DAT hydrogel.
Collapse
|
27
|
Breast-Associated Adipocytes Secretome Induce Fatty Acid Uptake and Invasiveness in Breast Cancer Cells via CD36 Independently of Body Mass Index, Menopausal Status and Mammary Density. Cancers (Basel) 2019; 11:cancers11122012. [PMID: 31847105 PMCID: PMC6966437 DOI: 10.3390/cancers11122012] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/29/2019] [Accepted: 12/10/2019] [Indexed: 12/26/2022] Open
Abstract
Breast adiposity is correlated with body mass index, menopausal status and mammary density. We here wish to establish how these factors influence the cross-talk between breast adipocytes and normal or malignant breast cells. Adipocyte-derived stem cells (ASCs) were obtained from healthy women and classified into six distinct groups based on body mass index, menopausal status and mammary density. The ASCs were induced to differentiate, and the influence of their conditioned media (ACM) was determined. Unexpectedly, there were no detectable differences in adipogenic differentiation and secretion between the six ASC groups, while their corresponding ACMs had no detectable influence on normal breast cells. In clear contrast, all ACMs profoundly influenced the proliferation, migration and invasiveness of malignant breast cells and increased the number of lipid droplets in their cytoplasm via increased expression of the fatty acid receptor CD36, thereby increasing fatty acid uptake. Importantly, inhibition of CD36 reduced lipid droplet accumulation and attenuated the migration and invasion of the breast cancer cells. These findings suggest that breast-associated adipocytes potentiate the invasiveness of breast cancer cells which, at least in part, is mediated by metabolic reprogramming via CD36-mediated fatty acid uptake.
Collapse
|
28
|
Yin GN, Wang L, Lin XN, Shi L, Gao ZL, Han FC, Li P, Jin YC, Suh JK, Ryu JK, Wang X, Jin HR. Combination of stromal vascular fraction and Ad-COMP-Ang1 gene therapy improves long-term therapeutic efficacy for diabetes-induced erectile dysfunction. Asian J Androl 2019; 20:465-472. [PMID: 29667617 PMCID: PMC6116680 DOI: 10.4103/aja.aja_16_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Men with diabetic erectile dysfunction (ED) respond poorly to the currently available oral phosphodiesterase-5 inhibitors. Therefore, functional therapies for diabetic ED are needed. Stromal vascular fraction (SVF) and the adenovirus-mediated cartilage oligomeric matrix angiopoietin-1 (Ad-COMP-Ang1) gene are known to play critical roles in penile erection. We previously reported that SVF and Ad-COMP-Ang1 have only a short-term effect in restoring erectile function. Further improvements to ED therapy are needed for long-lasting effects. In the present study, we aimed to test if the combination of SVF and Ad-COMP-Ang1 could extend the erection effect in diabetic ED. We found that the combination therapy showed a long-term effect in restoring erectile function through enhanced penile endothelial and neural cell regeneration. Combination therapy with SVF and Ad-COMP-Ang1 notably restored cavernous endothelial cell numbers, pericyte numbers, endothelial cell–cell junctions, decreased cavernous endothelial cell permeability, and promoted neural regeneration for at least 4 weeks in diabetic mice. In summary, this is an initial description of the long-term effect of combination therapy with SVF and Ad-COMP-Ang1 in restoring erectile function through a dual effect on endothelial and neural cell regeneration. Such combination therapy may have therapeutic potential for the treatment of diabetic ED.
Collapse
Affiliation(s)
- Guo-Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
| | - Lin Wang
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai 264000, China
| | - Xiang-Nan Lin
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai 264000, China
| | - Lei Shi
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai 264000, China
| | - Zhen-Lin Gao
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai 264000, China
| | - Feng-Chan Han
- Key Laboratory for Genetic Hearing Disorders in Shandong, Binzhou Medical University, Yantai 264003, China
| | - Ping Li
- Key Laboratory for Genetic Hearing Disorders in Shandong, Binzhou Medical University, Yantai 264003, China
| | - Yin-Chuan Jin
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong 264000, China
| | - Jun-Kyu Suh
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
| | - Xiong Wang
- Reproductive Medicine Center, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China
| | - Hai-Rong Jin
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai 264000, China
| |
Collapse
|
29
|
Annamalai RT, Hong X, Schott NG, Tiruchinapally G, Levi B, Stegemann JP. Injectable osteogenic microtissues containing mesenchymal stromal cells conformally fill and repair critical-size defects. Biomaterials 2019; 208:32-44. [PMID: 30991216 PMCID: PMC6500486 DOI: 10.1016/j.biomaterials.2019.04.001] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/28/2019] [Accepted: 04/01/2019] [Indexed: 12/18/2022]
Abstract
Repair of complex fractures with bone loss requires a potent, space-filling intervention to promote regeneration of bone. We present a biomaterials-based strategy combining mesenchymal stromal cells (MSC) with a chitosan-collagen matrix to form modular microtissues designed for delivery through a needle to conformally fill cavital defects. Implantation of microtissues into a calvarial defect in the mouse showed that osteogenically pre-differentiated MSC resulted in complete bridging of the cavity, while undifferentiated MSC produced mineralized tissue only in apposition to native bone. Decreasing the implant volume reduced bone regeneration, while increasing the MSC concentration also attenuated bone formation, suggesting that the cell-matrix ratio is important in achieving a robust response. Conformal filling of the defect with microtissues in a carrier gel resulted in complete healing. Taken together, these results show that modular microtissues can be used to augment the differentiated function of MSC and provide an extracellular environment that potentiates bone repair.
Collapse
Affiliation(s)
- Ramkumar T Annamalai
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, United States
| | - Xiaowei Hong
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, United States
| | - Nicholas G Schott
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, United States
| | | | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, United States
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, United States.
| |
Collapse
|
30
|
Polly SS, Nichols AEC, Donnini E, Inman DJ, Scott TJ, Apple SM, Werre SR, Dahlgren LA. Adipose-Derived Stromal Vascular Fraction and Cultured Stromal Cells as Trophic Mediators for Tendon Healing. J Orthop Res 2019; 37:1429-1439. [PMID: 30977556 DOI: 10.1002/jor.24307] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 03/18/2019] [Indexed: 02/04/2023]
Abstract
Adipose-derived stromal vascular fraction (SVF) is a heterogeneous population of cells that yields a homogeneous population of plastic-adherent adipose tissue-derived stromal cells (ASC) when culture-expanded. SVF and ASC have been used clinically to improve tendon healing, yet their mechanism of action is not fully elucidated. The objective of this study was to investigate the potential for ASC to act as trophic mediators for tendon healing. Flexor digitorum superficialis tendons and adipose tissue were harvested from adult horses to obtain SVF, ASC, and tenocytes. Growth factor gene expression was quantified in SVF and ASC in serial passages and growth factors were quantified in ASC-conditioned medium (CM). Microchemotaxis assays were performed using ASC-CM. Tenocytes were grown in co-culture with autologous ASC or allogeneic SVF. Gene expression for insulin-like growth factor 1 (IGF-1), stromal cell-derived factor-1α (SDF-1α), transforming growth factor-β1 (TGF-β1) and TGF-β3 was significantly higher in SVF compared to ASC. Concentrations were significantly increased in ASC-CM compared to controls for IGF-1 (4-fold) and SDF-1α (6-fold). Medium conditioned by ASC induced significant cell migration in a dose-dependent manner. Gene expression for collagen types I and III, decorin, and cartilage oligomeric matrix protein was modestly, but significantly increased following co-culture of tenocytes with autologous ASC. Our findings support the ability of SVF and ASC to act as trophic mediators in tendon healing, particularly through chemotaxis, which stands to critically impact the intrinsic healing response. In vivo studies to further delineate the potential for SVF and/or ASC to improve tendon healing are warranted. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1429-1439, 2019.
Collapse
Affiliation(s)
- Shelley S Polly
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Anne E C Nichols
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Elle Donnini
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Daniel J Inman
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Timothy J Scott
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Stephanie M Apple
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Stephen R Werre
- Laboratory for Statistical Design and Study Analysis, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Linda A Dahlgren
- Department of Large Animal Clinical Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| |
Collapse
|
31
|
Adipose-Derived Tissue in the Treatment of Dermal Fibrosis: Antifibrotic Effects of Adipose-Derived Stem Cells. Ann Plast Surg 2019; 80:297-307. [PMID: 29309331 DOI: 10.1097/sap.0000000000001278] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Treatment of hypertrophic scars and other fibrotic skin conditions with autologous fat injections shows promising clinical results; however, the underlying mechanisms of its antifibrotic action have not been comprehensively studied. Adipose-derived stem cells, or stromal cell-derived factors, inherent components of the transplanted fat tissue, seem to be responsible for its therapeutic effects on difficult scars. The mechanisms by which this therapeutic effect takes place are diverse and are mostly mediated by paracrine signaling, which switches on various antifibrotic molecular pathways, modulates the activity of the central profibrotic transforming growth factor β/Smad pathway, and normalizes functioning of fibroblasts and keratinocytes in the recipient site. Direct cell-to-cell communications and differentiation of cell types may also play a positive role in scar treatment, even though they have not been extensively studied in this context. A more thorough understanding of the fat tissue antifibrotic mechanisms of action will turn this treatment from an anecdotal remedy to a more controlled, timely administered technology.
Collapse
|
32
|
Najar M, Lombard CA, Fayyad-Kazan H, Faour WH, Merimi M, Sokal EM, Lagneaux L, Fahmi H. Th17 immune response to adipose tissue-derived mesenchymal stromal cells. J Cell Physiol 2019; 234:21145-21152. [PMID: 31041809 DOI: 10.1002/jcp.28717] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 12/11/2022]
Abstract
Adipose tissue-derived mesenchymal stromal cells (ASCs) hold the promise of achieving successful immunotherapeutic results due to their ability to regulate different T-cell fate. ASCs also show significant adaptability to environmental stresses by modulating their immunologic profile. Cell-based therapy for inflammatory diseases requires a detailed understanding of the molecular relation between ASCs and Th17 lymphocytes taking into account the influence of inflammation and cell ratio on such interaction. Accordingly, a dose-dependent increase in Th17 generation was only observed in high MSC:T-cell ratio with no significant impact of inflammatory priming. IL-23 receptor (IL-23R) expression by T cells was not modulated by ASCs when compared to levels in activated T cells, while ROR-γt expression was significantly increased reaching a maximum in high (1:5) unprimed ASC:T-cell ratio. Finally, multiplex immunoassay showed substantial changes in the secretory profile of 15 cytokines involved in the Th17 immune response (IL-1β, IL-4, IL-6, IL-10, IL-17A, IL-17F, IL-22, IL-21, IL-23, IL-25, IL-31, IL-33, IFN-γ, sCD40, and TNF-α), which was modulated by both cell ratio and inflammatory priming. These findings suggest that Th17 lymphocyte pathway is significantly modulated by ASCs that may lead to immunological changes. Therefore, future ASC-based immunotherapy should take into account the complex and detailed molecular interactions that depend on several factors including inflammatory priming and cell ratio.
Collapse
Affiliation(s)
- Mehdi Najar
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), and Department of Medicine, Montreal, Quebec, Canada.,Laboratory of Physiology, Ethnopharmacology and Genetics, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Catherine A Lombard
- Institut de Recherche Expérimentale and Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Université Catholique de Louvain, Brussels, Belgium
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
| | - Wissam H Faour
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Makram Merimi
- Laboratory of Physiology, Ethnopharmacology and Genetics, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco.,Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Etienne M Sokal
- Institut de Recherche Expérimentale and Clinique (IREC), Laboratory of Pediatric Hepatology and Cell Therapy, Université Catholique de Louvain, Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Brussels, Belgium
| | - Hassan Fahmi
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), and Department of Medicine, Montreal, Quebec, Canada
| |
Collapse
|
33
|
Keeley TP, Mann GE. Defining Physiological Normoxia for Improved Translation of Cell Physiology to Animal Models and Humans. Physiol Rev 2019; 99:161-234. [PMID: 30354965 DOI: 10.1152/physrev.00041.2017] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The extensive oxygen gradient between the air we breathe (Po2 ~21 kPa) and its ultimate distribution within mitochondria (as low as ~0.5-1 kPa) is testament to the efforts expended in limiting its inherent toxicity. It has long been recognized that cell culture undertaken under room air conditions falls short of replicating this protection in vitro. Despite this, difficulty in accurately determining the appropriate O2 levels in which to culture cells, coupled with a lack of the technology to replicate and maintain a physiological O2 environment in vitro, has hindered addressing this issue thus far. In this review, we aim to address the current understanding of tissue Po2 distribution in vivo and summarize the attempts made to replicate these conditions in vitro. The state-of-the-art techniques employed to accurately determine O2 levels, as well as the issues associated with reproducing physiological O2 levels in vitro, are also critically reviewed. We aim to provide the framework for researchers to undertake cell culture under O2 levels relevant to specific tissues and organs. We envisage that this review will facilitate a paradigm shift, enabling translation of findings under physiological conditions in vitro to disease pathology and the design of novel therapeutics.
Collapse
Affiliation(s)
- Thomas P Keeley
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London , London , United Kingdom
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London , London , United Kingdom
| |
Collapse
|
34
|
Smieszek A, Kornicka K, Szłapka-Kosarzewska J, Androvic P, Valihrach L, Langerova L, Rohlova E, Kubista M, Marycz K. Metformin Increases Proliferative Activity and Viability of Multipotent Stromal Stem Cells Isolated from Adipose Tissue Derived from Horses with Equine Metabolic Syndrome. Cells 2019; 8:E80. [PMID: 30678275 PMCID: PMC6406832 DOI: 10.3390/cells8020080] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/18/2019] [Accepted: 01/21/2019] [Indexed: 12/17/2022] Open
Abstract
In this study, we investigated the influence of metformin (MF) on proliferation and viability of adipose-derived stromal cells isolated from horses (EqASCs). We determined the effect of metformin on cell metabolism in terms of mitochondrial metabolism and oxidative status. Our purpose was to evaluate the metformin effect on cells derived from healthy horses (EqASCHE) and individuals affected by equine metabolic syndrome (EqASCEMS). The cells were treated with 0.5 μM MF for 72 h. The proliferative activity was evaluated based on the measurement of BrdU incorporation during DNA synthesis, as well as population doubling time rate (PDT) and distribution of EqASCs in the cell cycle. The influence of metformin on EqASC viability was determined in relation to apoptosis profile, mitochondrial membrane potential, oxidative stress markers and BAX/BCL-2 mRNA ratio. Further, we were interested in possibility of metformin affecting the Wnt3a signalling pathway and, thus, we determined mRNA and protein level of WNT3A and β-catenin. Finally, using a two-tailed RT-qPCR method, we investigated the expression of miR-16-5p, miR-21-5p, miR-29a-3p, miR-140-3p and miR-145-5p. Obtained results indicate pro-proliferative and anti-apoptotic effects of metformin on EqASCs. In this study, MF significantly improved proliferation of EqASCs, which manifested in increased synthesis of DNA and lowered PDT value. Additionally, metformin improved metabolism and viability of cells, which correlated with higher mitochondrial membrane potential, reduced apoptosis and increased WNT3A/β-catenin expression. Metformin modulates the miRNA expression differently in EqASCHE and EqASCEMS. Metformin may be used as a preconditioning agent which stimulates proliferative activity and viability of EqASCs.
Collapse
Affiliation(s)
- Agnieszka Smieszek
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
| | - Katarzyna Kornicka
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
| | - Jolanta Szłapka-Kosarzewska
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
| | - Peter Androvic
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Biocev, 252 50 Vestec, Czech Republic.
- Laboratory of Growth Regulators, Faculty of Science, Palacky University, 78371 Olomouc, Czech Republic.
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Biocev, 252 50 Vestec, Czech Republic.
| | - Lucie Langerova
- Gene Core BIOCEV, Průmyslová 595, Vestec 252 50, Czech Republic.
| | - Eva Rohlova
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Biocev, 252 50 Vestec, Czech Republic.
- Department of Anthropology and Human Genetics, Faculty of Science, Charles University, 128 43 Prague, Czech Republic.
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology CAS, Biocev, 252 50 Vestec, Czech Republic.
- TATAA Biocenter AB, 411 03 Gothenburg, Sweden.
| | - Krzysztof Marycz
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences, 50-375 Wroclaw, Poland.
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392 Giessen, Germany.
| |
Collapse
|
35
|
In Vitro and In Vivo Osteogenic Differentiation of Human Adipose-Derived Stromal Cells. Methods Mol Biol 2018. [PMID: 30414122 DOI: 10.1007/978-1-4939-8904-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Adipose-derived stromal cells (ASCs) are a promising population of cells that may be useful for the regeneration of human tissue defects. ASCs are capable of forming bone tissue in vitro and in vivo. Further work is required to determine the optimal conditions that will allow human ASCs to regenerate tissue in clinically significant tissue defects. Here we present three experimental protocols that are indispensable for the study of ASC osteogenic activity.
Collapse
|
36
|
Hillers LE, D'Amato JV, Chamberlin T, Paderta G, Arendt LM. Obesity-Activated Adipose-Derived Stromal Cells Promote Breast Cancer Growth and Invasion. Neoplasia 2018; 20:1161-1174. [PMID: 30317122 PMCID: PMC6187054 DOI: 10.1016/j.neo.2018.09.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 09/09/2018] [Accepted: 09/16/2018] [Indexed: 01/07/2023] Open
Abstract
Obese women diagnosed with breast cancer have an increased risk for metastasis, and the underlying mechanisms are not well established. Within the mammary gland, adipose-derived stromal cells (ASCs) are heterogeneous cells with the capacity to differentiate into multiple mesenchymal lineages. To study the effects of obesity on ASCs, mice were fed a control diet (CD) or high-fat diet (HFD) to induce obesity, and ASCs were isolated from the mammary glands of lean and obese mice. We observed that obesity increased ASCs proliferation, decreased differentiation potential, and upregulated expression of α-smooth muscle actin, a marker of activated fibroblasts, compared to ASCs from lean mice. To determine how ASCs from obese mice impacted tumor growth, we mixed ASCs isolated from CD- or HFD-fed mice with mammary tumor cells and injected them into the mammary glands of lean mice. Tumor cells mixed with ASCs from obese mice grew significantly larger tumors and had increased invasion into surrounding adipose tissue than tumor cells mixed with control ASCs. ASCs from obese mice demonstrated enhanced tumor cell invasion in culture, a phenotype associated with increased expression of insulin-like growth factor-1 (IGF-1) and abrogated by IGF-1 neutralizing antibodies. Weight loss induced in obese mice significantly decreased expression of IGF-1 from ASCs and reduced the ability of the ASCs to induce an invasive phenotype. Together, these results suggest that obesity enhances local invasion of breast cancer cells through increased expression of IGF-1 by mammary ASCs, and weight loss may reverse this tumor-promoting phenotype.
Collapse
Affiliation(s)
- Lauren E Hillers
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI 53706
| | - Joseph V D'Amato
- Department of Comparative Biosciences, School of Veterinary Medicine, University Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706
| | - Tamara Chamberlin
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI 53706
| | - Gretchen Paderta
- Department of Comparative Biosciences, School of Veterinary Medicine, University Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706
| | - Lisa M Arendt
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI 53706; Department of Comparative Biosciences, School of Veterinary Medicine, University Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706.
| |
Collapse
|
37
|
Baxter-Holland M, Dass CR. Pigment epithelium-derived factor: a key mediator in bone homeostasis and potential for bone regenerative therapy. J Pharm Pharmacol 2018; 70:1127-1138. [DOI: 10.1111/jphp.12942] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 05/19/2018] [Indexed: 01/02/2023]
Abstract
Abstract
Objectives
Pigment epithelium-derived factor (PEDF), a multifunctional endogenous glycoprotein, has a very wide range of biological actions, notably in bone homeostasis. The question has been raised regarding the place of PEDF in the treatment of bone disorders and osteosarcoma, and its potential for tumour growth suppression.
Methods
The PubMed database was used to compile this review.
Key findings
Pigment epithelium-derived factor's actions in osteoid tissues include promoting mesenchymal stem cell commitment to osteoblasts, increasing matrix mineralisation, and promoting osteoblast proliferation. It shows potential to improve therapeutic outcomes in treatment of multiple cancer types and regrowth of bone after trauma or resection in animal studies. PEDF may possibly have a reduced adverse effect profile compared with current osteo-regenerative treatments; however, there is currently very limited evidence regarding the safety or efficacy in human models.
Summary
Pigment epithelium-derived factor is very active within the body, particularly in osseous tissue, and its physiological actions give it potential for treatment of both bone disorders and multiple tumour types. Further research is needed to ascertain the adverse effects and safety profile of PEDF as a therapeutic agent.
Collapse
Affiliation(s)
- Mia Baxter-Holland
- School of Pharmacy and Biomedical Science, Curtin University, Perth, WA, Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Science, Curtin University, Perth, WA, Australia
- Curtin Health Innovation Research Institute, Bentley, WA, Australia
| |
Collapse
|
38
|
Haahr MK, Harken Jensen C, Toyserkani NM, Andersen DC, Damkier P, Sørensen JA, Sheikh SP, Lund L. A 12-Month Follow-up After a Single Intracavernous Injection of Autologous Adipose-Derived Regenerative Cells in Patients with Erectile Dysfunction Following Radical Prostatectomy: An Open-Label Phase I Clinical Trial. Urology 2018; 121:203.e6-203.e13. [PMID: 29958973 DOI: 10.1016/j.urology.2018.06.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 05/30/2018] [Accepted: 06/11/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To explore safety in adipose-derived regenerative cells (ADRC) therapy, treating erectile dysfunction (ED). METHODS Twenty-one patients with ED after radical prostatectomy, with no signs of recovery using conventional therapy, received a single intracavernous injection of autologous ADRC and were followed for 1 year. Six men were incontinent, and 15 were continent at inclusion. The primary (safety of ADRC therapy) and secondary endpoints (sexual function) were evaluated at 1, 3, 6, and 12 months after ADRC injection by registration of adverse events and validated questionnaires using the international index of erectile function-5 and erection hardness score. RESULTS No serious adverse events occurred, but 8 reversible minor events related to the liposuction were noted. Eight out of 15 (53%) patients in the continent group reported erectile function sufficient for intercourse at 12 months. Baseline median international index of erectile function-5 scores (6.0; interquartile range [IQR] 3) were unchanged 1 month after the treatment, but significantly increased after 6 to 7 (IQR 17). This effect was sustained at 12 months (median 8; IQR 14). We did not see any improvements in erectile function in the group of incontinent men or among men with ED prior to radical prostatectomy. CONCLUSION Intracavernous injection of ADRC is safe in this phase 1 study with a 12-month follow-up.
Collapse
Affiliation(s)
- Martha Kirstine Haahr
- Department of Urology, Odense University Hospital, Odense, Denmark; The Danish Centre for Regenerative Medicine (www.danishcrm.com), Odense University Hospital, Odense, Denmark; Clinical Institute, University of Southern Denmark, Odense, Denmark.
| | - Charlotte Harken Jensen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Navid Mohamadpour Toyserkani
- Department of Plastic Surgery, Odense University Hospital, Odense, Denmark; The Danish Centre for Regenerative Medicine (www.danishcrm.com), Odense University Hospital, Odense, Denmark
| | - Ditte Caroline Andersen
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark; The Danish Centre for Regenerative Medicine (www.danishcrm.com), Odense University Hospital, Odense, Denmark
| | - Per Damkier
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Jens Ahm Sørensen
- Department of Plastic Surgery, Odense University Hospital, Odense, Denmark; The Danish Centre for Regenerative Medicine (www.danishcrm.com), Odense University Hospital, Odense, Denmark
| | - Søren Paludan Sheikh
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark; Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark; The Danish Centre for Regenerative Medicine (www.danishcrm.com), Odense University Hospital, Odense, Denmark
| | - Lars Lund
- Department of Urology, Odense University Hospital, Odense, Denmark; The Danish Centre for Regenerative Medicine (www.danishcrm.com), Odense University Hospital, Odense, Denmark; Clinical Institute, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
39
|
Comparative Analysis between the In Vivo Biodistribution and Therapeutic Efficacy of Adipose-Derived Mesenchymal Stromal Cells Administered Intraperitoneally in Experimental Colitis. Int J Mol Sci 2018; 19:ijms19071853. [PMID: 29937494 PMCID: PMC6073850 DOI: 10.3390/ijms19071853] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 06/15/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as a promising treatment for inflammatory diseases. The immunomodulatory effect of MSCs takes place both by direct cell-to-cell contact and by means of soluble factors that leads to an increased accumulation of regulatory immune cells at the sites of inflammation. Similar efficacy of MSCs has been described regardless of the route of administration used, the inflammation conditions and the major histocompatibility complex context. These observations raise the question of whether the migration of the MSCs to the inflamed tissues is a pre-requisite to achieve their beneficial effect. To address this, we examined the biodistribution and the efficacy of intraperitoneal luciferase-expressing human expanded adipose-derived stem cells (Luci-eASCs) in a mouse model of colitis. Luci-eASC-infused mice were stratified according to their response to the Luci-eASC treatment. According to the stratification criteria, there was a tendency to increase the bioluminescence signal in the intestine at the expense of a decrease in the bioluminescence signal in the liver in the “responder” mice. These data thus suggest that the accumulation of the eASCs to the inflamed tissues is beneficial for achieving an optimal modulation of inflammation.
Collapse
|
40
|
Annamalai RT, Turner PA, Carson WF, Levi B, Kunkel S, Stegemann JP. Harnessing macrophage-mediated degradation of gelatin microspheres for spatiotemporal control of BMP2 release. Biomaterials 2018; 161:216-227. [PMID: 29421557 PMCID: PMC5831261 DOI: 10.1016/j.biomaterials.2018.01.040] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/24/2018] [Accepted: 01/25/2018] [Indexed: 02/07/2023]
Abstract
Biomaterials-based approaches to harnessing the immune and inflammatory responses to potentiate wound healing hold important promise. Bone fracture healing is characterized by an acute inflammatory phase, followed by a transition to a regenerative and repair phase. In this study, we developed genipin-crosslinked gelatin microspheres designed to be preferentially degraded by inflammatory (M1) macrophages. Highly crosslinked (>90%) microspheres allowed efficient incorporation of bioactive bone morphogenetic protein 2 (BMP2), a potent stimulator of osteogenesis in progenitor cells, via electrostatic interactions. Release of BMP2 was directly correlated with degradation of the gelatin matrix. Exposure of microspheres to polarized murine macrophages showed that degradation was significantly higher in the presence of M1 macrophages, relative to alternatively activated (M2) macrophages and unpolarized controls. Microsphere degradation in the presence of non-inflammatory cells resulted in very low degradation rates. The expression of matrix metalloproteinases (MMPs) and tissue inhibitors of MMP (TIMPs) by macrophages were consistent with the observed phenotype-dependent degradation rates. Indirect co-culture of BMP2-loaded microspheres and macrophages with isolated adipose-derived mesenchymal stem cells (MSC) showed that M1 macrophages produced the strongest osteogenic response, comparable to direct supplementation of the culture medium with BMP2. Controlled release systems that are synchronized with the inflammatory response have the potential to provide better spatiotemporal control of growth factor delivery and therefore may improve the outcomes of recalcitrant wounds.
Collapse
Affiliation(s)
| | - Paul A Turner
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA
| | | | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, USA
| | - Steven Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, USA
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA.
| |
Collapse
|
41
|
Gaubys A, Papeckys V, Pranskunas M. Use of Autologous Stem Cells for the Regeneration of Periodontal Defects in Animal Studies: a Systematic Review and Meta-Analysis. J Oral Maxillofac Res 2018; 9:e3. [PMID: 30116515 PMCID: PMC6090251 DOI: 10.5037/jomr.2018.9203] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 05/24/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To overview preclinical animal trials and quantify the effect size that stem cell therapy has on the regeneration of periodontal tissue complex. MATERIAL AND METHODS A systematic MEDLINE (PubMed) online library search was conducted for preclinical animal studies in vivo , using autologous periodontal ligament, dental pulp, cementum, alveolar periosteal, gingival margin or adipose stem cell types for periodontal tissue complex regeneration purposes. Studies had to be published between 2007.09.01 and 2017.09.01 in the English language. RESULTS Online library search yielded 2099 results. After the title, abstract and full-text screening ten studies fit inclusion criteria and were pooled into meta-analysis. Overall the stem cell regenerative therapy had a statistically significant positive influence on the periodontal tissue regeneration when compared to the control groups. The biggest influence was made to the regeneration of cementum (standardised mean difference [SMD] 2.25 [95% confidence interval (CI) = 1.31 to 3.2]) while the smallest influence was made to the alveolar bone (SMD 1.47 [95% CI = 0.7 to 2.25]) the effect size for periodontal ligament regeneration was (SMD 1.8 [95% CI = 1 to 2.59]). Subgroup analysis showed statistically significant (P < 0.05) differences between different cell types in the alveolar bone and cementum regeneration groups and in alveolar bone group in relation to scaffold materials. CONCLUSIONS Stem cell therapy has a positive impact on periodontal tissue complex regeneration. Such therapy has the biggest influence on cementum regeneration meanwhile alveolar bone regeneration is influenced by the least amount. However more and less diverse preclinical studies are needed to have a greater statistical power in future meta-analyses.
Collapse
Affiliation(s)
- Algimantas Gaubys
- Department of Maxillofacial Surgery, Lithuanian University of Health Sciences, KaunasLithuania.
| | - Valdas Papeckys
- Department of Maxillofacial Surgery, Lithuanian University of Health Sciences, KaunasLithuania.
| | - Mindaugas Pranskunas
- Department of Maxillofacial Surgery, Lithuanian University of Health Sciences, KaunasLithuania.
| |
Collapse
|
42
|
Hoffman JM, Sideri A, Ruiz JJ, Stavrakis D, Shih DQ, Turner JR, Pothoulakis C, Karagiannides I. Mesenteric Adipose-derived Stromal Cells From Crohn's Disease Patients Induce Protective Effects in Colonic Epithelial Cells and Mice With Colitis. Cell Mol Gastroenterol Hepatol 2018; 6:1-16. [PMID: 29928668 PMCID: PMC6008259 DOI: 10.1016/j.jcmgh.2018.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/01/2018] [Indexed: 12/14/2022]
Abstract
Mesenteric adipose tissue hyperplasia is a hallmark of Crohn's disease (CD). Recently, we showed that mesenteric adipose-derived stromal cells (ADSCs) from CD, ulcerative colitis, and control patients synthesize and release adipokines in a disease-dependent manner. Here we examined the expression profiles of CD and control patient-derived mesenteric ADSCs and studied the effects of their extracellular mediators on colonocyte signaling in vitro and experimental colitis in vivo. ADSCs were isolated from mesenteric fat of control and CD patients. Microarray profiling and network analysis were performed in ADSCs and human colonocytes treated with conditioned media from cultured ADSCs. Mice with acute colitis received daily injections of conditioned media from patient-derived ADSCs, vehicle, or apolactoferrin. Proliferative responses were evaluated in conditioned media-treated colonocytes and mouse colonic epithelium. Total protein was isolated from cultured colonocytes after treatment with apolactoferrin for Western blot analysis of phosphorylated intracellular signaling kinases. Microarray profiling revealed differential mRNA expression in CD patient-derived ADSCs compared with controls, including lactoferrin. Administration of CD patient-derived medium or apolactoferrin increased colonocyte proliferation compared with controls. Conditioned media from CD patient-derived ADSCs or apolactoferrin attenuated colitis severity in mice and enhanced colonocyte proliferation in vivo. ADSCs from control and CD patients show disease-dependent inflammatory responses and alter colonic epithelial cell signaling in vitro and in vivo. Furthermore, we demonstrate lactoferrin production by adipose tissue, specifically mesenteric ADSCs. We suggest that mesenteric ADSC-derived lactoferrin may mediate protective effects and participate in the pathophysiology of CD by promoting colonocyte proliferation and the resolution of inflammation.
Collapse
Key Words
- ADSC, adipose-derived stromal cell
- CD, Crohn’s disease
- DSS, dextran sodium sulfate
- IBD, inflammatory bowel disease
- IBS, irritable bowel syndrome
- IL, interleukin
- Inflammatory Bowel Disease
- Intestinal Epithelium
- Mesenteric Adipose Tissue
- PCR, polymerase chain reaction
- Preadipocytes
- RT, reverse-transcriptase
- TNBS, trinitrobenzenesulfonic acid
- VEGF, vascular endothelial growth factor
- i.c., intracolonic
Collapse
Affiliation(s)
- Jill M. Hoffman
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California,Jill Hoffman, PhD, Inflammatory Bowel Disease Center, Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, 675 Charles E. Young Drive South, MRL Building 1220, Los Angeles, California 90095. fax: (310) 825-3542
| | - Aristea Sideri
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Jonathan J. Ruiz
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Dimitris Stavrakis
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - David Q. Shih
- Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, California
| | - Jerrold R. Turner
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Charalabos Pothoulakis
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Iordanes Karagiannides
- Center for Inflammatory Bowel Diseases, Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California,Correspondence Address correspondence to: Iordanes Karagiannides, PhD, Inflammatory Bowel Disease Center, Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, 675 Charles E. Young Drive South, MRL Building 1220, Los Angeles, California 90095. fax: (310) 825-3542.
| |
Collapse
|
43
|
Sun X, Gupta K, Wu B, Zhang D, Yuan B, Zhang X, Chiang HC, Zhang C, Curiel TJ, Bendeck MP, Hursting S, Hu Y, Li R. Tumor-extrinsic discoidin domain receptor 1 promotes mammary tumor growth by regulating adipose stromal interleukin 6 production in mice. J Biol Chem 2018; 293:2841-2849. [PMID: 29298894 DOI: 10.1074/jbc.ra117.000672] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/31/2017] [Indexed: 12/25/2022] Open
Abstract
Discoidin domain receptor 1 (DDR1) is a collagen receptor that mediates cell communication with the extracellular matrix (ECM). Aberrant expression and activity of DDR1 in tumor cells are known to promote tumor growth. Although elevated DDR1 levels in the stroma of breast tumors are associated with poor patient outcome, a causal role for tumor-extrinsic DDR1 in cancer promotion remains unclear. Here we report that murine mammary tumor cells transplanted to syngeneic recipient mice in which Ddr1 has been knocked out (KO) grow less robustly than in WT mice. We also found that the tumor-associated stroma in Ddr1-KO mice exhibits reduced collagen deposition compared with the WT controls, supporting a role for stromal DDR1 in ECM remodeling of the tumor microenvironment. Furthermore, the stromal-vascular fraction (SVF) of Ddr1 knockout adipose tissue, which contains committed adipose stem/progenitor cells and preadipocytes, was impaired in its ability to stimulate tumor cell migration and invasion. Cytokine array-based screening identified interleukin 6 (IL-6) as a cytokine secreted by the SVF in a DDR1-dependent manner. SVF-produced IL-6 is important for SVF-stimulated tumor cell invasion in vitro, and, using antibody-based neutralization, we show that tumor promotion by IL-6 in vivo requires DDR1. In conclusion, our work demonstrates a previously unrecognized function of DDR1 in promoting tumor growth.
Collapse
Affiliation(s)
- Xiujie Sun
- Department of Molecular Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Kshama Gupta
- Department of Molecular Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Bogang Wu
- Department of Molecular Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Deyi Zhang
- Department of Molecular Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Bin Yuan
- Department of Molecular Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Xiaowen Zhang
- Department of Molecular Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Huai-Chin Chiang
- Department of Molecular Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Chi Zhang
- Department of Molecular Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Tyler J Curiel
- Department of Molecular Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229
| | - Michelle P Bendeck
- Ted Rogers Center for Heart Research, University of Toronto, Toronto, Ontario M5G 1M1, Canada
| | - Stephen Hursting
- Department of Nutrition, Nutrition Research Institute, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Yanfen Hu
- Department of Molecular Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229.
| | - Rong Li
- Department of Molecular Medicine, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas 78229.
| |
Collapse
|
44
|
Foster AD, Clark N, Davis TA. Induction of Skin Allograft Transplantation Tolerance in Mice Using Human Adipose Derived Stromal Cells. Methods Mol Biol 2018; 1773:73-91. [PMID: 29687382 DOI: 10.1007/978-1-4939-7799-4_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Murine models of allograft transplantation are valuable for understanding the immunological mechanisms of allograft acceptance and rejection, the evaluation of immunosuppressive drugs and strategies, and the restoration of functional defects. Herein, we describe methods to create a skin murine allograft surgical model and how to administer adipose-derived stromal cells (ASC) with limited numbers of donor bone marrow to create stable multilineage donor cell chimerism and indefinite immunological tolerance.
Collapse
Affiliation(s)
- Anthony D Foster
- Regenerative Medicine Department, Naval Medical Research Center, Silver Spring, MD, USA
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Nicholas Clark
- Regenerative Medicine Department, Naval Medical Research Center, Silver Spring, MD, USA
| | - Thomas A Davis
- Regenerative Medicine Department, Naval Medical Research Center, Silver Spring, MD, USA.
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
45
|
Zanata F, Shaik S, Devireddy RV, Wu X, Ferreira LM, Gimble JM. Cryopreserved Adipose Tissue-Derived Stromal/Stem Cells: Potential for Applications in Clinic and Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 951:137-146. [PMID: 27837560 DOI: 10.1007/978-3-319-45457-3_11] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adipose-Derived Stromal/Stem Cells (ASC) have considerable potential for regenerative medicine due to their abilities to proliferate, differentiate into multiple cell lineages, high cell yield, relative ease of acquisition, and almost no ethical concerns since they are derived from adult tissue. Storage of ASC by cryopreservation has been well described that maintains high cell yield and viability, stable immunophenotype, and robust differentiation potential post-thaw. This ability is crucial for banking research and for clinical therapeutic purposes that avoid the morbidity related to repetitive liposuction tissue harvests. ASC secrete various biomolecules such as cytokines which are reported to have immunomodulatory properties and therapeutic potential to reverse symptoms of multiple degenerative diseases/disorders. Nevertheless, safety regarding the use of these cells clinically is still under investigation. This chapter focuses on the different aspects of cryopreserved ASC and the methods to evaluate their functionality for future clinical use.
Collapse
Affiliation(s)
- Fabiana Zanata
- Federal University of Sao Paulo, Sao Paulo, SP, Brazil
- Center for Stem Cell Research & Regenerative Medicine, Tulane University, New Orleans, LA, USA
| | - Shahensha Shaik
- Department of Mechanical Engineering, Louisiana State University, Baton Rouge, LA, USA
| | - Ram V Devireddy
- Department of Mechanical Engineering, Louisiana State University, Baton Rouge, LA, USA
| | - Xiying Wu
- La Cell LLC, New Orleans BioInnovation Center, Suite 304, 1441 Canal Street, New Orleans, LA, 70112, USA
| | | | - Jeffrey M Gimble
- Center for Stem Cell Research & Regenerative Medicine, Tulane University, New Orleans, LA, USA.
- La Cell LLC, New Orleans BioInnovation Center, Suite 304, 1441 Canal Street, New Orleans, LA, 70112, USA.
| |
Collapse
|
46
|
Turner L. ClinicalTrials.gov, stem cells and ‘pay-to-participate’ clinical studies. Regen Med 2017; 12:705-719. [DOI: 10.2217/rme-2017-0015] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Numerous US businesses that engage in direct-to-consumer advertising of stem cell interventions that are not US FDA-approved also recruit clients by listing ‘pay-to-participate’ studies listed on ClinicalTrials.gov . Individuals considering enrolling in such studies and NIH officials responsible for overseeing the database need to be aware that some businesses are using the registry to promote unapproved stem cell interventions that study subjects are charged to receive. Inclusion of such studies in ClinicalTrials.gov reveals that the database needs better screening tools. In particular, screening should evaluate whether studies submitted to the registry have been reviewed and permitted to proceed by the FDA in the case of clinical studies requiring FDA clearance in addition to institutional review board approval.
Collapse
Affiliation(s)
- Leigh Turner
- Center for Bioethics, School of Public Health & College of Pharmacy, University of Minnesota, N520 Boynton, 410 Church Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
47
|
Bae CR, Hino J, Hosoda H, Arai Y, Son C, Makino H, Tokudome T, Tomita T, Kimura T, Nojiri T, Hosoda K, Miyazato M, Kangawa K. Overexpression of C-type Natriuretic Peptide in Endothelial Cells Protects against Insulin Resistance and Inflammation during Diet-induced Obesity. Sci Rep 2017; 7:9807. [PMID: 28852070 PMCID: PMC5574992 DOI: 10.1038/s41598-017-10240-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/02/2017] [Indexed: 02/08/2023] Open
Abstract
The endogenous peptide C-type natriuretic peptide (CNP) binds its receptor, guanylyl cyclase B (GCB), and is expressed by endothelial cells in diverse tissues. Because the endothelial cells of visceral adipose tissue have recently been reported to play a role in lipid metabolism and inflammation, we investigated the effects of CNP on features of obesity by using transgenic (Tg) mice in which CNP was placed under the control of the Tie2 promoter and was thus overexpressed in endothelial cells (E-CNP). Here we show that increased brown adipose tissue thermogenesis in E-CNP Tg mice increased energy expenditure, decreased mesenteric white adipose tissue (MesWAT) fat weight and adipocyte hypertrophy, and prevented the development of fatty liver. Furthermore, CNP overexpression improved glucose tolerance, decreased insulin resistance, and inhibited macrophage infiltration in MesWAT, thus suppressing pro-inflammation during high-fat diet–induced obesity. Our findings indicate an important role for the CNP produced by the endothelial cells in the regulation of MesWAT hypertrophy, insulin resistance, and inflammation during high-fat diet–induced obesity.
Collapse
Affiliation(s)
- Cho-Rong Bae
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Jun Hino
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.
| | - Hiroshi Hosoda
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Yuji Arai
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Cheol Son
- Division of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.,Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Hisashi Makino
- Division of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Takeshi Tokudome
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Tsutomu Tomita
- Division of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.,Biobank, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Toru Kimura
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Takashi Nojiri
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Kiminori Hosoda
- Division of Endocrinology and Metabolism, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.
| |
Collapse
|
48
|
The Holy Grail of Orthopedic Surgery: Mesenchymal Stem Cells-Their Current Uses and Potential Applications. Stem Cells Int 2017; 2017:2638305. [PMID: 28698718 PMCID: PMC5494105 DOI: 10.1155/2017/2638305] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/16/2017] [Indexed: 02/07/2023] Open
Abstract
Only select tissues and organs are able to spontaneously regenerate after disease or trauma, and this regenerative capacity diminishes over time. Human stem cell research explores therapeutic regenerative approaches to treat various conditions. Mesenchymal stem cells (MSCs) are derived from adult stem cells; they are multipotent and exert anti-inflammatory and immunomodulatory effects. They can differentiate into multiple cell types of the mesenchyme, for example, endothelial cells, osteoblasts, chondrocytes, fibroblasts, tenocytes, vascular smooth muscle cells, and sarcomere muscular cells. MSCs are easily obtained and can be cultivated and expanded in vitro; thus, they represent a promising and encouraging treatment approach in orthopedic surgery. Here, we review the application of MSCs to various orthopedic conditions, namely, orthopedic trauma; muscle injury; articular cartilage defects and osteoarthritis; meniscal injuries; bone disease; nerve, tendon, and ligament injuries; spinal cord injuries; intervertebral disc problems; pediatrics; and rotator cuff repair. The use of MSCs in orthopedics may transition the practice in the field from predominately surgical replacement and reconstruction to bioregeneration and prevention. However, additional research is necessary to explore the safety and effectiveness of MSC treatment in orthopedics, as well as applications in other medical specialties.
Collapse
|
49
|
Bora P, Majumdar AS. Adipose tissue-derived stromal vascular fraction in regenerative medicine: a brief review on biology and translation. Stem Cell Res Ther 2017; 8:145. [PMID: 28619097 PMCID: PMC5472998 DOI: 10.1186/s13287-017-0598-y] [Citation(s) in RCA: 312] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Adipose/fat tissue provides an abundant source of stromal vascular fraction (SVF) cells for immediate administration and can also give rise to a substantial number of cultured, multipotent adipose-derived stromal cells (ADSCs). Recently, both SVF and ADSCs have gained wide-ranging translational significance in regenerative medicine. Initially used for cosmetic breast enhancement, this mode of treatment has found use in many diseases involving immune disorders, tissue degeneration, and ischaemic conditions. In this review, we try to address several important aspects of this field, outlining the biology, technology, translation, and challenges related to SVF- and ADSC-based therapies. Starting from the basics of SVF and ADSC isolation, we touch upon recently developed technologies, addressing elements of novel methods and devices under development for point-of-care isolation of SVF. Characterisation of SVF cells and ADSCs is also an evolving area and we look into unusual expression of CD34 antigen as an interesting marker for such purposes. Based on reports involving different cells of the SVF, we draw a potential mode of action, focussing on angiogenesis since it involves multiple cells, unlike immunomodulation which is governed predominantly by ADSCs. We have looked into the latest research, experimental therapies, and clinical trials which are utilising SVF/ADSCs in conditions such as multiple sclerosis, Crohn’s disease, peripheral neuropathy, osteoarthritis, diabetic foot ulcer, and so forth. However, problems have arisen with regards to the lack of proper regulatory guidelines for such therapies and, since the introduction of US Food and Drug Administration draft guidelines and the Reliable and Effective Growth for Regenerative Health Options that Improve Wellness (REGROW) Act, the debate became more public with regards to safe and efficacious use of these cells.
Collapse
Affiliation(s)
- Pablo Bora
- Stempeutics Research Private Limited, Akshay Tech Park, # 72&73, 2nd Floor, EPIP Zone, Phase 1, Whitefield, Bangalore, 560066, India.,Present Address: Department of Molecular Biology & Genetics, Faculty of Science, Jihočeská univerzita v Českých Budějovicích (University of South Bohemia), Branišovská 31, 37005, České Budějovice, Czech Republic
| | - Anish S Majumdar
- Stempeutics Research Private Limited, Akshay Tech Park, # 72&73, 2nd Floor, EPIP Zone, Phase 1, Whitefield, Bangalore, 560066, India.
| |
Collapse
|
50
|
Lopez-Santalla M, Mancheño-Corvo P, Escolano A, Menta R, DelaRosa O, Abad JL, Büscher D, Redondo JM, Bueren JA, Dalemans W, Lombardo E, Garin MI. Biodistribution and Efficacy of Human Adipose-Derived Mesenchymal Stem Cells Following Intranodal Administration in Experimental Colitis. Front Immunol 2017. [PMID: 28642759 PMCID: PMC5462906 DOI: 10.3389/fimmu.2017.00638] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have a large potential in cell therapy for treatment of inflammatory and autoimmune diseases, thanks to their immunomodulatory properties. The encouraging results in animal models have initiated the translation of MSC therapy to clinical trials. In cell therapy protocols with MSCs, administered intravenously, several studies have shown that a small proportion of infused MSCs can traffic to the draining lymph nodes (LNs). This is accompanied with an increase of different types of regulatory immune cells in the LNs, suggesting the importance of migration of MSCs to the LNs in order to contribute to immunomodulatory response. Intranodal (IN), also referred as intralymphatic, injection of cells, like dendritic cells, is being proposed in the clinic for the treatment of cancer and allergy, showing that this route of administration is clinically safe and efficient. In this study, we investigated, for the first time, the biodistribution and the efficacy of Luciferase+ adipose-derived MSCs (Luci-eASCs), infused through the inguinal LNs (iLNs), in normal mice and in inflamed mice with colitis. Most of the Luci-eASCs remain in the iLNs and in the adipose tissue surrounding the inguinal LNs. A small proportion of Luci-eASCs can migrate to other locations within the lymphatic system and to other tissues and organs, having a preferential migration toward the intestine in colitic mice. Our results show that the infused Luci-eASCs protected 58% of the mice against induced colitis. Importantly, a correlation between the response to eASC treatment and a higher accumulation of eASCs in popliteal, parathymic, parathyroid, and mesenteric LNs were found. Altogether, these results suggest that IN administration of eASCs is feasible and may represent an effective strategy for cell therapy protocols with human adipose-derived MSCs in the clinic for the treatment of immune-mediated disorders.
Collapse
Affiliation(s)
- Mercedes Lopez-Santalla
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | | | - Amelia Escolano
- Gene Regulation in Cardiovascular Remodeling and Inflammation Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | | | | | | | | | - Juan M Redondo
- Gene Regulation in Cardiovascular Remodeling and Inflammation Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Juan A Bueren
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | | | | | - Marina I Garin
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBER-ER), Madrid, Spain.,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| |
Collapse
|