1
|
Liu M, Lu F, Feng J. Aging and homeostasis of the hypodermis in the age-related deterioration of skin function. Cell Death Dis 2024; 15:443. [PMID: 38914551 PMCID: PMC11196735 DOI: 10.1038/s41419-024-06818-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 02/01/2024] [Accepted: 06/10/2024] [Indexed: 06/26/2024]
Abstract
Adipose tissues in the hypodermis, the crucial stem cell reservoir in the skin and the endocrine organ for the maintenance of skin homeostasis undergo significant changes during skin aging. Dermal white adipose tissue (dWAT) has recently been recognized as an important organ for both non-metabolic and metabolic health in skin regeneration and rejuvenation. Defective differentiation, adipogenesis, improper adipocytokine production, and immunological dissonance dysfunction in dWAT lead to age-associated clinical changes. Here, we review age-related alterations in dWAT across levels, emphasizing the mechanisms underlying the regulation of aging. We also discuss the pathogenic changes involved in age-related fat dysfunction and the unfavorable consequences of accelerated skin aging, such as chronic inflammaging, immunosenescence, delayed wound healing, and fibrosis. Research has shown that adipose aging is an early initiation event and a potential target for extending longevity. We believe that adipose tissues play an essential role in aging and form a potential therapeutic target for the treatment of age-related skin diseases. Further research is needed to improve our understanding of this phenomenon.
Collapse
Affiliation(s)
- Meiqi Liu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Jingwei Feng
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong, 510515, People's Republic of China.
| |
Collapse
|
2
|
Kohlhauser M, Tuca A, Kamolz LP. The efficacy of adipose-derived stem cells in burn injuries: a systematic review. Cell Mol Biol Lett 2024; 29:10. [PMID: 38182971 PMCID: PMC10771009 DOI: 10.1186/s11658-023-00526-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/18/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Burn injuries can be associated with prolonged healing, infection, a substantial inflammatory response, extensive scarring, and eventually death. In recent decades, both the mortality rates and long-term survival of severe burn victims have improved significantly, and burn care research has increasingly focused on a better quality of life post-trauma. However, delayed healing, infection, pain and extensive scar formation remain a major challenge in the treatment of burns. ADSCs, a distinct type of mesenchymal stem cells, have been shown to improve the healing process. The aim of this review is to evaluate the efficacy of ADSCs in the treatment of burn injuries. METHODS A systematic review of the literature was conducted using the electronic databases PubMed, Web of Science and Embase. The basic research question was formulated with the PICO framework, whereby the usage of ADSCs in the treatment of burns in vivo was determined as the fundamental inclusion criterion. Additionally, pertinent journals focusing on burns and their treatment were screened manually for eligible studies. The review was registered in PROSPERO and reported according to the PRISMA statement. RESULTS Of the 599 publications screened, 21 were considered relevant to the key question and were included in the present review. The included studies were almost all conducted on rodents, with one exception, where pigs were investigated. 13 of the studies examined the treatment of full-thickness and eight of deep partial-thickness burn injuries. 57,1 percent of the relevant studies have demonstrated that ADSCs exhibit immunomodulatory effects during the inflammatory response. 16 studies have shown improved neovascularisation with the use of ADSCs. 14 studies report positive influences of ADSCs on granulation tissue formation, while 11 studies highlight their efficacy in promoting re-epithelialisation. 11 trials demonstrated an improvement in outcomes during the remodelling phase. CONCLUSION In conclusion, it appears that adipose-derived stem cells demonstrate remarkable efficacy in the field of regenerative medicine. However, the usage of ADSCs in the treatment of burns is still at an early experimental stage, and further investigations are required in order to examine the potential usage of ADSCs in future clinical burn care.
Collapse
Affiliation(s)
- Michael Kohlhauser
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria.
| | - Alexandru Tuca
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
- Department of Surgery, State Hospital Güssing, Güssing, Austria
| | - Lars-Peter Kamolz
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
- COREMED-Cooperative Centre for Regenerative Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, Graz, Austria
| |
Collapse
|
3
|
Ye P, Gu R, Zhu H, Chen J, Han F, Nie X. SOX family transcription factors as therapeutic targets in wound healing: A comprehensive review. Int J Biol Macromol 2023; 253:127243. [PMID: 37806414 DOI: 10.1016/j.ijbiomac.2023.127243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
The SOX family plays a vital role in determining the fate of cells and has garnered attention in the fields of cancer research and regenerative medicine. It also shows promise in the study of wound healing, as it actively participates in the healing processes of various tissues such as skin, fractures, tendons, and the cornea. However, our understanding of the mechanisms behind the SOX family's involvement in wound healing is limited compared to its role in cancer. Gaining insight into its role, distribution, interaction with other factors, and modifications in traumatized tissues could provide valuable new knowledge about wound healing. Based on current research, SOX2, SOX7, and SOX9 are the most promising members of the SOX family for future interventions in wound healing. SOX2 and SOX9 promote the renewal of cells, while SOX7 enhances the microvascular environment. The SOX family holds significant potential for advancing wound healing research. This article provides a comprehensive review of the latest research advancements and therapeutic tools related to the SOX family in wound healing, as well as the potential benefits and challenges of targeting the SOX family for wound treatment.
Collapse
Affiliation(s)
- Penghui Ye
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Rifang Gu
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; School Medical Office, Zunyi Medical University, Zunyi 563006, China
| | - Huan Zhu
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Jitao Chen
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China
| | - Felicity Han
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xuqiang Nie
- Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China; College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
4
|
Schneider I, Calcagni M, Buschmann J. Adipose-derived stem cells applied in skin diseases, wound healing and skin defects: a review. Cytotherapy 2023; 25:105-119. [PMID: 36115756 DOI: 10.1016/j.jcyt.2022.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/17/2022] [Accepted: 08/11/2022] [Indexed: 01/18/2023]
Abstract
Adipose tissue presents a comparably easy source for obtaining stem cells, and more studies are increasingly investigating the therapeutic potential of adipose-derived stem cells. Wound healing, especially in chronic wounds, and treatment of skin diseases are some of the fields investigated. In this narrative review, the authors give an overview of some of the latest studies concerning wound healing as well as treatment of several skin diseases and concentrate on the different forms of application of adipose-derived stem cells.
Collapse
Affiliation(s)
| | - Maurizio Calcagni
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Johanna Buschmann
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
5
|
Bellei B, Migliano E, Picardo M. Therapeutic potential of adipose tissue-derivatives in modern dermatology. Exp Dermatol 2022; 31:1837-1852. [PMID: 35102608 DOI: 10.1111/exd.14532] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 12/14/2022]
Abstract
Stem cell-mediated therapies in combination with biomaterial and growth factor-based approaches in regenerative medicine are rapidly evolving with increasing application beyond the dermatologic field. Adipose-derived stem cells (ADSCs) are the more frequently used adult stem cells due to their abundance and easy access. In the case of volumetric defects, adipose tissue can take the shape of defects, restoring the volume and enhancing the regeneration of receiving tissue. When regenerative purposes prevail on volume restoration, the stromal vascular fraction (SVF) rich in staminal cells, purified mesenchymal stem cells (MSCs) or their cell-free derivatives grafting are favoured. The therapeutic efficacy of acellular approaches is explained by the fact that a significant part of the natural propensity of stem cells to repair damaged tissue is ascribable to their secretory activity that combines mitogenic factors, cytokines, chemokines and extracellular matrix components. Therefore, the secretome's ability to modulate multiple targets simultaneously demonstrated preclinical and clinical efficacy in reversing pathological mechanisms of complex conditions such atopic dermatitis (AD), vitiligo, psoriasis, acne and Lichen sclerosus (LS), non-resolving wounds and alopecia. This review analysing both in vivo and in vitro models gives an overview of the clinical relevance of adipose tissue-derivatives such as autologous fat graft, stromal vascular fraction, purified stem cells and secretome for skin disorders application. Finally, we highlighted the major disease-specific limitations and the future perspective in this field.
Collapse
Affiliation(s)
- Barbara Bellei
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Emilia Migliano
- Department of Plastic and Regenerative Surgery, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Mauro Picardo
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| |
Collapse
|
6
|
The Role of Gap Junctions in the Generation of Smooth Muscle Cells from Bone Marrow Mesenchymal Stem Cells. DISEASE MARKERS 2022; 2022:1491327. [PMID: 35990247 PMCID: PMC9391152 DOI: 10.1155/2022/1491327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022]
Abstract
Background. Studies have shown that stem cell transplantation can improve smooth muscle cell (SMC) regeneration and remodelling. Gap junctions can enhance the cytoprotective effects of neighbouring cells. We investigated the effect of gap junctions on the differentiation of bone marrow mesenchymal stem cells (BMSCs) into SMCs. Materials and Methods. Rat BMSCs and SMCs were obtained from the bone marrow and bladder of Sprague-Dawley rats, respectively. Flow cytometry and multilineage differentiation were performed to assess the characteristics of these cells. BMSCs and SMCs were incubated together in cocultures in the presence and absence of heptanol, an uncoupler of gap junctions. Cocultures were divided into three groups consisting of a contact coculture, noncontact coculture, and contact coculture plus heptanol groups. The expression of BMSC-specific markers and the effect of gap junctions on the differentiation of BMSCs were evaluated by performing real-time reverse transcription-polymerase chain reaction, immunofluorescence analysis, and western blotting after cocultures. Results. CD90 and CD44 were markedly expressed, and CD31 and CD45 were weakly or not expressed in BMSCs. The cells also showed good osteogenic and adipogenic differentiation ability. Compared with the noncontact coculture group, the SMC markers such as α-SMA, calponin, and connexin43 increased in the contact coculture group. The effect of contact in the coculture group was significantly weakened by heptanol. Conclusions. The results suggested that gap junctions play an important role in the generation of SMCs from BMSCs. The formation of SMCs can potentially be used to repair the sphincter muscle of patients with stress urinary incontinence.
Collapse
|
7
|
Zhu Z, Zhang X, Hao H, Xu H, Shu J, Hou Q, Wang M. Exosomes Derived From Umbilical Cord Mesenchymal Stem Cells Treat Cutaneous Nerve Damage and Promote Wound Healing. Front Cell Neurosci 2022; 16:913009. [PMID: 35846563 PMCID: PMC9279568 DOI: 10.3389/fncel.2022.913009] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/02/2022] [Indexed: 11/13/2022] Open
Abstract
Wound repair is a key step in the treatment of skin injury caused by burn, surgery, and trauma. Various stem cells have been proven to promote wound healing and skin regeneration as candidate seed cells. Therefore, exosomes derived from stem cells are emerging as a promising method for wound repair. However, the mechanism by which exosomes promote wound repair is still unclear. In this study, we reported that exosomes derived from umbilical cord mesenchymal stem cells (UC-MSCs) promote wound healing and skin regeneration by treating cutaneous nerve damage. The results revealed that UC-MSCs exosomes (UC-MSC-Exo) promote the growth and migration of dermal fibroblast cells. In in vitro culture, dermal fibroblasts could promote to nerve cells and secrete nerve growth factors when stimulated by exosomes. During the repair process UC-MSC-Exo accelerated the recruitment of fibroblasts at the site of trauma and significantly enhanced cutaneous nerve regeneration in vivo. Interestingly, it was found that UC-MSC-Exo could promote wound healing and skin regeneration by recruiting fibroblasts, stimulating them to secrete nerve growth factors (NGFs) and promoting skin nerve regeneration. Therefore, we concluded that UC-MSC-Exo promote cutaneous nerve repair, which may play an important role in wound repair and skin regeneration.
Collapse
Affiliation(s)
- Ziying Zhu
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, China
- The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- *Correspondence: Ziying Zhu,
| | - Xiaona Zhang
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, China
| | - Haojie Hao
- The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Heran Xu
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, China
| | - Jun Shu
- The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Qian Hou
- The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- Medical Innovation Research Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- Qian Hou,
| | - Min Wang
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing, China
- Min Wang,
| |
Collapse
|
8
|
Zhang Z, Chen W, Tiemessen DM, Oosterwijk E, Kouwer PHJ. A Temperature-Based Easy-Separable (TempEasy) 3D Hydrogel Coculture System. Adv Healthc Mater 2022; 11:e2102389. [PMID: 35029325 PMCID: PMC11469334 DOI: 10.1002/adhm.202102389] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/10/2021] [Indexed: 12/13/2022]
Abstract
Interactions between different cell types are crucial for their behavior in tissues, but are rarely considered in 3D in vitro cell culture experiments. One reason is that such coculture experiments are sometimes difficult to perform in 3D or require specialized equipment or know-how. Here, a new 3D cell coculture system is introduced, TempEasy, which is readily applied in any cell culture lab. The matrix material is based on polyisocyanide hydrogels, which closely resemble the mechanical characteristics of the natural extracellular matrix. Gels with different gelation temperatures, seeded with different cells, are placed on top of each other to form an indirect coculture. Cooling reverses gelation, allowing cell harvesting from each layer separately, which benefits downstream analysis. To demonstrate the potential of TempEasy , human adipose stem cells (hADSCs) with vaginal epithelial fibroblasts are cocultured. The analysis of a 7-day coculture shows that hADSCs promote cell-cell interaction of fibroblasts, while fibroblasts promote proliferation and differentiation of hADSCs. TempEasy provides a straightforward operational platform for indirect cocultures of cells of different lineages in well-defined microenvironments.
Collapse
Affiliation(s)
- Zhaobao Zhang
- Institute for Molecules and MaterialsRadboud University NijmegenHeyendaalseweg 135Nijmegen6525 AJThe Netherlands
| | - Wen Chen
- Institute for Molecules and MaterialsRadboud University NijmegenHeyendaalseweg 135Nijmegen6525 AJThe Netherlands
| | - Dorien M. Tiemessen
- Department of UrologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterGeert Grooteplein Zuid 28Nijmegen6525 GAThe Netherlands
| | - Egbert Oosterwijk
- Department of UrologyRadboud Institute for Molecular Life SciencesRadboud University Medical CenterGeert Grooteplein Zuid 28Nijmegen6525 GAThe Netherlands
| | - Paul H. J. Kouwer
- Institute for Molecules and MaterialsRadboud University NijmegenHeyendaalseweg 135Nijmegen6525 AJThe Netherlands
| |
Collapse
|
9
|
Bellei B, Migliano E, Picardo M. Research update of adipose tissue-based therapies in regenerative dermatology. Stem Cell Rev Rep 2022; 18:1956-1973. [PMID: 35230644 DOI: 10.1007/s12015-022-10328-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/01/2022] [Indexed: 12/09/2022]
Abstract
Mesenchymal stromal/stem cells (MSCs) have a spontaneous propensity to support tissue homeostasis and regeneration. Among the several sources of MSCs, adipose-derived tissue stem cells (ADSCs) have received major interest due to the higher mesenchymal stem cells concentration, ease, and safety of access. However, since a significant part of the natural capacity of ADSCs to repair damaged tissue is ascribable to their secretory activity that combines mitogenic factors, cytokines, chemokines, lipids, and extracellular matrix components, several studies focused on cell-free strategies. Furthermore, adipose cell-free derivatives are becoming more attractive especially for non-volumizing purposes, such as most dermatological conditions. However, when keratinocytes, fibroblasts, melanocytes, adipocytes, and hair follicle cells might not be locally sourced, graft of materials containing concentrated ADSCs is preferred. The usage of extracellular elements of adipose tissue aims to promote a self-autonomous regenerative microenvironment in the receiving area restoring physiological homeostasis. Hence, ADSCs or their paracrine activity are currently being studied in several dermatological settings including wound healing, skin fibrosis, burn, and aging.The present work analyzing both preclinical and clinical experiences gives an overview of the efficacy of adipose tissue-derivatives like autologous fat, the stromal vascular fraction (SVF), purified ADSCs, secretome and extracellular matrix graft in the field of regenerative medicine for the skin.
Collapse
Affiliation(s)
- Barbara Bellei
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Via Elio Chianesi 53, 00144, Rome, Italy.
| | - Emilia Migliano
- Department of Plastic and Reconstructive Surgery, San Gallicano Dermatological Institute, IRCCS, Via Elio Chianesi 53, 00144, Rome, Italy
| | - Mauro Picardo
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, Via Elio Chianesi 53, 00144, Rome, Italy
| |
Collapse
|
10
|
Immunomodulation of Skin Repair: Cell-Based Therapeutic Strategies for Skin Replacement (A Comprehensive Review). Biomedicines 2022; 10:biomedicines10010118. [PMID: 35052797 PMCID: PMC8773777 DOI: 10.3390/biomedicines10010118] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 12/12/2022] Open
Abstract
The immune system has a crucial role in skin wound healing and the application of specific cell-laden immunomodulating biomaterials emerged as a possible treatment option to drive skin tissue regeneration. Cell-laden tissue-engineered skin substitutes have the ability to activate immune pathways, even in the absence of other immune-stimulating signals. In particular, mesenchymal stem cells with their immunomodulatory properties can create a specific immune microenvironment to reduce inflammation, scarring, and support skin regeneration. This review presents an overview of current wound care techniques including skin tissue engineering and biomaterials as a novel and promising approach. We highlight the plasticity and different roles of immune cells, in particular macrophages during various stages of skin wound healing. These aspects are pivotal to promote the regeneration of nonhealing wounds such as ulcers in diabetic patients. We believe that a better understanding of the intrinsic immunomodulatory features of stem cells in implantable skin substitutes will lead to new translational opportunities. This, in turn, will improve skin tissue engineering and regenerative medicine applications.
Collapse
|
11
|
Md Fadilah NI, Mohd Abdul Kader Jailani MS, Badrul Hisham MAI, Sunthar Raj N, Shamsuddin SA, Ng MH, Fauzi MB, Maarof M. Cell secretomes for wound healing and tissue regeneration: Next generation acellular based tissue engineered products. J Tissue Eng 2022; 13:20417314221114273. [PMID: 35923177 PMCID: PMC9340325 DOI: 10.1177/20417314221114273] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/01/2022] [Indexed: 12/20/2022] Open
Abstract
Wound represents a significant socioeconomic burden for both affected individuals and as a whole healthcare system. Accordingly, stem cells have garnered attention due to their differentiation capacity and ability to aid tissue regeneration by releasing biologically active molecules, found in the cells' cultivated medium which known as conditioned medium (CM) or secretomes. This acellular approach provides a huge advantage over conventional treatment options, which are mainly used cellular treatment at wound closure. Interestingly, the secretomes contained the cell-secreted proteins such as growth factors, cytokines, chemokines, extracellular matrix (ECM), and small molecules including metabolites, microvesicles, and exosomes. This review aims to provide a general view on secretomes and how it is proven to have great potential in accelerating wound healing. Utilizing the use of secretomes with its secreted proteins and suitable biomaterials for fabrications of acellular skin substitutes can be promising in treating skin loss and accelerate the healing process.
Collapse
Affiliation(s)
- Nur Izzah Md Fadilah
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| | | | - Muhd Aliff Iqmal Badrul Hisham
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| | - Nithiaraj Sunthar Raj
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| | - Sharen Aini Shamsuddin
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| | - Manira Maarof
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| |
Collapse
|
12
|
Ma J, Zhang Z, Wang Y, Shen H. Investigation of miR-126-3p loaded on adipose stem cell-derived exosomes for wound healing of full-thickness skin defects. Exp Dermatol 2021; 31:362-374. [PMID: 34694648 DOI: 10.1111/exd.14480] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 09/01/2021] [Accepted: 10/16/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To investigate the function of miR-126-3p loaded on adipose stem cell (ADSC)-derived exosomes (ADSC-Exos) in wound healing of full-thickness skin defects. METHODS ADSCs transfected with miR-126-3p mimic, miR-126-3p inhibitor or pcDNA3.1-PIK3R2, or PKH26-marked ADSC-Exos were cultured with fibroblasts or human umbilical vein endothelial cells (HUVECs). The proliferation and migration rates of fibroblasts and angiogenesis of HUVECs were measured. Rats with full-thickness skin defects were injected with ADSC-Exos or exosomes extracted from ADSCs transfected with miR-126-3p inhibitor and the wound healing rates were measured. The wound bed, collagen deposition and angiogenesis in injured rats were assessed. RESULTS ADSC-Exos could be ingested by fibroblasts and HUVECs. Co-incubation with ADSCs or ADSC-Exos promoted the proliferation and migration of fibroblasts and angiogenesis of HUVECs, which was further enhanced by miR-126-3p overexpression. Inhibition of ADSC-Exos or miR-126-3p or PIK3R2 overexpression suppressed the proliferation and migration of fibroblasts and angiogenesis of HUVECs. ADSC-derived exosomal miR-126-3p increased wound healing rate, collagen deposition and newly formed vessels in injured rats. CONCLUSION ADSC-derived exosomal miR-126-3p promotes wound healing of full-thickness skin defects by targeting PIK3R2.
Collapse
Affiliation(s)
- Jie Ma
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Zhaofeng Zhang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Yinmin Wang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Hua Shen
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| |
Collapse
|
13
|
Lv M, Zhang S, Jiang B, Cao S, Dong Y, Cao L, Guo S. Adipose-derived stem cells regulate metabolic homeostasis and delay aging by promoting mitophagy. FASEB J 2021; 35:e21709. [PMID: 34143518 DOI: 10.1096/fj.202100332r] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/26/2021] [Accepted: 05/17/2021] [Indexed: 12/12/2022]
Abstract
Tissues undergo a process of degeneration as the body ages. Mesenchymal stem cells (MSCs) have been found to have major potential in delaying the aging process in tissues and organs. However, the mechanism underlying the anti-aging effects of MSC is not clear which limits clinical applications. In this study, we used adipose-derived mesenchymal stem cells (ADSCs) to perform anti-aging treatments on senescent cells and progeroid animal models. Following intervention with ADSCs, replicative senescence was delayed and metabolic homeostasis was transformed from catabolism to anabolism. Metabolomic tests were used to analyze different metabolites. We found that ADSCs acted to accelerate mitophagy which eliminated intracellular ROS and improved the quality of mitochondria. These processes acted to regulate the cellular metabolic homeostasis and ultimately delayed the process of aging. Allogeneic stem cell therapy in a Progeria animal model (DNA polymerase gamma (POLG) knockin, mitochondrial dysfunction) also showed that ADSC therapy can improve alopecia and kyphosis by promoting mitophagy. Our research confirms for the first time that allogeneic stem cell therapy can improve aging-related symbols and phenotypes through mitochondrial quality control. These results are highly significant for the future applications of stem cells in aging-related diseases.
Collapse
Affiliation(s)
- Mengzhu Lv
- Department of Plastic Surgery, China Medical University the First Hospital, Shenyang, P.R. China
| | - Simeng Zhang
- Department of Medical Oncology, China Medical University the First Hospital, Shenyang, P.R. China
| | - Bo Jiang
- Key Laboratory of Medical Cell Biology, Ministry of Education, Shenyang, P.R. China.,Institute of Translational Medicine, China Medical University, Shenyang, P.R. China.,Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, P.R. China
| | - Sunrun Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education, Shenyang, P.R. China.,Institute of Translational Medicine, China Medical University, Shenyang, P.R. China.,Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, P.R. China
| | - Yuqing Dong
- Department of Plastic Surgery, China Medical University the First Hospital, Shenyang, P.R. China
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education, Shenyang, P.R. China.,Institute of Translational Medicine, China Medical University, Shenyang, P.R. China.,Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, P.R. China
| | - Shu Guo
- Department of Plastic Surgery, China Medical University the First Hospital, Shenyang, P.R. China
| |
Collapse
|
14
|
Bounds K, Colmer-Hamood JA, Myntti M, Jeter RM, Hamood AN. The influence of a biofilm-dispersing wound gel on the wound healing process. Int Wound J 2021; 19:553-572. [PMID: 34263993 PMCID: PMC8874046 DOI: 10.1111/iwj.13653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/14/2021] [Accepted: 06/21/2021] [Indexed: 01/02/2023] Open
Abstract
Topical antimicrobials that reduce the bacterial bioburden within a chronically‐infected wound may have helpful or harmful effects on the healing process. We used murine models of full‐thickness skin wounds to determine the effects of the novel biofilm‐dispersing wound gel (BDWG) and its gel base on the healing of uninfected wounds. The rate of wound closure over 19 days was comparable among the BDWG‐treated (BT) wounds and the controls. Compared with the controls, histology of the BT wounds showed formation of a stable blood clot at day 1, more neovascularisation and reepithelialisation at day 3, and more organised healing at day 7. Fluorescence‐activated cell sorting analysis showed a lower percentage of neutrophils in wounded tissues of the BT group at days 1 and 3, and significantly more M2 macrophages at day 3. Levels of proinflammatory cytokines and chemokines were increased over the uninjured baseline within the wounds of all treatment groups but the levels were significantly lower in the BT group at day 1, modulating the inflammatory response. Our results suggest that BDWG does not interfere with the wound healing process and may enhance it by lowering inflammation and allowing transition to the proliferative stage of wound healing by day 3.
Collapse
Affiliation(s)
- Kayla Bounds
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, USA.,Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Jane A Colmer-Hamood
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas, USA.,Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Matthew Myntti
- Research and Development, Next Science LLC, Jacksonville, Florida, USA
| | - Randall M Jeter
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Abdul N Hamood
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas, USA.,Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| |
Collapse
|
15
|
Human Adipose-Derived Stem/Stromal Cells Promote Proliferation and Migration in Head and Neck Cancer Cells. Cancers (Basel) 2021; 13:cancers13112751. [PMID: 34206064 PMCID: PMC8199568 DOI: 10.3390/cancers13112751] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Fat grafts obtained from a minimal invasive liposuction device contain multipotent stem cells termed adipose-derived stem/stromal cells (ASCs). ASCs can be used for their proposed wound healing relevant characteristics, including for tissue defects in cancer patients. For head and neck cancers, little is known about the effects of ASCs on tumor cells. Using supernatants of ASCs from five patients in different functional experiments, this study aimed to investigate how ASCs influence tumor growth, invasive properties, and neoangiogenesis. The data show that all mentioned characteristics are promoted by fat graft stem cells in vitro in head and neck cancer cell lines. Although clinical relevance of these in vitro findings is unclear, due to the lack of in vivo and clinical data, fat grafts should be used cautiously and complete removal of tumor should be ensured before augmentation in head and neck cancer patients is performed. Abstract Human adipose-derived stem/stromal cells (ASCs) are increasingly used as auto-transplants in regenerative medicine to restore tissue defects or induce wound healing, especially in cancer patients. The impact of ASCs on squamous cell carcinoma of the upper aerodigestive tract (UAT) including head and neck and esophageal squamous cell carcinoma (HNSCC and ESCC) is not yet fully understood. ASCs were cultured from subcutaneous, abdominal lipoaspirates of five patients, who received auto-transplants to the head and neck. Supernatants were tested for paracrine effects in functional in vitro assays of proliferation of HNSCC tumor cell line FaDu and ESCC cell line Kyse30, and their cell migration/invasion capacities in Boyden chambers, in addition to endothelial tube formation assay using human umbilical vein endothelial cells (HUVECs). All ASC-derived supernatants enhanced proliferation of FaDu cells, invasive migration, and tube formation by HUVECs, compared to controls. Of five patients’ lipoaspirates, ASC-derived supernatants of four patients increased proliferation and invasive migration in Kyse30 cells. The data suggests that ASCs can promote tumor cell proliferation, invasiveness, and neo-angiogenesis in these tumor cell lines of the UAT and HUVEC in a paracrine manner. Although clinical studies on the subject of oncological safety are still needed, these findings emphasize the importance of complete tumor removal before ASCs are used in the head and neck.
Collapse
|
16
|
Murlistyarini S, Aninda LP, Widyarti S, Endharti AT, Sardjono TW. Exosomes of Adipose-derived Stem Cells Conditioned Media Promotes Retinoblastoma and Forkhead-Box M1 Protein Expression. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.6195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: In the senescence process, the retinoblastoma (Rb) protein binds to E2F in hypophosphorylated conditions, preventing the cell to enter the S-phase in the cell cycle. Human Forkhead Box M1 (FOXM1) protein, key regulator G1/S and G2/M phases, decreases in the senescence process. Many studies have been carried out to reverse this system, one of which used exosomes of adipose-derived stem c ells conditioned media (ADSC-CM). These exosomes contain a variety of specific proteins which have pro-proliferation properties, however, little is known on the role of these exosomes toward the change of phosphorylated Rb and FOXM1.
AIM: This study aims to find out the involvement of exosomes of ADSC-CM on these two proteins on senescence human dermal fibroblasts (HDFs).
METHODS: In vitro experiment was undergone randomization sample and non-blinded pre-/post-test control group. The primary culture of senescent HDFs was transfected with exosomes of ADSC-CM; then, its effect on migration and senescence reversal was observed through analyzing Sa-β-gal, Rb, and FOXM1 protein expression.
RESULTS: The expression of Sa-β-gal was higher in the control group. Our result demonstrated the exosome of ADSC-CM significantly induced the expression of Rb and FOXM1 protein in senescent HDFs (p < 0.05).
CONCLUSION: It proved that exosomes of ADSC-CM could shift the senescent fibroblast into metabolically active cells.
Collapse
|
17
|
Liu SJ, Meng MY, Han S, Gao H, Zhao YY, Yang Y, Lin ZY, Yang LR, Zhu K, Han R, Huang WW, Wang RQ, Yang LL, Wang WJ, Li L, Wang XD, Hou ZL, Liao LW, Yang L. Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes Ameliorate HaCaT Cell Photo-Aging. Rejuvenation Res 2021; 24:283-293. [PMID: 33607932 DOI: 10.1089/rej.2020.2313] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Umbilical cord mesenchymal stem cells (UCMSCs) have been identified as a potentially ideal cell type for use in regenerative therapeutic contexts owing to their excellent paracrine secretory abilities and other desirable properties. Previous work has shown that stem cell-derived exosomes can effectively reduce skin aging, but few studies have specifically focused on the role of UCMSC-derived exosomes in this context. In this study, we isolated exosomes derived from UCMSCs grown in a three-dimensional culture system and explored their ability to modulate the photo-aging of HaCaT keratinocytes. Cell viability and proliferation were assessed using CCK8 assay, whereas wound healing and transwell assays were used to assess cell migratory capabilities. UVB irradiation (60 mJ/cm2) was used to induce photo-aging of HaCaT cells. TUNEL and SA-β-Gal staining were used to explore HaCaT cell apoptosis and senescence, respectively, whereas real-time quantitative PCR was used to assess the expression of relevant genes at the mRNA level. We found that UCMSC-derived exosomes were able to enhance normal HaCaT cell proliferation and migration while also inhibiting UVB-induced damage to these cells. These exosomes also reduced HaCaT cell apoptosis and senescence, increasing collagen type I expression and reducing matrix metalloproteinase (MMP1) expression in photo-aged HaCaT cells. Together, these findings indicate that UCMSC-derived exosomes have the potential to be used therapeutically to suppress skin aging.
Collapse
Affiliation(s)
- Shi-Jie Liu
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Ming-Yao Meng
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Shen Han
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Hui Gao
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Yi-Yi Zhao
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Yang Yang
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Zhu-Ying Lin
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Li-Rong Yang
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Kai Zhu
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Rui Han
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Wen-Wen Huang
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Run-Qing Wang
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Li-Li Yang
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Wen-Ju Wang
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Lin Li
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Xiao-Dan Wang
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Zong-Liu Hou
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Li-Wei Liao
- Central Laboratory of Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China.,Yunnan Cell Biology and Clinical Translation Research Center, Kunming, Yunnan, People's Republic of China.,Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Li Yang
- Department of Geriatrics, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| |
Collapse
|
18
|
Ni X, Shan X, Xu L, Yu W, Zhang M, Lei C, Xu N, Lin J, Wang B. Adipose-derived stem cells combined with platelet-rich plasma enhance wound healing in a rat model of full-thickness skin defects. Stem Cell Res Ther 2021; 12:226. [PMID: 33823915 PMCID: PMC8022317 DOI: 10.1186/s13287-021-02257-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/01/2021] [Indexed: 12/20/2022] Open
Abstract
Background Wound healing is impaired in patients with diabetes due to the multifactorial etiology of the disease, which limits the therapeutic efficacy of various approaches. This study hypothesizes that the combination of adipose-derived stem cells (ADSCs) and platelet-rich plasma (PRP) might achieve optimally efficient diabetic wound healing. Methods ADSCs were isolated from the adipose tissues of Sprague-Dawley (SD) rats. PRP was prepared by using a two-step centrifugation technique. A diabetic wound model was established on the backs of SD rats to evaluate the effect of ADSCs incorporated into PRP. Hematoxylin and eosin staining, immunofluorescence, and immunohistochemistry were performed to observe the changes in neovascularization. ELISA and Western blot were utilized to detect the angiogenesis-related protein expression levels. The proliferation of endothelial cells was assessed by the MTS assay. Results ADSCs incorporated into PRP induced a higher wound closure rate than ADSCs, PRP, and negative control. The expression levels of VEGF, p-STAT3, and SDF-1 in the ADSC+PRP group were higher than those in the other groups. Moreover, the proliferation of endothelial cells was strongly stimulated by treatment with the combination of ADSC-conditioned medium (ADSC-CM) and PRP. Conclusions PRP enhanced diabetic wound healing induced by ADSCs, and its promoting effect involved neovascularization.
Collapse
Affiliation(s)
- Xuejun Ni
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Xiuying Shan
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Lili Xu
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Wenjun Yu
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Mingliang Zhang
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Chen Lei
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Nating Xu
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Junyu Lin
- Department of Thyroid and Breast Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Biao Wang
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
19
|
Khazaei S, Keshavarz G, Bozorgi A, Nazari H, Khazaei M. Adipose tissue-derived stem cells: a comparative review on isolation, culture, and differentiation methods. Cell Tissue Bank 2021; 23:1-16. [PMID: 33616792 DOI: 10.1007/s10561-021-09905-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/01/2021] [Indexed: 01/14/2023]
Abstract
Adipose tissue-derived stem cells (ADSCs) are an available source of mesenchymal stem cells with the appropriate capacity to in vitro survive, propagate, and differentiate into cells from three lineages of ectoderm, mesoderm, and endoderm. The biological features of ADSCs depend on the donor physiology and health status, isolation procedure, culture conditions, and differentiation protocols used. Adipose tissue samples are provided by surgery and lipoaspiration-based methods and subjected to various mechanical and chemical digestion techniques to finally generate a heterogeneous mixture named stromal vascular fraction (SVF). ADSCs are purified through varied cell populations that exist within SVF and cultured under standard conditions to give rise to a highly rich resource of stem cells directly applied in the clinic or differentiated into a wide range of cells. The development and optimization of conventional isolation, expansion, and differentiation methods seem noteworthy to preserve the desirable biological functions of ADSCs in pre-clinical and clinical investigations.
Collapse
Affiliation(s)
- Saber Khazaei
- Department of Endodontics, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ghazal Keshavarz
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Azam Bozorgi
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Nazari
- Department of Orofacial Surgery, School of Dentistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
20
|
Skin Immunomodulation during Regeneration: Emerging New Targets. J Pers Med 2021; 11:jpm11020085. [PMID: 33573342 PMCID: PMC7911085 DOI: 10.3390/jpm11020085] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/25/2020] [Accepted: 01/07/2021] [Indexed: 02/08/2023] Open
Abstract
Adipose-Derived Stem Cells (ADSC) are present within the hypodermis and are also expected to play a pivotal role in wound healing, immunomodulation, and rejuvenation activities. They orchestrate, through their exosome, the mechanisms associated to cell differentiation, proliferation, and cell migration by upregulating genes implicated in different functions including skin barrier, immunomodulation, cell proliferation, and epidermal regeneration. ADSCs directly interact with their microenvironment and specifically the immune cells, including macrophages and T and B cells, resulting in differential inflammatory and anti-inflammatory mechanisms impacting, in return, ADSCs microenvironment and thus skin function. These useful features of ADSCs are involved in tissue repair, where the required cell proliferation, angiogenesis, and anti-inflammatory responses should occur rapidly in damaged sites. Different pathways involved have been reported such as Growth Differentiation Factor-11 (GDF11), Tumor Growth Factor (TGF)-β, Metalloproteinase (MMP), microRNA, and inflammatory cytokines that might serve as specific biomarkers of their immunomodulating capacity. In this review, we try to highlight ADSCs’ network and explore the potential indicators of their immunomodulatory effect in skin regeneration and aging. Assessment of these biomarkers might be useful and should be considered when designing new clinical therapies using ADSCs or their specific exosomes focusing on their immunomodulation activity.
Collapse
|
21
|
Safi R, Malek E, Nemer G, Sayed R, Eid E, Khalil S, Nasser N, Abbas O, Mohsen-Kanson T, Kurban M. Comparative characterization of sun exposed and sun protected skin-derived mesenchymal-like stem cells in variegate porphyria and healthy individuals. PHOTODERMATOLOGY PHOTOIMMUNOLOGY & PHOTOMEDICINE 2020; 37:202-213. [PMID: 33259115 DOI: 10.1111/phpp.12635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 10/24/2020] [Accepted: 11/24/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND PURPOSE We hypothesized that upon sun exposure, a sub-population of primary skin-derived mesenchymal-like cells is deleteriously affected and thus contribute to the chronic inflammatory state in autosomal recessive variegate porphyria patients. The aim of this study was to isolate and characterize the mesenchymal-like stem cells from different areas of the skin in a porphyria patient (sun exposed, SE, and sun protected, SP) and to compare them with cells from a healthy individual. METHODS The proliferation rate and the migration ability of SE and SP cells were evaluated in the presence of an antioxidant compound, N-acetylcysteine. A co-culture of SE-damaged cells with the conditioned medium from the enriched mesenchymal cell-like SP population was performed in order to regenerate the dermal injured tissue after sun exposure in patients. RESULTS Results showed that the percentage of CD105+ cells varies between 3.9% in SP and 5% in SE of the healthy individual and between 3.6% and 1.4% in SP and SE in the porphyria patient, respectively. The osteogenic differentiation potential was lower in the porphyria patient when compared to the control. Furthermore, the expression of stem cell markers was more pronounced in SE than in SP cells of both control and porphyria. The use of N-acetyl cysteine did not show any beneficial effects on porphyria SE cells. Treatment with SP-conditioned medium slightly increased the expression of stem cell markers in SE of porphyria patient. CONCLUSION In conclusion, the pool of mesenchymal stem-like SE cells is affected in variegate porphyria patient along with modification of their self-renewal and differentiation properties.
Collapse
Affiliation(s)
- Rémi Safi
- Faculty of Medicine, Dermatology Department, American University of Beirut, Beirut, Lebanon
| | - Elie Malek
- Faculty of Sciences, Biology Department, Lebanese University, Zahle, Lebanon
| | - Georges Nemer
- Faculty of Medicine, Biochemistry and Molecular Genetics Department, American University of Beirut, Beirut, Lebanon
| | - Reem Sayed
- Faculty of Medicine, Dermatology Department, American University of Beirut, Beirut, Lebanon
| | - Edward Eid
- Faculty of Medicine, Dermatology Department, American University of Beirut, Beirut, Lebanon
| | - Samar Khalil
- Faculty of Medicine, Dermatology Department, American University of Beirut, Beirut, Lebanon
| | - Nourhane Nasser
- Faculty of Medicine, Dermatology Department, American University of Beirut, Beirut, Lebanon
| | - Ossama Abbas
- Faculty of Medicine, Dermatology Department, American University of Beirut, Beirut, Lebanon
| | - Tala Mohsen-Kanson
- Faculty of Sciences, Biology Department, Lebanese University, Zahle, Lebanon
| | - Mazen Kurban
- Faculty of Medicine, Dermatology Department, American University of Beirut, Beirut, Lebanon.,Faculty of Medicine, Biochemistry and Molecular Genetics Department, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
22
|
Predicting quality decay in continuously passaged mesenchymal stem cells by detecting morphological anomalies. J Biosci Bioeng 2020; 131:198-206. [PMID: 33121889 DOI: 10.1016/j.jbiosc.2020.09.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 01/21/2023]
Abstract
With rapid advances in cell therapy, technologies enabling both consistency and efficiency in cell manufacturing are becoming necessary. Morphological monitoring allows practical quality maintenance in cell manufacturing facilities, but relies heavily on human skill. For more reproducible and data-driven quality evaluation, image-based morphological analysis provides multiple advantages over manual observation. Our group has investigated the performance of multiple morphological parameters obtained from time-course images to non-invasively and quantitatively predict cellular quality using machine learning algorithms. Although such morphology-based computational models succeeded in early cell quality predictions, it was difficult to introduce our approach in cell manufacturing facilities owing to data variation issues. Since manufacturing facilities have fixed their protocol to minimize anomalies as much as possible, most accumulated data are normal, and anomalies are scarce. Thus, our morphological analysis had to adapt to such practical situation where it was difficult to observe a wide range of data variations, including both normal samples and anomalies, which is typically essential to improve most machine learning models' performance. In the present study, we introduce a practical morphological analysis concept by investigating the performance of anomalous quality decay discrimination during the continuous passaging of human mesenchymal stem cells (hMSCs). Combining the visualization method and asymmetric statistic discrimination, we describe an effective morphology-based, in-process quality monitoring concept to detect quality anomalies throughout cell culture process. Our results showed that the use of morphological parameters to reflect cellular population heterogeneity can predict hMSC quality decay within 6 h after seeding.
Collapse
|
23
|
Zhang R, Han S, Ren N, Liang L, Liang N, Liu F, Chen Y, Li D, Liu W, Liu H, Sun C. Topographical regulation of stem cell differentiation by plant-derived micro/nanostructures. NANOSCALE 2020; 12:18305-18312. [PMID: 32869818 DOI: 10.1039/d0nr02765k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
This study examines the osteogenic differentiation promotion effect of micro/nanostructures of raffia on human adipose-derived stem cells to confirm the potential application of plant-derived micro/nanotopographies in tissue regeneration. The results confirm that the nanorod array on the front surface and the honeycomb-like microstructure on the back surface of raffia can not only regulate the adhesion, spreading, and migration of stem cells but also promote the osteogenic differentiation of the stem cells at a subsequent stage of cell culture. The osteocalcin expressions by the 21-day cultured cells on the front and back surfaces of raffia were 55-fold and 36-fold higher compared to the expression on a tissue culture plate. This indicates that plant-derived micro/nanotopographies can significantly promote stem cell differentiation. Furthermore, a general strategy for the application of plant-derived materials to stem-cell differentiation and bone-tissue engineering is suggested.
Collapse
Affiliation(s)
- Ruitong Zhang
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, P. R. China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Mazini L, Rochette L, Malka G. Adipose-Derived Stem Cells (ADSCs) and Growth Differentiation Factor 11 (GDF11): Regenerative and Antiaging Capacity for the Skin. Regen Med 2020. [DOI: 10.5772/intechopen.91233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
25
|
Li T, Zhou X, Wang J, Liu Z, Han S, Wan L, Sun X, Chen H. Adipose-derived mesenchymal stem cells and extracellular vesicles confer antitumor activity in preclinical treatment of breast cancer. Pharmacol Res 2020; 157:104843. [PMID: 32360582 DOI: 10.1016/j.phrs.2020.104843] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/30/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023]
Abstract
Both antitumor and protumor property of mesenchymal stem cells (MSCs) have been demonstrated. We hypothesize that this contradiction is due to the heterogeneity of MSC subsets and that extracellular vesicles (EVs) from distinct MSC subsets can transfer the corresponding antitumor activities. Here we evaluated the antitumor activities of two subsets of adipose-derived mesenchymal stem cells (ADSCs) and ADSC-derived EVs (ADSC-EVs) in immunocompetent syngeneic mouse models of breast cancer. We identified CD90high and CD90low ADSC subsets and demonstrated that CD90high ADSCs could be converted into CD90low ADSCs by stimulation with LPS. CD90low ADSCs and its derived EVs significantly inhibited tumor growth in tumor-bearing mice. Benefit of tumor control were associated with decreased tumor cell proliferation and migration, and enhanced tumor cell apoptosis mediated by ADSC-EVs. Antioncogenic miRNA-16-5p loaded CD90low ADSC-EVs further significantly enhanced antitumor activities. Taken together, this study represents the first attempt to apply our newly identified antitumor ADSCs and its derived EVs in preclinical treatment of breast cancer. This study also provides the evidence that EVs can serve as a novel and effective therapeutics or drug delivery vesicle. This new therapeutic approach could be potentially applicable to breast cancer and many other types of cancer.
Collapse
Affiliation(s)
- Tao Li
- Jiangsu Key Laboratory of Clinical Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Xiaohe Zhou
- Jiangsu Key Laboratory of Clinical Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jingzhe Wang
- Jiangsu Key Laboratory of Clinical Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Ziyao Liu
- Jiangsu Key Laboratory of Clinical Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Sen Han
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Lu Wan
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Xiaochun Sun
- Jiangsu Key Laboratory of Clinical Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Huabiao Chen
- Vaccine and Immunotherapy Center, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Experimental Therapeutics and Molecular Imaging Laboratory, Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
26
|
Concentration Dependent Effect of Human Dermal Fibroblast Conditioned Medium (DFCM) from Three Various Origins on Keratinocytes Wound Healing. Int J Mol Sci 2020; 21:ijms21082929. [PMID: 32331278 PMCID: PMC7215860 DOI: 10.3390/ijms21082929] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022] Open
Abstract
Fibroblasts secrete many essential factors that can be collected from fibroblast culture medium, which is termed dermal fibroblast conditioned medium (DFCM). Fibroblasts isolated from human skin samples were cultured in vitro using the serum-free keratinocyte-specific medium (Epilife (KM1), or define keratinocytes serum-free medium, DKSFM (KM2) and serum-free fibroblast-specific medium (FM) to collect DFCM-KM1, DFCM-KM2, and DFCM-FM, respectively). We characterised and evaluated the effects of 100-1600 µg/mL DFCM on keratinocytes based on attachment, proliferation, migration and gene expression. Supplementation with 200-400 µg/mL keratinocyte-specific DFCM-KM1 and DFCM-KM2 enhanced the attachment, proliferation and migration of sub-confluent keratinocytes, whereas 200-1600 µg/mL DFCM-FM significantly increased the healing rate in the wound healing assay, and 400-800 µg/mL DFCM-FM was suitable to enhance keratinocyte attachment and proliferation. A real-time (RT2) profiler polymerase chain reaction (PCR) array showed that 42 genes in the DFCM groups had similar fold regulation compared to the control group and most of the genes were directly involved in wound healing. In conclusion, in vitro keratinocyte re-epithelialisation is supported by the fibroblast-secreted proteins in 200-400 µg/mL DFCM-KM1 and DFCM-KM2, and 400-800 µg/mL DFCM-FM, which could be useful for treating skin injuries.
Collapse
|
27
|
Gersch RP, Raum JC, Calvert C, Percec I. Fibroblasts Derived From Human Adipose Stem Cells Produce More Effective Extracellular Matrix and Migrate Faster Compared to Primary Dermal Fibroblasts. Aesthet Surg J 2020; 40:108-117. [PMID: 30873530 DOI: 10.1093/asj/sjz071] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The efficacy of adipose-derived stem cells (ASCs) to improve wound healing has been extensively investigated. Unfortunately, no consistent reports have described significant improvements in healing time or outcomes in large-scale clinical trials utilizing human ASCs. Primarily, these studies examined undifferentiated ASCs as opposed to specific cells differentiated from ASCs. OBJECTIVES The authors sought to examine the properties of fibroblasts differentiated from human ASCs (dFib cells) compared with those of primary dermal fibroblasts. METHODS ASCs were isolated from healthy female patients, differentiated into dFib cells, and compared with intra-patient primary dermal fibroblasts for morphology, extracellular matrix (ECM) marker expression, and cell migration employing qPCR, western blot, and scratch test assays. RESULTS De novo differentiated fibroblasts produce higher levels of the healthy ECM markers Elastin, Fibronectin, and Collagen 1 compared with primary fibroblasts. In contrast, dFib cells have reduced expression of the scar tissue markers αSMA, Collagen 3, and MMP-1. Further, dFib cells close scratch defects more quickly than primary dermal fibroblasts (32 ± 12.85 hours vs 64 ± 13.85 hours, P < 0.01) in a scratch test assay. CONCLUSIONS These data suggest that fibroblasts newly differentiated from human ASCs migrate well and produce a robust ECM, the combination of which may contribute to improved wound healing, and thus should be further investigated.
Collapse
Affiliation(s)
- Robert P Gersch
- Department of Surgery, Division of Plastic Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Jeffrey C Raum
- Department of Surgery, Division of Plastic Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Catherine Calvert
- Department of Surgery, Division of Plastic Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Ivona Percec
- Department of Surgery, Division of Plastic Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
28
|
Human Adipose-Derived Stem Cells Promote Seawater-Immersed Wound Healing by Activating Skin Stem Cells via the EGFR/MEK/ERK Pathway. Stem Cells Int 2019; 2019:7135974. [PMID: 32082387 PMCID: PMC7012271 DOI: 10.1155/2019/7135974] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/31/2019] [Accepted: 11/09/2019] [Indexed: 12/22/2022] Open
Abstract
Seawater (SW) immersion can increase the damage of skin wounds and produce refractory wounds. However, few studies have been conducted to investigate the mechanisms of SW immersion on skin wounds. In our current study, we investigated the effect of human adipose-derived stem cells (hADSCs) on the repair of SW-treated full-thickness skin wounds and the underlying mechanisms. The results showed that SW immersion could reduce the expression of EGF and suppress the activation of the MEK/ERK signaling pathway. At the same time, the proliferation and migration of skin stem cells were inhibited by SW immersion, resulting in delayed wound healing. However, hADSCs significantly accelerated the healing of SW-immersed skin wounds by promoting cell proliferation and migration through the aforementioned mechanisms. Our results indicate a role for hADSCs in the repair of seawater-immersed skin wounds and suggest a potential novel treatment strategy for seawater-immersed wound healing.
Collapse
|
29
|
Paganelli A, Benassi L, Rossi E, Magnoni C. Extracellular matrix deposition by adipose-derived stem cells and fibroblasts: a comparative study. Arch Dermatol Res 2019; 312:295-299. [PMID: 31616972 DOI: 10.1007/s00403-019-01997-8] [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: 07/09/2019] [Revised: 09/21/2019] [Accepted: 10/03/2019] [Indexed: 02/05/2023]
Abstract
Cell-based strategies are today widely studied as possible therapies for wound healing. In this setting, fibroblasts play a key role since they are the main dermal cellular component and are responsible for extracellular matrix secretion. Several works report on the possibility of using fibroblast-derived extracellular matrix scaffolds for wound healing in skin injuries. While fibroblast-based substitutes have already been intensively studied by other groups, we focused our attention on the possibility of creating an adipose-derived stem cell (ADSC)-induced dermal scaffold for wound healing. ADSCs are a particular subset of mesenchymal stem cells present in the stromal vascular fraction of the adipose tissue. The aim of our work was to compare the ability of ADSCs and fibroblast to produce in vitro a scaffolding material, both in terms of collagen and fibronectin production. ADSCs turned out to be capable of efficiently producing a collagen and fibronectin-containing dermal matrix upon stimulation with ascorbic acid. We observed fibronectin and collagen production by ADSCs to be even more abundant when compared to fibroblasts'. Our results support the use of ADSC-induced sheets instead of fibroblast-based dermal substitutes as wound-healing strategies in full-thickness skin injuries.
Collapse
Affiliation(s)
- Alessia Paganelli
- Surgical, Medical and Dental Department of Morphological Sciences Related To Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy.
| | - Luisa Benassi
- Surgical, Medical and Dental Department of Morphological Sciences Related To Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Elena Rossi
- Surgical, Medical and Dental Department of Morphological Sciences Related To Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Cristina Magnoni
- Surgical, Medical and Dental Department of Morphological Sciences Related To Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| |
Collapse
|
30
|
Paganelli A, Benassi L, Pastar I, Pellegrini M, Azzoni P, Vaschieri C, Pisciotta A, Carnevale G, Pellacani G, Magnoni C. In vitro Engineering of a Skin Substitute Based on Adipose-Derived Stem Cells. Cells Tissues Organs 2019; 207:46-57. [PMID: 31261153 DOI: 10.1159/000501071] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/21/2019] [Indexed: 11/19/2022] Open
Abstract
In the field of wound healing, stem cell-based strategies are gaining importance for their regenerative potential. Adipose-derived stem cells (ADSCs) are a particular subset of mesenchymal stem cells present in the stromal-vascular fraction of the adipose tissue, today considered very attractive for their relative abundance and accessibility in the human body. However, ADSCs are still not routinely used in normal clinical practice. Several studies have also reported ADSC transplantation in association with biomaterials in an attempt to enhance the local retention and growth rate of the cells. The aim of our study was to evaluate the ability of ADSCs to build a dermal scaffold to be potentially used as a dermal substitute in the field of wound healing, with optimal biocompatibility and mechanical properties. ADSCs were defined as CD90-, CD73-, and CD105-positive cells. ADSCs turned out to be capable of secreting all the main components of the extracellular matrix (ECM) upon stimulation, thus efficiently producing a collagen and fibronectin-containing dermal matrix. We also checked whether the ADSC-produced dermal scaffold could be seeded with keratinocytes. The scaffolding material directly produced by ADSCs has several advantages when compared to the commercially available ones: it is easily obtained from the patients and it is 100% biocompatible and supports cell-ECM interaction. Moreover, it represents a possible powerful therapeutic tool for patients with chronic ulcers since it appears to be potentially grafted with keratinocytes layers, thus bypassing the classical two-step grafting procedure.
Collapse
Affiliation(s)
- Alessia Paganelli
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy,
| | - Luisa Benassi
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Irena Pastar
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, Miami, Florida, USA
| | - Massimo Pellegrini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Paola Azzoni
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Vaschieri
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandra Pisciotta
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Gianluca Carnevale
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanni Pellacani
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Magnoni
- Surgical, Medical and Dental Department of Morphological Sciences Related to Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
31
|
Maarof M, Mh Busra MF, Lokanathan Y, Bt Hj Idrus R, Rajab NF, Chowdhury SR. Safety and efficacy of dermal fibroblast conditioned medium (DFCM) fortified collagen hydrogel as acellular 3D skin patch. Drug Deliv Transl Res 2019; 9:144-161. [PMID: 30547385 DOI: 10.1007/s13346-018-00612-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Skin substitutes are one of the main treatments for skin loss, and a skin substitute that is readily available would be the best treatment option. However, most cell-based skin substitutes require long production times, and therefore, patients endure long waiting times. The proteins secreted from the cells and tissues play vital roles in promoting wound healing. Thus, we aimed to develop an acellular three-dimensional (3D) skin patch with dermal fibroblast conditioned medium (DFCM) and collagen hydrogel for immediate treatment of skin loss. Fibroblasts from human skin samples were cultured using serum-free keratinocyte-specific media (KM1 or KM2) and serum-free fibroblast-specific medium (FM) to obtain DFCM-KM1, DFCM-KM2, and DFCM-FM, respectively. The acellular 3D skin patch was soft, semi-solid, and translucent. Collagen mixed with DFCM-KM1 and DFCM-KM2 showed higher protein release compared to collagen plus DFCM-FM. In vitro and in vivo testing revealed that DFCM and collagen hydrogel did not induce an immune response. The implantation of the 3D skin patch with or without DFCM on the dorsum of BALB/c mice demonstrated a significantly faster healing rate compared to the no-treatment group 7 days after implantation, and all groups had complete re-epithelialization at day 17. Histological analysis confirmed the structure and integrity of the regenerated skin, with positive expression of cytokeratin 14 and type I collagen in the epidermal and dermal layer, respectively. These findings highlight the possibility of using fibroblast secretory factors together with collagen hydrogel in an acellular 3D skin patch that can be used allogeneically for immediate treatment of full-thickness skin loss.
Collapse
Affiliation(s)
- Manira Maarof
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaccob Latiff, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Mohd Fauzi Mh Busra
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaccob Latiff, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Yogeswaran Lokanathan
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaccob Latiff, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Ruszymah Bt Hj Idrus
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaccob Latiff, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Nor Fadilah Rajab
- Bioserasi Laboratory, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor Darul Ehsan, Malaysia
| | - Shiplu Roy Chowdhury
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaccob Latiff, Cheras, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
32
|
Pesaraklou A, Mahdavi-Shahri N, Hassanzadeh H, Ghasemi M, Kazemi M, Mousavi NS, Matin MM. Use of cerium oxide nanoparticles: a good candidate to improve skin tissue engineering. ACTA ACUST UNITED AC 2019; 14:035008. [PMID: 30754036 DOI: 10.1088/1748-605x/ab0679] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Today advancements in nanotechnology have made extensive progress in tissue engineering. Application of cerium oxide nanoparticles (CeO2) has improved regenerative medicine due to their antioxidant properties. In this study, nanoparticles were used to increase the efficacy of skin substitutes. Human skin samples were decellularized using four methods and studied via histological stainings and DNA content analyses. Then CeO2 dispersing and its stability were investigated. The prepared acellular dermal matrices (ADMs) were immersed in CeO2 suspension and their effects were evaluated on growth of cultured human adipose derived-mesenchymal stem cells (hAd-MSCs) using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and histological methods. Moreover, their antioxidant properties were assessed based on DPPH degradation. Changes in the collagen contents of the scaffolds containing cells and CeO2 were also determined by electron microscopy and their tensile strength was compared to ADM. Our results indicated that use of trypsin/NaOH protocol resulted in most efficient cell removal while maintaining extracellular matrix (ECM) architecture. Among different dispersal methods, the approach using Dulbecco's modified Eagle's medium (DMEM), wetting with fetal bovine serum (FBS) and ultrasonic bath resulted in the best stability. Furthermore, it was shown that CeO2 not only had no toxicity on the cells, but also increased the growth and survival of hAd-MSCs by about 27%, improved free radical scavenging, as well as the amount of collagen and tensile strength of the scaffolds containing nanoparticles compared to the ADM. It can be concluded that the combination of ADM/CeO2/hAd-MSCs could be a step forward in skin tissue engineering.
Collapse
Affiliation(s)
- Atefeh Pesaraklou
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | | | | | | | | | | | | |
Collapse
|
33
|
Shi B, Wei W, Qin X, Zhao F, Duan Y, Sun W, Li D, Cao Y. Mapping theme trends and knowledge structure on adipose-derived stem cells: a bibliometric analysis from 2003 to 2017. Regen Med 2018; 14:33-48. [PMID: 30547725 DOI: 10.2217/rme-2018-0117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM To investigate the theme trends and knowledge structure of adipose-derived stem cells (ADSCs) related literatures by using bibliometric analysis. MATERIALS & METHODS Co-word analysis, strategic diagram and social network analysis were employed. RESULTS In line with strategic diagrams, ADSC differentiation and transplantation as main undeveloped themes in 2003-2007 were partially replaced by regeneration medicine and ADSCs for myocardial infarction in 2008 to 2012, and then partially replaced by miRNAs in ADSC genetics and nerve regeneration in 2013 to 2017. Based on social network analysis, regenerative medicine/methods, myocardial infarction/therapy, as well as miRNAs/genetics, and nerve regeneration/physiology were considered the emerging hot spots in 2008 to 2012 and 2013 to 2017. CONCLUSION The undeveloped themes and emerging hot spots could be considered as new research topics.
Collapse
Affiliation(s)
- Bei Shi
- Department of Physiology, College of Life Science, China Medical University, Shenyang 110122, PR China.,Functional Laboratory Center, College of Basic Medical Science, China Medical University, Shenyang 110122, PR China
| | - Wenjuan Wei
- Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, PR China.,Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, PR China
| | - Xin Qin
- Department of Physiology, College of Life Science, China Medical University, Shenyang 110122, PR China.,Biomedical Technology Cluster, Hong Kong Science and Technology Parks Corporation, 2 Science Park West Avenue, Hong Kong
| | - Fangkun Zhao
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110004, PR China
| | - Yucen Duan
- Department of Physiology, College of Life Science, China Medical University, Shenyang 110122, PR China
| | - Weinan Sun
- Department of Physiology, College of Life Science, China Medical University, Shenyang 110122, PR China
| | - Da Li
- Centerof Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Yu Cao
- Department of Physiology, College of Life Science, China Medical University, Shenyang 110122, PR China
| |
Collapse
|
34
|
Lai F, Kakudo N, Morimoto N, Taketani S, Hara T, Ogawa T, Kusumoto K. Platelet-rich plasma enhances the proliferation of human adipose stem cells through multiple signaling pathways. Stem Cell Res Ther 2018; 9:107. [PMID: 29661222 PMCID: PMC5902971 DOI: 10.1186/s13287-018-0851-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/18/2018] [Accepted: 03/20/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Platelet-rich plasma (PRP) is an autologous blood product that contains a high concentration of several growth factors. Platelet-derived growth factor (PDGF)-BB is a potential mitogen for human adipose-derived stem cells (hASCs). PRP stimulates proliferation of hASCs; however, the signaling pathways activated by PRP remain unclear. METHODS hASCs were cultured with or without PRP or PDGF-BB, and proliferation was assessed. hASCs were also treated with PRP or PDGF-BB with or without imatinib, which is a PDGF receptor tyrosine kinase inhibitor, or sorafenib, which is a multikinase inhibitor. Inhibition of cell proliferation was examined using anti-PDGF antibody (Abcam, Cambridge, UK), by cell counting. We assessed the effects of inhibitors of various protein kinases such as ERK1/2, JNK, p38, and Akt on the proliferation of hASCs. RESULTS The proliferation was remarkably promoted in cells treated with either 1% PRP or 10 ng/ml PDGF-BB, and both imatinib and sorafenib inhibited this proliferation. Anti-PDGF antibody (0.5 and 2 μg/ml) significantly decreased the proliferation of hASCs compared with control. PRP-mediated hASC proliferation was blocked by inhibitors of ERK1/2, Akt, and JNK, but not by an inhibitor of p38. CONCLUSIONS PRP promotes hASC proliferation, and PDGF-BB in PRP plays a major role in inducing the proliferation of hASCs. PRP promotes hASC proliferation via ERK1/2, PI3K/Akt, and JNK signaling pathways.
Collapse
Affiliation(s)
- Fangyuan Lai
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Natsuko Kakudo
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan.
| | - Naoki Morimoto
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Shigeru Taketani
- Department of Microbiology, Kansai Medical University, Osaka, 573-1010, Japan
| | - Tomoya Hara
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan.,Department of Oral Implantology, Osaka Dental University, Osaka, 573-1121, Japan
| | - Takeshi Ogawa
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| | - Kenji Kusumoto
- Department of Plastic and Reconstructive Surgery, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka, 573-1010, Japan
| |
Collapse
|
35
|
Miana VV, González EAP. Adipose tissue stem cells in regenerative medicine. Ecancermedicalscience 2018; 12:822. [PMID: 29662535 PMCID: PMC5880231 DOI: 10.3332/ecancer.2018.822] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Indexed: 12/26/2022] Open
Abstract
Adipose tissue-derived stem cells (ADSCs) are mesenchymal cells with the capacity for self-renewal and multipotential differentiation. This multipotentiality allows them to become adipocytes, chondrocytes, myocytes, osteoblasts and neurocytes among other cell lineages. Stem cells and, in particular, adipose tissue-derived cells, play a key role in reconstructive or tissue engineering medicine as they have already proven effective in developing new treatments. The purpose of this work is to review the applications of ADSCs in various areas of regenerative medicine, as well as some of the risks associated with treatment with ADSCs in neoplastic disease.
Collapse
Affiliation(s)
- Vanesa Verónica Miana
- Centre for Advanced Studies in Humanities and Health Sciences, Interamerican Open University, Buenos Aires, Argentina
| | - Elio A Prieto González
- Centre for Advanced Studies in Humanities and Health Sciences, Interamerican Open University, Buenos Aires, Argentina
| |
Collapse
|