Regenerative and stem cell-based techniques for facial rejuvenation

Buy Lab Tests Online

BadassBlues

Well-Known Member
Exp Biol Med (Maywood). 2021 Aug; 246(16): 1829–1837.
Published online 2021 Jun 8. doi: 10.1177/15353702211020701
PMCID: PMC8381699
PMID: 34102897

Regenerative and stem cell-based techniques for facial rejuvenation​

J Sarah Crowley,[IMG alt="corresponding author"]https://www.ncbi.nlm.nih.gov/corehtml/pmc/pmcgifs/corrauth.gif[/IMG] Amy Liu,[IMG alt="corresponding author"]https://www.ncbi.nlm.nih.gov/corehtml/pmc/pmcgifs/corrauth.gif[/IMG] and Marek Dobke[IMG alt="corresponding author"]https://www.ncbi.nlm.nih.gov/corehtml/pmc/pmcgifs/corrauth.gif[/IMG]
Author information Copyright and License information PMC Disclaimer


Go to:

Abstract​

This review discusses the most novel ideas and modalities being incorporated into facial rejuvenation. Recent innovative techniques include the use of regenerative stem cell techniques and regeneration supportive modalities such as nano-technology or gene therapies. This review aims to investigate approaches that are less well known and lacking established evidence in order to proactively study these techniques prior to them becoming popularized. These applications and relevant research were reviewed in the context of both surgical and non-surgical modalities in clinical practice. Future directions include the concept of “precision cosmetic medicine” utilizing gene editing and cellular therapies to tailor rejuvenation techniques based on each individual’s genetic make-up and therefore needs.

This review is focused on evaluation of methods of facial rejuvenation that have not yet been established as the “typical,” mainstream surgical and non-surgical approaches. The ever-reaching goal to obtain the “fountain of youth” makes this a rapidly evolving field in the scope of regenerative medicine. However, these types of changes are frequently investigated and ahead of regulatory and credentialing bodies. Practitioners use products “off-label” on a not infrequent basis, and updated reviews such as this can help to inform clinicians about the evolving standards of care and risks prior to regulatory process completion, while helping patients to achieve their goals of obtaining youth and improving quality of life.

Introduction​

Facial rejuvenation is a rapidly growing field as patients seek to obtain “the fountain of youth.” Moreover, there has been a significant amount of dedicated research with attempts to delay or reverse aging on both a cellular and macroscale level. A thorough approach to facial rejuvenation necessitates a multi-modal approach that addresses all of the changes that occur with aging including damage to the skin, volume loss of all the tissues of the face including fat and bone, and tissue laxity. With careful evaluation of all of these facets, it is clear that a surgical facelift alone would not address the volume loss and skin damage.

With aging, there are recognized molecular and histologic skin changes including fibroblast senescence, flattened dermal-epidermal junctions leading to the appearance of atrophy, and decrease in Langerhans and dermal cells. 1 Additionally, the facial bone structure also changes with age; the orbital aperture increases in width and area, and the mandible becomes thinner. 2 One of the most effective and logical ways to achieve facial rejuvenation is through regenerative aesthetics, which utilizes the tissue’s own natural potential to combat cell senescence and tissue atrophy through repairing of aging cells and the tissue matrix.

Regenerative interventions are defined as those leading towards renewal, restoration, and regrowth of damaged tissue.35 Advances in molecular biology, genetics, and medical technology as well as empirical clinical experience provide a basis of interventions utilizing potential direct regenerative properties (e.g. stem cells) or those having indirect potential by modulating mesenchymal cells or tissue milieu (e.g. gene/cellular therapy techniques, nanotechnology). 6 Additionally, with the advent of artificial intelligence (AI), we can utilize methods of imaging, molecular, and genetic studies to objectively evaluate patients prior to facial rejuvenation procedures. 7 Moreover, in the future, data-driven simulations can be utilized to predict tissue “needs” and predict outcomes. 7 This concept can be used in conjunction with the direct and already established use of stem cells.

With the combination of this multi-faceted approach, comes the idea of precision medicine for facial rejuvenation, whereby the goals are tailored to an individual’s genetic make-up and needs. This review aims to provide an updated resource on the dynamic changing of approaches for facial rejuvenation to give patients and practitioners a thorough understanding to both surgical and non-surgical approaches to restoring youth.

Autologous fat and regenerative cell types​

At the core of facial rejuvenation is the use of the regenerative properties of stem cells derived from autologous fat. The use of autologous fat for regenerative or reparative techniques was first described by Neuber in 1893 when he described harvesting a patient’s arm fat in order to correct facial scarring contour deformities. 8 Since that time there have been numerous methods, and described applications for the use of fat grafting to the body, but one of the most popularized purposes today is for facial rejuvenation and contouring.812

Today, indications for fat grafting to the face for facial rejuvenation include sun damage, volume deficiency such as orbital, tear trough, and temporal hollowing, skin laxity, and rhytids. 8 In addition, fat grafting can be combined with other procedures such as facelifts, blepharoplasty, and laser resurfacing treatments to improve outcomes of facial rejuvenation through a multi-modal technique. Furthermore, fat grafts are biocompatible, clinically versatile, safe and provide a natural appearance. 3

A well-described problem with fat grafting is the variable rates of fat grafting survival. This can range from 25 to 70%,13,14 leading to great efforts to improve fat harvesting, processing, and grafting methods.13,14 Coleman was one of the first to describe that the key to enhancing fat grafting survival was to inject in “miniscule amounts,” thereby increasing the contact of the fat to surrounding vascular tissues. 15 Additionally, protocols developed by investigators from our group standardized the most efficacious ways for procurement, isolation, characterization, and evaluation of human mesenchymal cells. 16

There are different theories for adipocyte survival. One theory proposes that dying adipocytes stimulate phagocytosis leading to transformation of these “wandering cells” into embryonic fat cells, mature cells, and connective tissue, 18 while another theory suggests that the final amount of fat survival depends on the number of viable adipocytes that were grafted. 17 Strategies have been attempted to improve fat grafting outcomes and survival through the discovery of regenerative stem cells. Given the complexity of stem cells and their interaction with the host environment, it may be that specific conditions, such as stem cell dose and timing, determine the anti-senescence (or pro-senescence) fate of fat grafts. 16

Since Zuk et al. first identified regenerative stem cells in adipose tissue, the use of cell therapy for tissue regeneration has become a rapidly evolving field such that regenerative cells including fat cells, adipose-derived stem cells, stromal vascular fraction (SVF), nanofat, and platelet-rich plasma (PRP) have been described specifically for the use of facial rejuvenation.4,1820 Through the use of these different types of regenerative tissues, one’s youth, beauty, and even function can be restored.

Aspirated adipose tissue is made up of adipocytes and progenitor cells, and it is now known that these preadipocyte progenitor and adipocyte-derived stem cells are attributed to the long-term survival of fat grafting. 13 Adipose-derived stem cells (ADSCs) are an alternate source (as opposed to mesenchymal cells derived from bone) of adult multipotent stem cells found within the perivascular adipose stroma. 4 These cells have the ability of self-renewal, differentiation into other mesoderm derivatives, and have paracrine properties, with ability of secreting growth factors and promoting angiogenesis and anti-apoptosis. 21 Due to ease of harvest and ability to harvest in large quantities, ADSCs have become a frequently used adult stem cell population for regenerative medicine and supplementation of fat grafts for facial contouring. 22

Not only do these cells have the ability to differentiate, but studies have also shown the trophic abilities of these stem cells. It appears that fat processing and enrichment with autologous stem cells improves fat cell viability and clinical volume retention (Figure 1). Intravenous injection of ADSCs with fat grafting has helped to improve retention of the grafted fat as well as significantly higher adipogenesis gene expression and vasularity.23,24 Additionally, SVF, which is the substance created after collagenase digestion of perivascularr adipose tissue and stroma, contains numerous types of progenitor stem cells including ADSCs, pericytes, endothelial progenitor cells, hematopoietic cells, and fibroblasts. 25 These cells within the SVF secrete growth factors and cytokines contributing to its regenerative properties through stimulating tissue growth and angiogenesis. 24 Cell-assisted lipotransfer is the use of SVF-enriched fat grafts, and studies using this technique have shown that there is improved fat retention of the grafted fat versus just fat grafting alone.22,2628 A unique form of SVF was studied in the form of a gel product that has been processed to have high concentrations of ADSCs and other SVF cells without the pro-inflammatory lipids, therefore, producing long-term higher volume retention and rejuvenation effects. 28


[IMG alt="An external file that holds a picture, illustration, etc.Object name is 10.1177_15353702211020701-fig1.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8381699/bin/10.1177_15353702211020701-fig1.jpg[/IMG]

Figure 1.
Partitioning and processing of procured fat allows enrichment of the injectate with autologous stem cells. The Celution System equipment allows separation of stem cells within an hour and administration of the stem cell bolus back into the prepared fat for administration during the same operative procedure, in a sterile closed system.
 
Defy Medical TRT clinic doctor

BadassBlues

Well-Known Member

Stem Cells Therapy for Facial Rejuvenation​


Traditionally, dermal fillers have been used to plump up volume in the facial area and create a more youthful appearance by lifting the cheeks and enhancing blood flow. Stem Cells for facial rejuvenation is one of the best method out there that you might want to consider.
Stem cell facelifts use the healing powers of stem cells harvested from the fat throughout the body and using it as a more natural alternative to dermal fillers. Many scientists and doctors believe that the adult stem cells from PRP and fat enhance tissue healing, lead to the growth of new blood vessels, and ultimately produce a more youthful glow of skin. For these reasons, doctors offer stem cell therapies and treatments to smooth out wrinkles, and improve the skin’s overall texture and tone.

What is Stem Cell Therapy?​

In order to understand why stem cells are used as a facial rejuvenation alternative, it is important to first understand what stem cells are and what their function is. Of the trillions of cells that make up the human body, stem cells are by far the most intriguing category. They are among the few cells in the body that are able to transform between cell types and have the potential to become a needed cell type and replace worn and damaged cells that they encounter.
A large number of stem cells exist in fat tissue. When stem cells are transplanted with fat in the different parts of their body, the majority of them grown and exist in these new areas helping to form new tissue at the transplanted site. By using the regenerative properties of stem cells along with transplanted fat, a marked improvement of these tissues occurs.
Stem cell facial rejuvenation therapy is a non-surgical facial rejuvenation procedure that harnesses the science of stem cells to make the body create more collagen and fill in tissues with healthy blood flow, resulting in a glowing, youthful appearance.

What is a Stem Cell Facelift?​

The stem cell facelift is a simple technique that takes cells from your fat tissues, blood, bone marrow and uses them to restore elasticity, smoothness, and softness of the skin.
This gives the face a more youthful and vibrant appearance. This treatment reduces the need for surgery or injection of temporary fillers like Restylane, Juvederm and hyaluronic acid.
It is a true and complete facial rejuvenation procedure that does not involve surgery, avoiding any open incision and help in the process of “lifting” the skin itself. The treatment helps to improve the contour and shape of the face, improve skin color and irregularities caused by normal aging, as well as improving marks caused by exposure to the sun and environment.

Who Can Have This Procedure Done?​

The stem cell facelift is ideal for those with early or mild signs of facial aging and want to rejuvenate their skin without going under the knife. The treatment is perfect for those who want their wrinkles more shallow, their cheekbones more defined, or if you just want to get rid of other early or moderate signs of aging.

What Results Can I Expect From This Procedure?​

Stem cells can provide many benefits to patients. They can raise the volume and structure of the face and achieve a more youthful appearance, and also aid in the repair and rejuvenation of the skin for up to 18 months. This is possible because stem cells have the amazing ability to recognize and repair damaged tissue.
Patients who undergo stem cell therapies for facial lifting claim that their skin is thicker and more radiant, and that the benefits last for up to nine months. This treatment can help repair lines that have developed, help fill in sunken cheeks, and also remove bags under the eyes.
The duration of the results of the procedure depends on the individual aging process the patient has already experienced. Other factors that affect the success of the procedure include poor eating habits, exposure to sunlight, smoking, and proper skin care.

Technique and Protocol​

The facial rejuvenation procedure with stem cells is very simple and can be performed under local anesthesia. It’s fast, mostly free of pain or discomfort, and does not require hospital visits.
During the process, liposuction is used on belly fat or other areas of fat to gently remove stem cells. Stem cells are also harvest from your blood and bone marrow. Stem cells are then isolated, and then injected into the face. The power of stem cells improves skin, tighten it and makes wrinkles disappear.
The procedure does not only involve injecting stem cells into specific areas of the face, chin or lips, but also allows the specialist to sculpt or remove fat from specific areas to get a facial contour specifically designed to fit the needs of each patient.
Stem cells therapies are an effective and natural approach to combating the effects of aging. Talk to our doctor today Biotech Wellness Center about possibly engaging in stem cell therapy to rejuvenate your skin.
 

BadassBlues

Well-Known Member

Applications of Mesenchymal Stem Cells in Skin Regeneration and Rejuvenation​

Hantae Jo,1,† Sofia Brito,1,† Byeong Mun Kwak,2,3,† Sangkyu Park,4,* Mi-Gi Lee,5,* and Bum-Ho Bin1,4,*
Sung-Chul Jung, Academic Editor
Author information Article notes Copyright and License information PMC Disclaimer

Associated Data​

Data Availability Statement
Go to:

Abstract​

Mesenchymal stem cells (MSCs) are multipotent stem cells derived from adult stem cells. Primary MSCs can be obtained from diverse sources, including bone marrow, adipose tissue, and umbilical cord blood. Recently, MSCs have been recognized as therapeutic agents for skin regeneration and rejuvenation. The skin can be damaged by wounds, caused by cutting or breaking of the tissue, and burns. Moreover, skin aging is a process that occurs naturally but can be worsened by environmental pollution, exposure to ultraviolet radiation, alcohol consumption, tobacco use, and undernourishment. MSCs have healing capacities that can be applied in damaged and aged skin. In skin regeneration, MSCs increase cell proliferation and neovascularization, and decrease inflammation in skin injury lesions. In skin rejuvenation, MSCs lead to production of collagen and elastic fibers, inhibition of metalloproteinase activation, and promote protection from ultraviolet radiation-induced senescence. In this review, we focus on how MSCs and MSC-derived molecules improve diseased and aged skin. Additionally, we emphasize that induced pluripotent stem cell (iPSC)-derived MSCs are potentially advanced MSCs, which are suitable for cell therapy.
Keywords: mesenchymal stem cells, skin regeneration, wound healing, skin rejuvenation, antiaging, induced pluripotent stem cell
Go to:

1. Mesenchymal Stem Cells​

Mesenchymal stem cells (MSCs) were observed for the first time in bone marrow by Cohnheim in 1867, who discovered that these cells could be the source of fibroblasts involved in wound repair [1]. Later, MSCs were first isolated and cultured in 1968 by A. J. Friedenstein. Using cells prevenient from murine bone marrow, Friedenstein observed that transplanting cell colonies to semi-syngeneic animals could originate fibrous tissue, bone and bone containing bone marrow. However, only years after, it became clear that the works made by Friedenstein were due to cells with multipotent ability. The term “mesenchymal stem cells” was presented by Caplan in 1991, after his studies with human bone marrow research [2,3]. Since then, owing to their easy isolation, expansion, and multipotentiality, MSCs have been rapidly popularized as a promising therapeutic agent for regenerative medicine. To date, it is a hot topic of research that is being explored for multiple purposes. The International Society for Cellular Therapy (ISCT) has suggested at least three conditions that can characterize MSCs. First, MSCs must adhere to a plastic culture vessel and grow. Second, MSCs should have CD73, CD90, and CD105 as cell surface antigens. Also, CD11b, CD14, CD19, CD34, CD45, CD79α, and HLA-DR antigens, which are hematopoietic stem cell antigens, should not exist on MSCs. Third, MSCs must be able to differentiate into osteoblasts, adipocytes, and chondrocytes in vitro [4]. After the discovery of bone marrow-derived MSCs (BM-MSCs), several other MSC sources have been reported, including endometrium [5], dental pulp tissues [6], skeletal muscles [7], placenta [8], adipose tissue [9], umbilical cord blood [10], and Wharton’s jelly [11] are sources of MSCs. MSCs are suitable for cell therapy because: (a) They have stemness potency; b) They are easy to isolate from original tissues; (c) They have less severe ethical issues as compared to embryonic stem cells (ESC); (d) Unlike induced pluripotent stem cells (iPSC), they carry a lower risk of teratoma-formation [12,13]; and (e) They are useful for a variety of therapeutic applications because of their ability to migrate to damaged tissue by chemoattraction [14]. Hence, it is possible to apply MSCs for the treatment of tissues of different origins [15,16,17,18,19].
The skin is continuously exposed to a variety of injuries. In dermatology, MSCs have demonstrated the potential for skin regeneration in many reported cases [20,21]. Additionally, due to the modern population’s increased esthetic standards, the interest in keeping a youthful appearance has also increased. Therefore, skin rejuvenation using MSCs is a treatment that attracts attention [22,23,24]. This review focuses on recent applications of MSCs and MSC-derived appendages in skin regeneration and rejuvenation.
Go to:

2. Skin Structure​

The various layers of the skin have distinct structures and functions that work together to protect internal organs and serve diverse biological functions. The skin is composed of three major layers: epidermis, dermis, and hypodermis (Figure 1).

[IMG alt="An external file that holds a picture, illustration, etc.Object name is ijms-22-02410-g001.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7957487/bin/ijms-22-02410-g001.jpg[/IMG]

Figure 1
Schematic representation of the human skin structure.
The epidermis, the outermost layer, plays a major defensive role [25]. This layer protects the skin from damage and stress, while also limiting the passage of water and chemical absorption [26]. It is constituted predominantly of keratinocytes, which are present in the epidermis in different maturation states, constituting around 95% of the layer [27]. These cells produce multiple keratins, which are major structural proteins that provide strength to the skin [28]. The epidermis is subdivided into five distinct strata: stratum corneum, stratum lucidum, stratum granulosum, stratum spinosum, and stratum basale [29]. Keratinocyte stem cells are located in the basal layer and gradually differentiate across the layers until they become terminally differentiated in the stratum corneum, being gradually replaced by keratinocytes prevenient from the bottom layers [30,31]. In the stratum spinosum, Langerhans cells are involved in an immune response, and protect the skin from microbial agents [32]. Additionally, Merkel cells function as sensorial receptors for stimuli, including pain, temperature, and touch [33]. Melanocytes are also present, producing melanin and transferring it to keratinocytes. Melanin provides pigment to the skin and hair, and also protects the skin from damage by ultraviolet (UV) radiation [34,35].
Between the epidermis and dermis, there is a cutaneous basement membrane zone, which connects basal keratinocytes with collagen fibers located on the surface of the dermis [36]. The main function of this structure is to provide adhesion between both layers, Since the epidermis is avascular, the basement membrane zone allows oxygen and nutrient exchange from the vascular dermis to the epidermis [37].
The dermis plays a crucial role in cushioning the body and providing structure. This layer is arranged as a mesh-like network consisting of connective tissue, blood vessels, lymph vessels, and mast cells [38]. Connective tissue is mainly formed by fibroblasts, which are responsible for the synthesis of elastin and collagen proteins [39]. Elastin proteins play a role in assuring elasticity and resilience to the skin. Collagen fibers are structural proteins that play important roles in stretching and providing tensile strength to the skin [40]. Mast cells are responsible for the inflammatory response of the skin to combat microorganisms, allergens, and physical injury [41].
The hypodermis is the deepest layer of the skin, being mainly composed of adipocytes. A rich vascular plexus extends from this layer to the dermis, supplying it with blood. Furthermore, the hypodermis makes the connection of the skin to muscles. Deep wounds that reach the hypodermis, causing its loss or damage, constitute complicated cases for wound healing [42,43].
Human skin and rat skin differ in histological, phenotypic, immunological and molecular domains. Therefore, given that rodents are models more accessible for investigation, we should consider their differences when studying wound healing. Firstly, even though the epidermis, dermis and hypodermis are structurally similar in humans and mice, their thicknesses are distinct. Human skin is usually 5 to 10 layers of epidermis and over 100 μm of thickness, but mice skin is 2 to 3 layers of epidermis and less than 25 μm thick. As a result, mice skin has a lower barrier function and increased absorption than humans skin [44]. Furthermore, male mice dermis is 40% thicker than the female mice dermis. Furthermore, contrarily to humans, mice skin possesses panniculus carnosus, a thin muscle layer that gives the skin contraction properties. This muscle is responsible for almost 90% of the wound of mice closure process [45]. On the other hand, human skin wound closure depends on reepithelization and granulation tissue formations In research, using mice as skin models should take these different factors into account for experimental design and result interpretation. Additionally, concerning wound healing, inflammatory reactions control the healing capacity. Humans and mice have different percentages of leukocytes, 10–25% neutrophils and 75–90% lymphocytes in mice, and 50–70% and 30–50% respectively, in humans [46]. Despite that, the effects of these differences in wound healing are not clear. In addition, human skin neutrophils express defensin, an antimicrobial peptide that aids in the case of infection, but mice skin does not. [45,47,48]. Comparing to mice, several other mammals are physiologically more close to humans [49]. For example, the structure of skin and wound healing mechanisms of pigs are similar to humans. However, pigs are not as well researched physiologically as mice, since the cost of maintaining pigs in a lab is higher, and surgical operation is more complicated [49]. Considering these limitations, mice are more widely used for skin research [49].
Go to:

3. Applications of Mesenchymal Stem Cells in Skin Regeneration​

3.1. Wound Healing​

Wounds can be divided into acute and chronic, depending on the time and progress of the healing process. Additionally, post-infection and post-inflammatory wounds are also significant problems. Thus, it is essential to develop technologies to aid against skin loss due to wounds.
Typically, skin wound healing comprises four overlapping phases: hemostasis (coagulation), inflammation (infiltration of mononuclear cells), proliferation (epithelization, fibroplasia, angiogenesis) and maturation (collagen deposit, formation of scaring tissue) [50]. MSCs aid in all phases of the wound healing process. Application of MSCs for skin therapy can enhance wound healing and curtail scarring. MSCs migrate to the spot of skin injury, inhibit inflammation, and elevate the proliferation and differentiation potential of fibroblasts, epidermal cells, and endothelial cells (Figure 2) [51,52]. Recent studies have reported that MSC-derived cultured media (MSCs-CM), extracellular matrix (ECM), exosome, platelet-rich plasma (PRP), and cytokines treat injuries in diverse tissue types (Table 1).

[IMG alt="An external file that holds a picture, illustration, etc.Object name is ijms-22-02410-g002.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7957487/bin/ijms-22-02410-g002.jpg[/IMG]

Figure 2
MSCs healing mechanism in skin regeneration and rejuvenation.

Table 1​

Applications of mesenchymal stem cells (MSCs) in wound healing.

Wound Healing Process​

Treatment to MSCs​

Function of MSCs​

Source of MSCs​

Model​

Reference​

Anti-inflammation​

-​

Polarization of macrophages to an M2 phenotype​

BM​

MSCs co-culture with macrophage​

[53]​

MSC-derived exosome​

Polarization of macrophages to an M2 phenotype​

BM​

MSCs co-culture with macrophage​

[54]​

TNF-α, IL-6​

Polarization of macrophages to an M2 phenotype​

Gingiva​

MSCs co-culture with macrophage​

[15]​

TSG-6​

Polarization of macrophages to an M2 phenotype​

BM​

Diabetic mice model​

[55]​

TNF-α​

Limiting macrophage activation​

BM​

Skin injury mice model​

[56]​

siTSG-6
(negative effect)​

Polarization of macrophages to an M2 phenotype​

cAD​

Inflammatory bowel disease mice model​

[57]​

Proliferation​

CXCR4 antagonist
(negative effect)​

Chemotaxis of MSCs​

BM​

Burn mice model​

[58]​

PRP​

Chemotaxis of MSCs​

AF​

Transwell migration assay​

[59]​

PRP​

Fibroblast migration​

AD​

Wound healing assay in culture dish​

[60]​

PRP​

Re-epithelialization​

AD​

Skin injury mice model​

[61]​

PRP​

Chemotaxis of MSCs​

BM​

Chemotaxis device​

[62]​

EMPB​

Migration of MSCs​

Endogenous MSCs in mice​

Diabetic mice model​

[63]​

Cinnamtannin B-1​

Migration of MSCs​

Endogenous MSCs in mice​

Diabetic mice model​

[64]​

Angiogenesis​

Low-level laser therapy​

VEGF, bFGF secretion in the wound bed​

cAD​

Skin injury mice model​

[65]​

-​

CCL2​

Primary MSCs in CCL2-KO mice​

Skin injury mice model​

[66]​

Negative pressure wound therapy​

CD31, VEGF, α-SMA​

BM​

Skin injury mice model​

[67]​

Biomimetic hydrogel scaffold​

Wound
vascularization​

BM​

Skin injury mice model​

[68]​

Increase in wound closure​

Self-adaptive all-in-one delivery chip​

Skin nerve regeneration​

BM​

Skin injury mice model​

[69]​

Chitin nanofiber-based hydrogel​

Granulation tissue formation​

BM​

Skin injury mice model​

[70]​

-​

Collagen type VII​

iPSC​

Skin injury mice model​

[71]​

CTGF​

Fibroblast differentiation​

ESCs​

Skin pressure ulcer mice model​

[72]​

ECM​

VEGF, PDGF, EGF​

UCB​

Diabetic rat model​

[73]​


Open in a separate window
BM: bone marrow; cAD: canine adipose tissue; AD: adipose tissue; iPSC: induced pluripotent stem cells; AF: amniotic fluid; UCB: Umbilical cord blood, VEGF: vascular endothelial growth factor; bFGF: basic fibroblast growth factor; CCL2: chemokine (C-C motif) ligand 2; TNF-α: tumor necrosis factor-α; TSG-6: tumor necrosis factor-α–stimulated gene/protein-6; siTSG-6; CTGF: connective tissue growth factor; PDGF: platelet-derived growth factor; EGF: epidermal growth factor; TSG-6 siRNA transfection; KO: knockout; PRP: Platelet-rich plasma; EMPB: ethanol extract from Mallotus philippinensis, a plant in the spurge family, bark.
The inflammatory phase is important for the wound healing process, as it leads to the recruitment of immune cells to reduce pathogens and clear the injury. However, chronic inflammation can postpone skin healing. MSCs can inhibit inflammatory responses in several ways. Chiossone et al. (2016) showed that MSCs promote polarization of macrophages to an M2-like phenotype, a type of macrophage that reduces inflammation and immunosuppressive function [53]. Moreover, the MSC-induced M2-like phenotype macrophages (MMSC) interact with natural killer (NK) cells and inhibit the expression of NK activation-related proteins such as NKp44, CD25, CD69, and interferon-gamma (IFN-γ). Furthermore, MMSC can inhibit T cell proliferation by promoting the multiplication of Tregs [53]. Luz-Crawford et al. (2016) provided evidence of the critical role of interleukin-1 receptor antagonist (IL1RA) secreted by MSCs in inducing MMSC and inhibiting B cell maturation in an IL1RA knock-in mouse model [74]. Interleukin-1 (IL-1) is known to accelerate T-helper 17 (Th17) cell differentiation [75]. Th17 cells express IL-17, which is a marker of inflammatory cytokines in many tissues. Therefore, IL1RA decreases the differentiation of Th17 cells and causes an increase in the anti-inflammatory effect of the cells. Zhao et al. (2013) revealed that IL1RA from BM-MSCs inhibits the production and activity of IL-1 and TNF-α, which are pro-inflammatory cytokines [76]. These studies indicate that MSCs have anti-inflammatory ability through modulation of macrophage polarization and expression of anti-inflammatory cytokines (Figure 2).
In the proliferative phase, MSCs manipulate macrophages to recruit keratinocytes and fibroblasts (Figure 2). Macrophages release epidermal growth factor (EGF) and transforming growth factor-α (TGF-α) to stimulate the migration and proliferation of keratinocytes [77]. Fibroblasts increase the migration and proliferation of keratinocytes via EGF, fibronectin, and keratinocyte growth factor (KGF) [77]. Keratinocytes also stimulate fibroblasts by expressing fibronectin, laminin 332, and tenascin [77]. Li et al. (2015) showed that high glucose and lipopolysaccharide inhibits the migration and proliferation of rat keratinocytes [78]. Furthermore, MSCs-CM can stimulate the migration and proliferation of keratinocytes [78]. Smith et al. (2010) revealed that BM-MSCs release soluble signaling factors that increase migration, proliferation, and chemotaxis of dermal fibroblasts [79]. MSCs can lead to angiogenesis at the site of the wound (Figure 2). Rustad et al. (2012) showed that MSCs-CM within hydrogels increased VEGF expression levels and resulted in faster wound healing than an injection of only MSCs into the wounded skin area [68]. Furthermore, Qiu et al. (2020) noted that MSCs educated by exposure to exosomes from neonatal mouse serum significantly improved wound healing [80]. Moreover, they found that the exosomes of educated MSCs significantly increased wound healing by inducing angiogenesis [80]. Martin-Piedra et al. (2019) used AD-MSCs, dental pulp-derived MSCs (DP-MSCs), Wharton’s jelly-derived MSCs, and BM-MSCs for epidermal regeneration, by tissue engineering and surgical grafting in animal models. The study illustrated that the partial epithelial differentiation ability of these cells could be used to generate bioengineered human skin substitutes for epidermal repair [81].
Furthermore, another important aspect involved in skin wound healing is the recovery of nerve function [69]. Skin wound healing aims to recover the protective ability of skin, and restore neuronal excitation functions through nerve regeneration. First, endogenous MSCs migrate toward the injury site because of chemoattractants. Stromal cell-derived factor-1 (SDF-1) is a well-investigated chemoattractant for the recruitment of MSCs. In the second stage, MSCs promote neuronal regeneration. bFGF, nerve growth factor (NGF), and brain-derived neurotrophic factor (BDNF) are important secretory factors that promote nerve regeneration (Figure 2) [82].
The ECM exists within all tissues and organs and contributes crucially to physical scaffolding for the cellular construction and initiation of signaling bioactive factors [83]. ECM is composed of proteins, polysaccharides and water, but each tissue has an unique composition and topology [83]. Collagen, a component of the ECM of skin, facilitates the migration of keratinocytes to reconstruct the damaged epidermis, as collagen-based materials also improve wound healing [84,85]. Zhou et al. (2019) showed that a combination of AD-MSCs and their ECM increases wound healing [86]. Platelet-rich plasma (PRP) is a rich source of cytokines and growth factors important for wound healing, including EGF, bFGF, HGF, PDGF, TGF-β1 and VEGF [87]. Recent studies have shown that PRP has an anti-inflammatory effect and regulates macrophages to increase wound healing [87]. Hersant et al. (2019) showed that a treatment combining PRP and MSCs improves mouse wound closure and proangiogenic properties in wound sites [88]. Holmes et al. (2018) studied a treatment mixture of leukocyte-high PRP and bone marrow concentrate to induce the recruitment BM-MSCs in the microfluidic device [62]. Moreover, Paganelli et al. (2019) used MSCs derived from adipose tissue to build a dermal substitute for wound healing, with high biocompatibility and good mechanical properties [89]. In addition, Zhang et al. (2018) revealed that AD-MSCs increase wound healing via their paracrine function [90]. They showed that AD-MSC-derived exosomes improve wound healing by regulating the proliferation and migration of fibroblasts, and optimizing collagen deposition [90]. Furthermore, AD-MSC-derived conditioned media increased the migration of skin fibroblasts and elevated wound healing in vivo.
These reports illustrate that MSCs and MSC-derived cytokines, exosomes, ECM, PRP, and CM have wound healing capacity. Various cell-derived MSCs and their derived molecules have applications in wound healing (Table 1). These applications can be subjected to clinical trials, and optimized treatment plans and patient types can be decided.

3.2. Burn Injury​

Burns are one of the main injuries worldwide [50]. Burn injuries are classified as first to third-degree burns (1~3°). The recovery depends on the severity of the burn, and two weeks are needed for recovery from superficial burning and minimal scarring. Severe burn injury includes third-degree (3°) burns and damage to the full thickness of the skin [91]. Angiogenesis is vital for the blood supply required to heal severe burn injuries [92].
Recently, many studies have reported that MSCs aid in healing burn injuries (Figure 2) [93,94,95,96]. MSCs increase wound closure and angiogenesis, and minimize scarring. BM-MSCs induced burn healing in a rat model by the expression of collagen 1 and integrin α2β1 [97]. In the same burn injury rat model, umbilical cord-derived MSCs (UC-MSCs) promoted burn healing through an immunosuppressive effect [98]. However, the healing mechanism of burn injuries by MSCs is not fully understood yet. Additionally, the attachment of transplanted MSCs to wounds is limited. The rate of engraftment of MSCs into organs is less than 3%, as reported in heart [99], kidney [100], liver [101] and pancreatic [102] injury models [20]. Because of this poor engraftment of MSCs, detailed studies are needed to increase the probability of engraftment of MSCs in damaged skin. There are two methods to inject MSCs into the body: Firstly, MSCs can be delivered into the tissue locally, by diverse scaffolds embedding MSCs. Several scaffolds methods have been developed to help in the transplant of MSC for tissue engineering clinical therapy, being composed of biodegradable, ceramic, matrix, synthetic, or alternative materials [103]. Secondly, MSCs can be injected by intracardiac, intramuscular, or intraperitoneal injections. It is also possible to inject via intravascular injection, either by arterial (IA) or venous (IV) injections. Relevantly, Krean et al. (2013) showed that MSCs injection by IA is more effectively spread than IV injections [104]. By IV injection into the tail vein, MSCs clearly capture in the lungs, however, when MSCs were delivered by IA injection through the aortic arch, the cells were more equally spread in the entire animal body [104]. These two methods for injecting MSCs into the body should be more developed for increasing the rate of engraftment of MSCs engagement into the skin.
Go to:

4. Applications of Mesenchymal Stem Cells in Skin Rejuvenation​

4.1. Antiaging​

Aged skin is highly associated with an unpleasing esthetic, which occurs due to loss of function and structural degeneration of the skin [105]. This can result in more serious complications, including more susceptibility to diseases such as eczema, dermatitis, autoimmune disorders, and melanoma [106]. With aging, the skin naturally loses its collagen content and elastic fibers become deranged [107]. Additionally, aged skin demonstrates an increase in oxidant activity [108], and an increase in the production of matrix metalloproteases (MMP), which are typically involved in matrix degradation. Additionally, exposure to UV light is known to promote premature aging of the skin, namely photoaging (Figure 2) [109]. Thus, rejuvenation therapies, which focus on the prevention and reversal of skin aging are in high demand in our society, which increasingly aims to maintain a youthful appearance and improve their health.
AD-MSCs have been gaining attention in skin antiaging therapy because of their efficient re-epithelization and secretion of several growth factors necessary for skin regeneration [24]. In recent years, Charles-de-Sá et al. (2015) observed the histological and structural modifications in aged facial skin after the injection of expanded AD-MSCs, collected from fat removed by liposuction [110]. Treatment with AD-MSCs caused an increase in elastic fibers in the superficial layer of the dermis and modified the collagen and reticular fiber networks, which became more arranged. Subsequently, AD-MSCs were observed to induce complete regeneration of solar elastosis in photoaged skin [111]. The transplantation of AD-MSCs leads to complete regeneration of dermal elastic matrix components, including oxytalan, elaunin, and elastin fibrillary networks. In solar-aged skin, the normal elastin matrix is usually lost, and AD-MSC-mediated treatment successfully reversed the inhibition of precursor molecules involved in neoelastinogenesis. This was observed by their high immunoreactivity, which indicated a high de novo formation. Additionally, the elastotic abnormal elastin deposits in the deeper dermal layers were degraded and replaced by typically polymerized elastic fiber networks. This was hypothesized to have been caused by the activation of cathepsin K, which allows reparative and hyperplastic processes after sun exposure.
Another way to use AD-MSCs in antiaging therapy, in a “cell-free” method of treatment, is by using extracellular vesicles (EVs), which have several advantages over stem cells and their safety issues. Adipose-derived mesenchymal stem cells extracellular vesicles (AD-MSCs-EVs) have anti-photoaging potential and were analyzed as subcutaneous injections in photoaged mice models [112]. The treatment resulted in a decrease in skin wrinkles and promotion of epidermal cell proliferation. Additionally, macrophage infiltration and reactive oxygen species (ROS) production were reduced, which inhibited MMP activation and collagen degradation (Figure 2). Moreover, in vitro analysis showed increased fibroblast activity and protection from UVB-induced senescence.
Amniotic membrane-derived mesenchymal stem cells (AM-MSCs) have also gained popularity as agents for improving photoaging, due to their abundance, easy acquisition, growth factors, and cytokines. A study conducted in 2019 by Prakoeswa et al. used AM-MSC-conditioned medium to treat photoaged human patients, and microneedling was used to enhance penetration of the medium [113]. Clinical photoaging (pore, wrinkle, spot polarization, spot UV, and skin tone) improved in the treatment groups, as observed by the surface skin analysis system, Janus. AM-MSCs were predicted to improve proliferation and migration of dermal fibroblasts and epidermal keratinocytes, and increase collagen synthesis.
Furthermore, BM-MSCs have recently been observed to have beneficial effects on skin aging. In a study conducted by Liu et al. in 2017, the effects of BM-MSCs on skin aging were analyzed on mice models subjected to D-galactose-induced aging [114]. D-galactose is a monosaccharide sugar that is known to cause mitochondrial dysfunction and oxidative stress in cells. Treatment with BM-MSCs resulted in reduced antioxidant activity, as observed by a reduced content of malondialdehyde (MDA), which is formed by the degradation of polyunsaturated lipids by ROS, causing peroxidative tissue damage. Furthermore, superoxide dismutase (SOD) activity increased, demonstrating an improved dismutation of superoxide radicals to hydrogen peroxide and oxygen. Finally, the glutathione-peroxidase (GSP-Px) content also increased, leading to a better reduction of hydrogen peroxide to water, thus preventing lipid peroxidation.
Human umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) are known for their rapid proliferation and immunomodulatory capacity, while also being easy to isolate, in contrast to typical adult MSCs (Figure 2). These cells have also been a target for antiaging studies on skin. Kim et al. (2018) found that a conditioned medium of UCB-MSCs contained several growth factors such as EGF, bFGF, TGF-β, PDGF, hepatocyte growth factor (HGF), collagen type 1, and a rejuvenation factor called growth differentiation factor 11 (GDF-11) (Figure 2) [115]. Furthermore, a cream based on UCB-MSC-conditioned medium was used in vivo, and its effects on dermal density and wrinkles in human patients were analyzed. After daily treatment for four weeks, evaluation with digital micromirror devices demonstrated that skin density improved by 2.46% and eye-end wrinkles decreased. Another approach for skin rejuvenation is the use of EVs derived from UC-MSCs [116]. Engineered EVs (eEVs) were obtained using ultrasonication, and showed functions similar to those of naturally secreted EVs. Comparative tests demonstrated that eEVs promoted fibroblast proliferation and migration in vitro. They also increased the expression of proteins involved in the maintenance of the extracellular matrix, such as collagen, elastin, and fibronectin, and inhibited the expression of MMP-1 and MMP-3.

4.2. Hair Loss​

Androgenic alopecia is a form of hair loss that can occur in both men and women. In men, this condition is also referred to as male-pattern baldness. Male pattern hair loss converts testosterone in hair follicle cells into a more potent metabolite, dihydrotestosterone (DHT). DHT binds to the androgen receptor in the hair follicle, thereby lowering the cyclic AMP (cAMP) concentration in the cell. It reduces sugar metabolism in hair follicles and suppresses energy supply to shorten the hair follicle growth period. As a consequence, the duration of the resting phase of the hair increases, and the hair follicle gradually becomes thinner and shorter [117,118].
The role of stem cells located in the hair follicle bulge is vital for hair regeneration, involving the Wnt/β-catenin cycle [119]. The dermal papilla is essential for hair growth and hair loss occurs when the dermal papilla is inhibited from secreting growth factors [120,121]. Huang et al. (2016) investigated the interactions of dermal papilla cells with AD-MSCs in increasing hair formation [122]. Another study showed that when human amniotic fluid-MSCs-CM (AF-MSCs-CM) were injected subcutaneously around a full-thickness wound in rats, wound healing was facilitated and hair regrowth was observed at the wound site [123].
Similarly, there are reports that BM-MSCs play a role in wound repair and improve hair regrowth [124]. Dong et al. (2014) reported that over-expression of Wnt1a by BM-MSCs-CM stimulated the induction ability of mouse dermal papilla cells. Thus, BM-MSCs promote progression of hair cycle and lead to hair regeneration [123]. Park et al. (2019) showed that overexpression of Nanog by AF-MSCs promotes activity of dermal papilla cells and increases hair follicle recycling [125]. Rajendran et al. (2017) reported that mouse BM-MSCs-EVs stimulate proliferation and migration of dermal papilla cells. Fluorescence monitoring confirmed the uptake of BM-MSCs-EVs in dermal papilla cells which lead to the anagen stage of hair growth in a mouse model [126]. These results reveal that MSCs and MSC-derived appendages could be candidates for hair regrowth stimuli and hair loss treatment (Figure 2).
Go to:

5. Induced Pluripotent Stem Cell-derived Mesenchymal Stem Cells​

Typically, MSCs are harvested from adult adipose tissue, bone marrow, or umbilical cord. However, MSCs can be obtained from only approximately one-third of the umbilical cord blood. Furthermore, only 0.001–0.01% of the bone marrow cells allow harvesting of MSCs. Also, only 0.05% of adipose tissue from a donor can be used as a source of MSCs [127,128]. For the clinical application of MSCs into the human body, MSCs cell needs 1–3 × 106 cells/kg body weight [129,130,131,132,133]. The methods for MSCs harvesting are considered painful and difficult procedures, and require patient permission [134]. Furthermore, as mentioned above, the limited number of cells obtained from tissues is an issue. Additionally, restrained in vitro proliferation capacity is another difficulty in obtaining uniform populations for clinical trials. Since iPSCs are generated by somatic cells previously obtained from a patient, iPSCs are an easier and more ethical approach, comparing to MSCs concerning biopsy [135].
Takahashi et al. (2007) investigated the generation of iPSCs from adult human dermal fibroblasts by Yamanaka factors (Oct3/4, Sox2, Klf4, and c-Myc) through retroviral transduction [136]. Recent studies have shown that human iPSC-derived MSCs (iMSCs) are capable of aiding in various diseases. In a study by Lian et al. (2010) [137], MSCs were generated from iPSCs, with features similar to human BM-MSCs in terms of marker expression and differentiation potential (Figure 3). First, iPSCs were generated from IMR90 fibroblast cells by transduction of Oct4, Nanog, Sox2, and Lin28 factors through lentivirus transduction. To differentiate human iPSCs into MSCs, the authors used a published clinical protocol. iPSCs were placed on a gelatin-coated dish containing Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% serum replacement (SR), 10 ng/mL bFGF, 10 ng/mL PDGF-AB, and 10 ng/mL EGF, to promote the proliferation of MSCs. After 1 week, differentiated CD24−CD105+ iPSCs were harvested by fluorescence-activated cell sorting (FACS). The CD24−CD105+ cells were cultured in DMEM with 10% fetal calf serum, 5 ng/mL bFGF, 10 ng/mL PDGF-AB, and 10 ng/mL EGF. Adult human BM-MSCs were used as a control for the comparable characteristics of MSCs [137]. By transplantation of iMSCs into mice with severe hindlimb ischemia, the symptoms were reduced due to muscle regeneration and angiogenesis induced by iMSCs. Additionally, tests revealed that iMSCs and BM-MSCs have different capabilities in attenuating severe hindlimb ischemia by muscle regeneration, angiogenesis, and paracrine factor secretion. This result indicates that iMSCs have a better therapeutic capacity than BM-MSCs [137]. Additionally, Xu et al. (2019) revealed that iMSCs have different functions and gene expression patterns than BM-MSCs [138]. It was observed that the expression of CD73, CD90, and CD105, markers of MSCs, was significantly higher in iMSCs than in BM-MSCs. Further, iMSCs showed an increased expression of both KDR and MSX2 mRNA, as compared to BM-MSCs. Furthermore, BM-MSCs had a relatively high PDGFRα mRNA expression. These results suggest that iMSCs are distinguished from primary MSCs, and that iMSCs can be used in treatment methods which are beyond the limitations of primary MSCs [138].

[IMG alt="An external file that holds a picture, illustration, etc.Object name is ijms-22-02410-g003.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7957487/bin/ijms-22-02410-g003.jpg[/IMG]

Figure 3
Methodology for obtention of induced pluripotent stem cell (iPSCs)-derived MSCs.
Many experiments were attempted to differentiate iPSCs into MSCs. For example, Villa-Diaz et al. (2012) demonstrated that iPSCs cultured on synthetic substrates differentiated into MSCs (Figure 3) [139]. iPSCs were placed on poly [2-(methacryloyloxy)ethyl dimethyl-(3-sulfopropyl)ammonium hydroxide (PMEDSAH)-coated plates with human-cell-conditioned medium (hCCM) supplemented with 4 ng/mL bFGF2. PMEDSAH-coated plates were preincubated with hCCM for at least 48 h at 37 °C in a 5% CO2 incubator. Embryoid bodies (EBs) were formed, and cultured in suspension for 7 d with hCCM. Almost 70 EBs were cultured on 0.1% gelatin-coated dishes in α-minimum essential medium (α-MEM) with 10% fetal bovine serum (FBS), 200 mM L-glutamine, and 10 mM non-essential amino acids solution (NEAA), to promote the differentiation of iPSCs into MSCs. EBs were cultured for 2 weeks until the cells had a homogeneous fibroblastic morphology in the culture dish. The results showed that iMSCs were successfully differentiated, and had functional capacity, especially of bone formation in vivo [139]. In another method, SB-431542, a TGF-β inhibitor, was used to promote the differentiation of iPSCs into MSCs. iMSCs generated after 10 days of SB-431542 treatment revealed characteristics of MSCs such as differentiation potential and immunophenotype [140]. Several methods are currently being tried to obtain functionally useful iMSCs.
Recent studies demonstrate that iMSCs immune modulation and teratoma formation properties compare with MSCs or iPSCs. Soontararak et al. (2018) demonstrated that iMSCs have equal healing potential as AD-MSCs in mouse inflammatory bowel disease models [141]. Fu et al. (2012) and Gao et al. (2017) show that iMSCs modulate T-cell and dendritic cell function similarly to AD-MSCs or BM-MSCs [142,143]. Chow et al. (2017) discovered that iPSCs could lead to the formation of teratomas after 20 days of subcutaneous injection into immune-deficient mice. However, iMSCs intravascularly injected into dog models did not lead to the formation of tumors [144]. Wei et al. (2012) found that iMSCs did not result in teratoma formation in SCID mice models [145]. These results indicate that iMSCs could be a useful clinical therapy by immune modulation and safe for tumor formation rather than iPSCs.
Recently, several studies have been using iMSCs for skin regeneration and skin rejuvenation. Nakayama et al. (2018) successfully differentiated keratinocyte-derived iPSCs (KC-iPSCs). This group received keratinocytes from patients with human recessive dystrophic epidermolysis bullosa (RDEB) and obtained KC-iPSC-derived MSCs (KC-iMSCs) [71]. KC-iMSCs were injected subcutaneously and intravenously into immunodeficient mice with skin injury. After transplantation, human collagen VII was found at the dermal-epidermal junction, indicating successful wound healing. In addition, Kim et al. (2018) revealed that exosomes secreted by iMSCs (iMSCs-exo) increase the proliferation of human keratinocytes and dermal fibroblasts [146]. Also, according to a study by Veraitch et al. (2017), iMSCs improved the properties of dermal papilla cells and contributed to increasing the hair-like structure morphology in an immunodeficient mouse model [147]. Furthermore, Spitzhorn et al. (2019) reported that human iMSCs express genes related to rejuvenation. In this study, iPSCs were used to obtained induced fetal femur-derived MSCs and adult BM-MSCs. Both types of iMSCs had common MSC cell surface markers and expressed rejuvenation-related genes such as CDKN1C, DNMT3B, GCNT2, INHBE, and POU5F1P1. These results suggest that iMSCs can acquire rejuvenation-related genes regardless of donor age and MSC source. The iMSCs concept avoids the shortcomings associated with the use of adult MSCs. Therefore, iMSCs may prove useful for future applications in various clinical settings [148].
Go to:

6. Embryonic Stem Cells-Derived Mesenchymal Stem Cells​

Evans et al. (1981) first discovered embryonic stem cells (ESCs), originated from the inner cell mass of mouse blastocysts and, Thomson et al. (1998), was the first to report studies with human ESCs. These cells have the capacity to differentiate into all three germ layers (mesoderm, endoderm, and ectoderm). ESCs are considered to be able to overcome the limitations of adult stems cells, however, for clinical application, ESCs have higher risk of tumorigenicity, comparing to iPSCs, and the possibility of immune rejection. The major limitation of the development of ESC-based clinical therapies is the sacrifice of an embryo [135], which constitutes a major ethical issue. If overcome properly, ESC-derived MSCs (eMSCs) based clinical trials could be considered for skin regeneration and rejuvenation medicine.
Barberi et al. (2005) reported the first example of differentiation of ESCs into MSCs [149] and, over the years, research with eMSCs has been expanding [150,151,152]. Hwang et al. (2008) reported that transplanted eMSCs into the knee joint cartilage defect area promoted cartilage repair [153]. Furthermore, Laurila et al. (2009) transplanted eMSCs and BM-MSCs into rat ischemic model and revealed that the eMSCs and BM-MSCs have a similar capacity to enhance angiogenesis and cell proliferation due to secretion of growth factors [154]. Clinical application for skin regeneration and rejuvenation by eMSCs is still scarce, but former publications indicate that eMSC can be applied to regenerative medicine [155,156]. Yoon et al. (2018) first published evidence that eMSCs promote wound healing in pressure ulcers. It was demonstrated that eMSCs increase wound closure, vessel formation and expression of collagen type I and III, fibronectin, and fibroblast-specific protein-1 (FSP-1) [72].
Go to:

7. Conclusions​

Novel studies on MSCs have demonstrated their potential in skin therapy. Transplantation of MSCs is considered a powerful tool for regeneration and rejuvenation of skin, as MSCs are a promising source of skin cells. Recent advances have revealed that MSCs have many benefits in treating the skin. For example, research on MSCs demonstrates efficacy in healing, especially due to the improvement of immune function by macrophage activation and cytokine production. Furthermore, MSCs have been shown to improve skin conditions by ameliorating antioxidant activity, promoting cell proliferation, and improving overall skin morphology. However, further studies focusing on the underlying molecular mechanisms are still necessary to guarantee the safe implementation of these methods. Despite all the advantages and benefits of MSC therapy, there are still obstacles such as their low frequency in tissues and the limited proliferative potential of MSCs derived from adult sources. Thus, we propose iMSCs as a promising target for skin therapy research. Despite being relatively new, this technology has demonstrated great potential in stem cell research, considering the high self-renewal capacity and differentiation ability of iMSCs.
 

BadassBlues

Well-Known Member

In modern medicine, advanced aging is treated as a disease itself.

"If I do a heart surgery or any procedure on the heart, the number one question is how old is the patient? Because that determines the risk," Schwarz said.

He noted that over the last 20 years, there has been a shift in the medicine paradigm, moving from reactive medicine to what is known as regenerative medicine.

Reactive medicine is when someone comes to the doctor because their body hurts. They may have had an accident and the physician reacts to that injury, inflammation, infection, etc.

Today, doctors can repair damage and regenerate damaged tissues through various treatments, such as stem cells, which are not FDA-approved for any disease outside of certain forms of cancer.


No disease has ever been cured with stem cells. However, Schwarz noted that their use has seen an improvement in symptoms and the quality of life.
"The goal is to create an improved health and longer health span, not of the lifespan. So, we want people to be able to be active until high ages,"
Schwarz said.

Schwarz, who authored the book "The Secrets of Immortality," often speaks about how important it is not to believe that there is one injection or one pill that a person can take to solve their problems. Instead, what is needed is a whole lifestyle modification change, which includes diet, exercise, the discovery of nutrient deficiencies and regenerative medicine.
 

BadassBlues

Well-Known Member

Platelet-rich plasma: a narrative review​

Thomas Collins,1 Dinesh Alexander,2 and Bilal Barkatali2
Author information Copyright and License information PMC Disclaimer


Go to:

Abstract​

  • The aim of this article was to synopsize platelet-rich plasma (PRP) use in musculoskeletal pathologies through evidence-based assessment of the preparation, classification, mechanism of action and applications of PRP, thereby answering which PRP type is best for each clinical indication.
  • The literature search was performed using Medline, EMBASE and Cochrane Reviews databases for papers containing the key terms “platelet-rich plasma” AND “orthopaedics” AND (“classification” OR “mechanism of action” OR “preparation” OR “clinical application”). Generated papers were evaluated for pertinence in following areas: preparation, classification, mechanism of action, clinical application within orthopaedics. Non-English papers were excluded. Included studies were evaluated for quality.
  • Sixty studies were included in our review. There are many commercial PRP preparation kits with differing component concentrations. There is no consensus on optimal component concentrations. Multiple PRP classifications exist but none have been validated. Platelet-rich plasma acts via growth factors (GFs) released from α-granules within platelets. Growth factors have been shown to be beneficial in healing. Grossly elevated concentrations of GFs may have inhibitory effects on healing. Multiple systematic reviews show efficacy of PRP in tendinopathies, early osteoarthritis, acute muscle injuries and in combination with rotator cuff repair and anterior cruciate ligament reconstruction.
  • The literature suggests leukocyte-rich PRP (L-PRP) is more beneficial in tendinopathies and pure PRP (P-PRP) is more beneficial in cartilage pathology. However, different PRP preparations have not been directly compared in any pathology. Classification of PRP type is frequently not stated in research. Standardization of PRP research parameters is needed to streamline findings and generate clear indications for PRP types to yield maximum clinical benefit.

Cite this article: EFORT Open Rev 2021;6:225-235. DOI: 10.1302/2058-5241.6.200017
Keywords: orthopaedics, osteoarthritis, platelet-rich plasma, soft tissue, sports and exercise medicine
Go to:

Introduction​

Arthritis and soft tissue pathology make up the majority of orthopaedic referrals. An increasing proportion of patients are developing these pathologies at an earlier age, thereby producing a growing societal cost on healthcare and reduced productivity.14 This has led the drive to find treatments that can delay, or preferably cure, diagnoses that would otherwise require surgical intervention at an age or time when it would not typically be undertaken.
Platelet-rich plasma (PRP) is an autologous blood product acquired from part of the plasma fraction created via centrifugation of whole blood. By definition it has a platelet concentration above that of normal physiological levels.5
The term PRP originated in the 1970s by haematologists describing plasma with a platelet count higher than peripheral blood,6 which at the time was being used as a transfusion product in thrombocytopaenic patients.5 Since then it has been applied in multiple fields including plastic surgery, paediatric surgery, cardiac surgery, gynaecology, urology and ophthalmology.7 However, it is within the musculoskeletal field where there has been a surge of PRP use for multiple pathologies, largely due to widespread commercial interest following PRP use in professional sport.8
This review aims to synopsise PRP use in musculoskeletal pathologies through evidence-based assessment of the preparation, classification systems, mechanism of action and clinical applications of PRP and thereby answer, ‘What type of PRP is best for different clinical indications?’ We attempt to interpret the current viability of PRP within orthopaedics to help direct the focus of future research.
Go to:

Methods​

A literature search was performed using the Medline, EMBASE and Cochrane Reviews databases for all papers published between 1978 and 2019 containing the following key terms: “platelet-rich plasma” AND “orthopaedics” AND (“classification” OR “mechanism of action” OR “preparation” OR “clinical application”).

Selection criteria​

Yielded papers were evaluated independently by two authors and selected if containing pertinence to PRP regarding one or more of the following areas: preparation, classification, mechanism of action, or clinical application within trauma and orthopaedics. Papers not written in English and animal studies relating to applications were excluded. The authors acknowledge an element of language bias; however, using more modern publications limits the extent of exclusion.

Literature grading and analysis​

Studies were independently rated by two authors using the Oxford Centre for Evidence-based Medicine ‘Levels of evidence’ document9 and the Coleman modified score (CMS) when applicable.
Go to:

Results​

Sixty studies5,1068 were included in our analysis published between 1978 and 2019. Details of the literature search and data extraction can be seen in a flow diagram (Fig. 1).
[IMG alt="An external file that holds a picture, illustration, etc.Object name is eor-6-225-g001.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8142058/bin/eor-6-225-g001.jpg[/IMG]
Fig. 1
Study flow diagram.

Preparations​

Platelet-rich plasma can be formulated in multiple ways with no consensus on a definitive protocol that could be used internationally to standardize the formulation. The following section is based on level 5 evidence surrounding the preparation of PRP. The basis of the preparations relies on the concept of differential centrifugation.10 Each component of whole blood has a different specific gravity and, when spun in a centrifuge, separates into distinct layers.
There are two principle methods of producing PRP, the PRP method and the buffy-coat method.10 The PRP method uses fresh blood from venepuncture, which is placed in a centrifuge for a ‘soft’ spin to separate the red blood cells (RBCs). The supernatant plasma is then centrifuged in a ‘hard’ spin at higher speeds to obtain the platelet concentrate. The buffy-coat technique utilizes whole blood, pre-stored at room temperature (i.e. 20–24oC). It undergoes a ‘hard’ spin to separate it into three layers: RBCs, platelets and white blood cells (WBCs), and platelet-poor plasma. The supernatant plasma is removed, and the buffy-coat is separated. This layer undergoes a second low-speed spin to separate the WBCs, or a leukocyte filter can be used.
Centrifugation rate has proved important in determining the optimal platelet yield. Most PRP production involves two spins – for separation and then concentration; the main factors of these spins are the speed in rotations per minute (RPM) and duration. Sabarish et al11 studied three differing protocols involving spin rates from 1000–3600 RPM lasting 4 to 15 minutes respectively. They found that lower spin rates had higher platelet yields, hypothesizing that high rates could cause platelet clumping or disintegration.
Lansdown et al12 highlighted that patient factors also play a role in platelet concentration. Fasting patients had lower concentrations than those who ate a high-fat meal. The timings of venepuncture can also affect concentration, with the optimal time being in the afternoon. They also surmise from other literature that there appears to be an optimal platelet concentration. If it is too low, i.e. 0.5–1.5x whole blood concentration, there is no enhancement in bone regeneration. If too high, i.e. 6–11x whole blood concentration, then there is a paradoxical inhibitory effect on bone regeneration.
Arora et al14 reviewed some of the technical aspects in relation to PRP preparation. They stressed the importance of anticoagulants in preventing the coagulation cascade of collected blood. Ethylenediaminetetraacetic acid (EDTA) suppresses platelet degranulation and therefore is not recommended for PRP. Conversely, heparin can cause spontaneous aggregation of platelets in some individuals. Anticoagulant citrate dextrose-A (ACD-A) is the most commonly used in commercial kits. It maintains the optimal pH for platelets at 7.2 while the citrate binds to calcium preventing the coagulation cascade. They also recommend PRP be kept in small diameter tubes with caps (to minimize surface area to atmosphere) as the pH can increase by diffusion of CO2 out of the plasma, potentially causing spontaneous aggregation.
Etulain et al15 proposed an optimized protocol for PRP preparation. Their three-pronged approach centred on dilution, 4oC incubation and plasma cryoprecipitate supplementation. The combination of these had an additive effect with greater angiogenesis, greater secretion of vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), interleukin-17 (IL-17) and interleukin-8 (IL-8) which translated into faster and more efficient mouse-skin wound repair vs. non-optimized PRP.
Preparation of PRP is vitally important, as it will have a direct impact on the final composition of PRP. Degen et al16 looked into the compositional differences of five commercially available PRP systems. They found that platelet concentration and capture efficiency was similar amongst all systems, WBC concentration was significantly elevated in all systems compared to whole blood, and variation was seen with pH, RBC and neutrophil levels. The exact implications of differing compositional elements are still unknown and will be considered later in the article. What is clear is the heterogeneity amongst differing commercial PRP systems (Table 1). It is therefore inferable that this heterogeneity limits conclusions drawn from pooled PRP studies containing multiple PRP preparation systems.

Table 1.​

Component and preparation profile of commercial platelet-rich plasma (PRP) systems

Arthrex ACP Double Syringe (Arthrex, USA)​

Arthrex Angel System (Arthrex, USA)​

RegenKit A-PRP (RegenLab, Switzerland)​

MyCells (UK)/ Tropocells (UK)/ Cellenis PRP (Estar Medical, Israel)​

PRGF / Endoret (BTI, Spain)​

Glo PRP (Glofinn, Finland)​

PRP type

Plasma-based

Buffy-coat

Variant

Variant

Plasma-based

Variant

Starting volume

15 ml

40–180 ml

8 ml

10 ml

9 ml

9 ml

Platelets

2–3x (~2.5x)

Up to 18x

1.6x

2–5x (4.5x in 2ml)

2x

4–9x

WBCs

Reduction

Adjustable

Reduction

Reduction

Reduction

Increase

RBCs

Adjustable

Adjustable

Reduction

Reduction

Adjustable

No reduction possibility

PRP yield

4–6 ml

2–20 ml depending on composition

4 ml

2–3 ml

2 ml

Adjustable

Closed system

Yes

Yes

No

No

No

No

Needles involved

No

No

Yes

Yes

Yes

Yes

Principle

Centrifugation – closed transfer of PRP

Centrifugation with sensor/valve technology (light absorption) – PRP automatically collected in syringe

Separation gel – open needle transfer of PRP

Separation gel

Manual

Manual

Separation gel

No

No

Yes

Yes

No

No

Anticoagulant

No

Yes

Yes

Yes

Yes

Yes

Centrifugation steps

One

One

One

One

One

Two

Spinning parameters

1500 rpm / 5 min

Depending on program, 3000 rpm or 3500 rpm, 15–30 min

3400 rpm / 5min

1500 g / 10 min

580 / 8 min (+20 min clotting time)

1200 g / 5 min 1200 g / 10 min

Preparation time

10 min

25–40 min

10 min

25 min

30 min

25 min

Handling steps

⩽ 5

5–8

⩽ 5

> 10

5–8

8–10

Centrifuge

Specific

Specific

Specific

Specific

Specific

Specific


Open in a separate window

Ortho.pras (Proteal, Spain)​

Genesis CS (EmCyte, USA)​

PurePRP II (EmCyte, USA)​

Y-PRP (Ycellbio Medical, South Korea)​

Dr. PRP (SDD Medical Group, UK)​

PRP type

Variant

Buffy-coat

Buffy-coat

Buffy-coat

Buffy-coat

Starting volume

20 / 40 ml

30 / 60 ml

60 / 120 ml

15 ml

20 ml

Platelets

2.2x (in 4 ml PRP)

?

8x

7–9x

?

WBCs

Adjustable

Increase

Adjustable

Increase

Adjustable

RBCs

Adjustable

No reduction possibility

Adjustable

Reduction

Adjustable

PRP yield

4–10 ml

3–4 / 7 ml

7 / 14 ml

1–2 ml

5 ml

Closed system

No

Yes

Yes

No

No

Needles involved

No

No

No

Yes

Yes

Principle

Manual

Manual

Manual, PRP transferred to a separate container after first centrifugation

Manual

Manual, after first centrifugation plasma and RBC container are separated

Separation gel

No

No

No

No

No

Anticoagulant

Yes

Yes

Yes

Yes

Yes

Centrifugation steps

One

One

Two

One

Two

Spinning parameters

1800 rpm / 8 min

4400 rpm / 5 min

3800 rpm / 1.5 min; 3800 rpm / 5 min

3200–3600 rpm / 4 min

3400 rpm / 4 min; 3500 rpm / 2 min

Preparation time

20 min

15 min

20 min

20 min

20 min

Handling steps

8–10

5–8

5–8

5–8

> 10

Centrifuge

Specific

Specific

Specific

Specific

Specific


Open in a separate window

SW-PRP (Seawon meditech, South Korea)​

Biomet GPS (Zimmer Biomet, USA)​

Harvest SmartPReP (Terumo, USA)​

CPunT (Eltek Group, Italy)​

Magellan (Arteriocyte Medical Systems, USA)​

PRP type

Buffy-coat

Buffy-coat

Buffy-coat

Buffy-coat

Buffy-coat

Starting volume

25 ml

30 / 60 ml

20 / 60 ml

50 ml

30–160 ml

Platelets

?

9.3x

4.3–6.6x

4–5x

?

WBCs

Increase

Increase

Increase

Adjustable

Adjustable

RBCs

No reduction possibility

No reduction possibility

No reduction possibility

Adjustable

Adjustable

PRP yield

2 ml

3 / 6 ml

3 / 7 / 10 ml

10 ml

3–10 ml

Closed system

No

No

No

Yes

Yes

Needles involved

No

No

Yes

Yes

No

Principle

Manual, after first centrifugation plasma and RBC container are separated

Dual buoy system – extraction of PRP through separate luer port

Two chamber bucket + floating shelf – open needle transfer of PPP and PRP

Centrifugation – first separation automated in electromechanical device, second separation manual

Centrifugation with sensor/valve technology (light absorption) – PRP automatically collected in syringe

Separation gel

No

No

No

No

No

Anticoagulant

Yes

Yes

Yes

Yes

Yes

Centrifugation steps

Two

One

One

Two

One

Spinning parameters

3850 rpm / 7 min; 3850 rpm / 8 min

3200 rpm / 15 min

1000 g / 14 min

1200 rpm / 10 min; 1900 rpm / 10 min

Depending on programme, 12–17 min

Preparation time

40 min

30 min

20 min

30 min

25–30 min

Handling steps

> 10

5–8

8–10

> 10

5–8

Centrifuge

Specific

Specific

Specific

Specific

Specific


Open in a separate window
Note. ACP, autologous conditioned plasma; PRGF, plasma rich in growth factors; BTI, biotechnology institute; WBC, white blood cell; RBC, red blood cell. GPS, gravitational platelet system.
At a commercial level, there is a vast array of kits available to prepare PRP with varying concentration of platelet yields and no overall consensus on optimal component concentrations.17 A systematic review by Chahla et al18 found that, of 105 studies using PRP in orthopaedics, only 11 provided comprehensive reporting of the preparation protocol that could be used by subsequent investigators. Only 17 studies provided qualitative metrics on the composition of their PRP products. Greater transparency in the reporting of PRP used in clinical and laboratory studies is needed. This must be in conjunction with a unified classification system with international consensus. This must be the direction of future research if the full potential of PRP is to be realized.

Classifications​

Over the past decade, PRP use has grown significantly with numerous formulations currently available. Several authors have attempted to classify these preparations in order to give the orthopaedic community the means of comparing formulations, to find the optimal preparations for specific pathologies. The first such classification was proposed in 2009 by Ehrenfest et al19 (Table 2) who divided preparations by the presence of cell content and fibrin architecture. This qualitative classification gave a starting point but did not take into consideration other subpopulations of cells such as RBCs or neutrophils, which have an important role in the mechanism of action of PRP.

Table 2.​

Ehrenfest classification

Pure platelet-rich plasma (P-PRP)

Leukocyte-poor, low-density fibrin network

Leukocyte and platelet-rich plasma (L-PRP)

Contains leukocytes and low-density fibrin network

Pure platelet-rich fibrin (P-PRF)

Without leukocytes and high-density fibrin network

Leukocyte and platelet-rich fibrin (L-PRF)

Contains leukocytes and high-density fibrin network


Open in a separate window
DeLong et al20 developed on this classification and introduced a quantitative element. The PAW classification (Platelets, Activation, White blood cells) (Table 3) provides a nomenclature based on platelet concentration, activation and WBC count, including the neutrophil subgroup.

Table 3.​

PAW (Platelets, Activation, White blood cells) classification

Platelets

Concentration (/µL)

⩽ baseline
> baseline – 750,000
> 750,000 – 1,250,000
> 1,250,000

P1
P2
P3
P4

Activation

Exogenous

 

X

White blood cells (WBCs)

Total WBCs
Neutrophils

Above baseline
⩽ baseline
Above baseline
⩽ baseline

A
B
α
β


Open in a separate window
Mautner et al,21 however, argued that the PAW system did not address the effects of the RBC content on PRP preparations and recommended the PLRA classification (Platelet count, Leukocyte content, RBC content, Activation) (Table 4). This system was the first to recommend documentation of the volume of PRP used and the absolute platelet concentration. The authors also suggested the frequency of PRP treatments be recorded if multiple treatments were delivered.

Table 4​

PLRA (Platelet count, Leukocyte content, RBC content, Activation) classification

Criteria​

Final Score​

P Platelet count

____P
Volume Injected

_____M
Cells/µL

L Leucocyte content*

> 1%
< 1%

+

R Red blood cell content

> 1%
< 1%

+

A Activation**

Yes
No

+


Open in a separate window
*If white blood cells are present (+), percentage of neutrophils should be reported.
**The method of exogenous activation should be reported.
Magalon et al22 proposed a classification system (Table 5) focused on the quality of the preparation. The DEPA classification (Dose, Efficiency, Purity, Activation) analyses aspects of the production process that were not previously taken into consideration. However, it does not analyse the content of the PRP based on different cell types to the same quantitative level as the PLRA classification.

Table 5.​

DEPA (Dose, Efficiency, Purity, Activation) classification

Subgroup​

Description​

Dose of injected platelets

Very high
High
Medium
Low

> 5 Billion injected platelets
3–5 Billion
1–3 Billion
< 1 Billion

Efficiency of production

High
Medium
Low
Poor

Recovery rate in platelets > 90%
70–90%
30–70%
< 30%

Purity of PRP

Very pure
Pure
Heterogeneous
Whole-blood

Platelets in PRP > 90%
70–90%
30–70%
< 30%

Activation process

Autologous thrombin
Calcium chloride

 

Open in a separate window
The latest PRP classification system is MARSPILL (Method, Activation, RBCs, Spin, Platelet concentration, Image guided, Leukocyte concentration, Light activation), developed by Lana et al23 (Table 6). This is an amalgamation of previous systems, incorporating aspects of the manufacturing process as well as analysing the subgroups of cell types. They believe the focus on peripheral blood mononuclear cells is as important as platelet concentration, and they also incorporate newer concepts such as light activation and the use of image guidance to provide more elements to the classification system.

Table 6.​

MARSPILL classification

M

Method

Handmade
Machine

H
M

A

Activation

Activated
Non-activated

A+
A–

R

Red blood cells

Rich
Poor

RBC-R
RBC-P

S

Spin

One spin
Two spins

Sp1
Sp2

P

Platelet concentration

 

PL 2–3
PL 4–6
PL 6–8
PL 8–10

I

Image guided

Guided
Not-guided

G+
G–

L

Leukocyte concentration

Rich
Poor

Lc-R
Lc-P

L

Light activation

Activated
Not-activated

A+
A–


Open in a separate window
Numerous authors have developed classification systems for PRP, with each new generation taking into account additional factors that their predecessors had not considered. Due to the rapidly evolving nature of this field and the increased complexity of each preparation, there has not been widespread uptake of a single classification. In turn, none of the aforementioned classifications have been validated at an international consensus level. Barriers to widespread use of these classifications include the problem of oversimplification, such that researchers are wary of the earlier classification systems, which may classify their formulation as equal to other products that have not had favourable outcomes. Alternatively, if the trend of classifications becomes ever more complex, it may pose financial barriers to smaller research groups who may not have the resources to analyse their preparations to the same standard as large-scale pharmaceutical corporations.13

Mechanism of action​

Platelets are anucleated cytoplasmic fragments of megakaryocytes that differentiate down the myeloid cell lineage.24 They contain α-granules, often thought of as the storage units of platelets,25 which studies suggest contain an abundance of growth factors (GFs). These are believed to influence inflammation, angiogenesis, stem cell migration and cell proliferation.5 Platelets are well known to be the initiators of the healing process; however, not all tissues have a rich blood supply, for example tendons, ligaments and cartilage. This results in relatively low levels of GFs being available to these tissues to enact effective healing. Application of PRP to these, and other, areas can therefore introduce supra-physiological levels of GFs to theoretically stimulate resolution of chronic pathological processes. Commercial ELISA (Vector Laboratories, Burlingame, CA; Quantikine Immunoassay, R&D Systems, Minneapolis, Minnesota) and Luminex kits (Luminex Corporation, Austin, Texas) were used to accurately quantify GFs in software based statistical analysis in the following section.
Once recruited to an area of injury, platelet adhesion is facilitated through adhesive glycoproteins secreted by α-granules,26 including vitronectin, fibronectin, thrombospondin and von Willebrand factor.27,28 Once the clot is formed the platelets are activated,29 allowing the release of the GFs from α-granules to stimulate healing.
There are myriad GFs contained within α-granules, of which the complex interchange amongst them is hypothesized to be of additional benefit to the healing process beyond simply introducing a higher concentration of platelets at hypovascular sites.24
Growth factors enact their functions primarily via ligand binding to associated extracellular cell surface receptors, which signal intracellular cytoplasmic proteins to attach to phosphorylated tyrosine. This is followed by multiple phosphorylation and activation steps of protein kinases within the cytoplasm, finally leading to translocation of a phosphorylated kinase to the cell nucleus. This phosphorylates transcription factors enabling gene transcription and ultimately the execution of the encoded function.30,31
Growth factors contained within α-granules thought to be crucial to the efficacy of PRP include platelet-derived growth factor (PDGF), VEGF, the transforming growth factor-β superfamily (TGF-β), fibroblast growth factor (FGF) and insulin-like growth factor (IGF). PDGF is able to initiate callus formation via chemotaxis and mitogenesis of fibroblasts and chondrocytes,32,33 along with chemotaxis of mesenchymal stem cells (MSCs).34 The promotion of endothelial cell proliferation by PDGF also has an important role in angiogenesis.35 VEGF is involved in neovascularization through its strong endothelial chemokine and mitogenic properties.36 TGF-β is well established as a promoter of chondrogenesis,37 but has also been shown to: stimulate osteogenic MSC differentiation38 and undifferentiated mesenchymal cell proliferation; regulate the mitogenic effects of other GFs; and inhibit macrophage and lymphocyte proliferation.39 The FGF family is involved in multiple biological processes including osteoblastogenesis,38,40 growth and differentiation of chondrocytes and MSCs.39 IGF regulates the proliferation and maturation of chondrocytes41,42 and IGF-1 may down-regulate expression of programmed cell death 5 (PDCD5), thereby inhibiting apoptosis of osteoarthritic chondrocytes.43
In addition to GF release following platelet activation, Xie et al44 demonstrated that PRP also forms a fibrin gel, which acts as a conductive bioscaffold to allow incorporation of migrating cells for tendon healing. Entrapment of GFs within a fibrin matrix45,46 may hold the key to controlled release of GFs at the intended site of action. However, it is important to note that cellular response to GFs is limited by number of target receptors available on cell surfaces, therefore high platelet concentrations and subsequent GF release may not be of benefit.26 This may explain why PRP preparations with GFs over six times the physiological concentration may have an inhibitory effect.47
This leads on to an important point, that while there are many GFs that have been shown to have beneficial effects on cartilage, tendons, bone and other tissues, there are other components that can have negative effects such as pro-inflammatory cytokines, matrix metalloproteinases (MMPs) and interleukin-1β (IL-1β).48 For example, Browning et al49 demonstrated an increase in MMP-1 and MMP-3 in osteoarthritis (OA) synoviocytes incubated with PRP. Thereby suggesting PRP application to joints may lead to accelerated cartilage breakdown due to a pro-inflammatory response. Most in vitro studies support PRP use in cartilage tissue because of the ability to increase chondrocyte proliferation and production of matrix molecules whilst not affecting chondrogenic phenotype.50 However, the importance of platelet-derived GF dosage has also been highlighted through the different results they can produce.51
Perhaps the biggest area of controversy surrounding PRP is the concentration of cellular components, particularly leucocytes. There has been debate around whether leucocytes are adverse because of cytokines causing inflammation and subsequent weaker fibrotic tissue and/or proteases and reactive oxygen species they release,50 or beneficial as a result of cytokines that can prevent infection and improve healing.16 This is something we will explore in the following section.

Applications​

The ubiquitous nature of the mechanism of action of PRP suggests that, in theory, it can be applied to multiple pathologies to aid the body’s natural healing processes. We will look at these pathologies in detail and the type of PRP used (see Table 7). Unless stated, all the evidence included in this section is either level 1 (systematic review of randomized controlled trials [RCTs] or individual RCT) or level 2 (systematic review of cohort studies and RCTs). The Coleman Modified Scores given are the average of the papers analysed.

Table 7.​

Summarized platelet-rich plasma (PRP) evidence by indication and PRP type

Indication​

Findings​

PRP type studied

Osteoarthritis

Laver et al
Improved symptoms with PRP at 12-month follow up. Significantly improved in 7 of the 9 RCTs included.
Trend towards improved outcomes in younger patients/early OA changes with PRP.
Chang et al
Improved functional outcomes with PRP up to a year post treatment.
Less severe OA showed more benefit from PRP.

[IMG alt="An external file that holds a picture, illustration, etc.Object name is eor-6-225-i001.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8142058/bin/eor-6-225-i001.jpg[/IMG]
[IMG alt="An external file that holds a picture, illustration, etc.Object name is eor-6-225-i002.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8142058/bin/eor-6-225-i002.jpg[/IMG]

Lateral epicondylitis

Ben-Nafa and Munro
Improved outcomes for up to 2 years post treatment with PRP compared to corticosteroid injection.

[IMG alt="An external file that holds a picture, illustration, etc.Object name is eor-6-225-i003.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8142058/bin/eor-6-225-i003.jpg[/IMG]

Achilles tendinopathy

Gholami et al
No clinical benefit shown between PRP and saline placebo injection or eccentric loading programme.

[IMG alt="An external file that holds a picture, illustration, etc.Object name is eor-6-225-i004.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8142058/bin/eor-6-225-i004.jpg[/IMG]

Patella tendinosis

Dupley and Charalambous
Statistically significant improvement in functional scores at 6 months post treatment with PRP compared to dry needling or extracorporeal shockwave therapy (ESWT).

[IMG alt="An external file that holds a picture, illustration, etc.Object name is eor-6-225-i005.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8142058/bin/eor-6-225-i005.jpg[/IMG]

Rotator cuff disease

Kesikburun et al
No difference demonstrated between PRP and saline at any follow up point (followed up for 1 year) for rotator cuff tendinopathy or partial tears.
Rha et al
Significant functional improvement and greater symptomatic relief at 6 weeks to 6 months post treatment with PRP compared to dry needling for partial tears.

[IMG alt="An external file that holds a picture, illustration, etc.Object name is eor-6-225-i006.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8142058/bin/eor-6-225-i006.jpg[/IMG]
[IMG alt="An external file that holds a picture, illustration, etc.Object name is eor-6-225-i007.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8142058/bin/eor-6-225-i007.jpg[/IMG]

Acute muscle injury

Grassi et al
Meta-analysis demonstrated statistically significant reduction in return to sport time (7.17 days) with PRP compared to controls (none/haematoma evacuation/saline injection/platelet-poor plasma (PPP) injection).
Subgroup analysis of only double-blinded RCTs (both using P-PRP) showed no difference between PRP and controls (haematoma evacuation/saline injection).
Subgroup analysis of hamstring injuries (2 using L-PRP, 1 using P-PRP) showed no difference between PRP and controls (none/saline injection/PPP injection).

[IMG alt="An external file that holds a picture, illustration, etc.Object name is eor-6-225-i008.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8142058/bin/eor-6-225-i008.jpg[/IMG]

Surgical augmentation:
rotator cuff repair

Cohn et al
Of the 5 studies included, 1 showed less pain in the early post-operative period and increased strength of external rotation at 3 months post op with L-PRP + surgery. Another study showed a 20% reduction in re-rupture rate and significant improvement in shoulder function post op in PRP + surgery (PRP type unknown). The other 3 studies showed no significant differences with the addition of PRP.

[IMG alt="An external file that holds a picture, illustration, etc.Object name is eor-6-225-i009.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8142058/bin/eor-6-225-i009.jpg[/IMG]

Surgical augmentation:
ACL reconstruction

Figueroa et al
Of the 9 RCTs included, 2 studies showed PRP might reduce graft maturity time (one used L-PRP, the other type was unknown).

[IMG alt="An external file that holds a picture, illustration, etc.Object name is eor-6-225-i010.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8142058/bin/eor-6-225-i010.jpg[/IMG]

Sacroiliac joint instability

Ko et al
Clinically and statistically significant improvement in pain at 12 months post treatment with L-PRP. Clinically significant improvement still present at 4 years post treatment.

[IMG alt="An external file that holds a picture, illustration, etc.Object name is eor-6-225-i011.jpg"]https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8142058/bin/eor-6-225-i011.jpg[/IMG]


Open in a separate window
Note. P-PRP, pure platelet-rich plasma; L-PRP, leukocyte-rich platelet-rich plasma; RCT, randomized controlled trial; ACL, anterior cruciate ligament.

Tendinopathies​

The majority of research into PRP treatment for tendinopathy centres on lateral epicondylitis, where PRP has been shown through systematic review52 to have a better, albeit delayed, therapeutic effect compared to corticosteroid injection for up to two years post injection (CMS 53). Three of the five RCTs analysed used leukocyte-rich PRP (L-PRP), the others did not document the type of PRP used. On further analysis, the RCTs that showed the most significant improvements compared to corticosteroid, were those documenting L-PRP was used.
Systematic review and meta-analyses of studies assessing PRP efficacy in Achilles tendinopathy53 showed that PRP conferred no clinical benefit when compared to saline placebo or an eccentric loading programme (CMS 65). Two of the studies used L-PRP, the other did not document the type of PRP used.
A systematic review and meta-analysis of two RCTs assessing L-PRP efficacy for patellar tendinosis54 suggested that PRP was statistically better than dry needling or extracorporeal shockwave therapy at six months post treatment (CMS 66).
There have been two RCTs assessing PRP versus saline injection55 and dry needling56 respectively in the treatment of rotator cuff disease (tendinopathy or partial tears). Rha et al56 found that PRP provided more symptomatic relief and functional improvement (based on greater reduction in shoulder pain and disability index) at six weeks to six months post injection than dry needling (CMS 66). The type of PRP was not documented. Whereas, Kesikburun et al55 found no difference between L-PRP and saline injections at any follow-up point up to a year post injection (CMS 71).
The combined evidence for PRP efficacy in tendinopathies shows that in the studies where PRP has shown statistical improvement to control measures, it is L-PRP that has been used.

Cartilage pathology​

Laver et al57 reviewed all studies that assessed PRP for the treatment of degenerative cartilage pathology. A total of 29 studies were included, nine prospective RCTs, four prospective comparative studies, 14 case series, and two retrospective comparative studies. Of the nine RCTs, all reported improved symptoms with PRP groups at the final 12-month follow up, seven of which were significantly superior results. Generally, all studies appear to show overall positive results and clinical benefit from PRP, irrespective of methodological variation. Interestingly, there was a trend towards improved outcomes in either patients of younger age or early OA changes. Only one study followed up patients beyond 12 months (to two years). In this study, while there was symptomatic improvement at 12 months follow up; there was significant decrease in functional scores at two years, albeit still higher than the baseline level (CMS 61). Twenty studies used pure PRP (P-PRP), seven studies used L-PRP and two studies did not document PRP leukocyte content. Of the nine RCTs reporting improved outcomes, eight used P-PRP, while one used L-PRP. Whilst not directly investigated, these findings suggest P-PRP is more suitable to intra-articular pathology.
Further review and meta-analysis by Chang et al58 reinforced the findings of Laver et al.57 Specifically that less severe OA benefits more from PRP, and PRP is likely to be superior to hyaluronic acid for functional outcomes and have longer duration of action (up to a year).
A case series by Ko et al59 (level 4) has even shown L-PRP can significantly reduce chronic low back pain in patients with sacroiliac joint (SIJ) instability when injected under ultrasound guidance into the SIJ, lasting up to four years (CMS 59).

Acute muscle injuries​

A systematic review and meta-analysis of six RCTs assessing the effectiveness of PRP in reducing return to sport times, demonstrated that when taking into account all six studies, the return to sport time was significantly shorter (by 7.17 days) in the PRP group (CMS 67).60 However, when only the double-blinded studies or studies including only hamstring injury were included in the analysis, no significant difference was noted. In addition, re-injury rates were similar between PRP and controls across studies. There were no significant differences regarding pain, muscle strength, flexibility, muscle function or healing (on ultrasound scan or magnetic resonance imaging).60 Two studies used P-PRP, two used L-PRP, and two did not document PRP type. These findings suggest that when return to play as early as possible is the primary motivation (such as for professional sport) it can be worth using PRP. However, the results are varied and the type of PRP best suited is unknown.

Surgical augmentation​

Multiple studies have looked at the use of PRP as an augmentation for surgery to expedite healing and recovery time. The majority of studies assessing this are focussed on rotator cuff repair and anterior cruciate ligament (ACL) surgery. Cohn et al61 reviewed five RCTs assessing the effect of PRP versus no treatment in conjunction with rotator cuff repair. Only two of the studies showed any benefit. Randelli et al62 demonstrated less pain in the early post-operative period and increased strength of external rotation at three months post-operatively in the L-PRP group (CMS 76). Interestingly, subgroup analysis of grade 1 and 2 tears showed greater strength of external rotation from 3 to 24 months post-operatively, suggesting milder tears may benefit more from L-PRP. Jo et al63 looked at PRP efficacy in large rotator cuff tears and found that re-rupture was 20% lower in the PRP + surgery group compared with surgery alone, as well as the overall shoulder function being significantly better (CMS 73). However, the type of PRP used was not described. The other RCTs showed no significant differences in peri-operative morbidity, clinical outcomes of structural integrity between PRP + surgery and surgery alone. Two of the studies used P-PRP while the other did not specify the PRP classification. Overall, these results show L-PRP may be of benefit in rotator cuff repair. A 20% reduction in large tear re-rupture is certainly worth the addition of PRP. However, the type was not documented. Interestingly, of the three RCTs showing no benefit with these tendon injuries, two used P-PRP and the other was unspecified.
A systematic review of nine RCTs and two cohort studies assessing PRP use in ACL surgery64 (level 3) showed there is evidence that adding PRP to the graft or tunnels could be beneficial in expediting graft maturity (CMS 60). Seven studies used L-PRP, two used P-PRP and two did not document PRP type. Similarly to muscle injuries, where early return to play is a crucial, these finding suggest the addition of PRP during ACL reconstruction may be of benefit. However, the type of PRP is again unclear.
Go to:

Discussion​

The breadth of applications for PRP in orthopaedics is vast. There have been encouraging results in a multitude of studies focussing on different potential indications. However, the sheer scale of heterogeneity across studies makes it difficult to draw clear conclusions from promising results. In addition, many studies will group soft tissue injuries together in their analysis, thereby further compounding the heterogeneity and potentially obscuring the true impact that PRP may have on specific soft tissue pathologies.
Too often the classification of PRP is not made clear, making it difficult to establish trends of PRP efficacy for differing pathologies. This is especially important when many clinicians are matching the type of PRP to specific pathologies, based on loose clinical indications from studies where the primary aim was not the comparison of PRP types for specific pathologies. Throughout the applications section we have highlighted the PRP type used in each study and, in conjunction with the results, thereby suggested which PRP type appears more effective for each indication based on the analysed evidence. However, it must be emphasized that no specific trends of impaired or improved outcomes of one PRP type over another have been observed for any indication. This is due to the methodologies of the analysed studies not being specifically designed to answer the question ‘Which PRP type is best for this indication?’ Therefore, as there is no direct comparison between these two PRP formulations for any indication, definitive conclusions cannot be made. This highlights the importance for future research to compare PRP formulation efficacy across applications, or at least state clearly what PRP formulation is being used so it can be accurately classified.
While there has been no direct comparison of PRP types for different applications within the literature, L-PRP appears to be more effective in chronic tendinopathies. This is due to the natural first stage of tendon healing including inflammation from leucocytes and catabolic cytokines.66 In contrast, P-PRP seems to be more beneficial in cartilage pathology.65,67 This may be because L-PRP has been shown to cause a significantly greater acute inflammatory response and increased synoviocyte cell death.67,68
Go to:

Conclusion​

Going forward, there needs to be standardization of certain parameters regarding PRP research. Murray et al69 have produced a comprehensive 23-statement checklist that all future clinical studies in PRP should adhere to, with the aim of streamlining PRP research towards yielding robust evidence.
 
Buy Lab Tests Online
Defy Medical TRT clinic

Sponsors

enclomiphene
nelson vergel coaching for men
Discounted Labs
TRT in UK Balance my hormones
Testosterone books nelson vergel
Register on ExcelMale.com
Trimix HCG Offer Excelmale
Thumos USA men's mentoring and coaching
Testosterone TRT HRT Doctor Near Me

Online statistics

Members online
4
Guests online
4
Total visitors
8

Latest posts

bodybuilder test discounted labs
Top