Open access peer-reviewed chapter

A New Cell Stem Concept for Pelvic Floor Disorders Prevention and Treatment – Endometrial Mesenchymal Stem Cells

Written By

Manuela Cristina Russu

Submitted: 05 August 2022 Reviewed: 12 September 2022 Published: 29 October 2022

DOI: 10.5772/intechopen.108010

From the Edited Volume

Possibilities and Limitations in Current Translational Stem Cell Research

Edited by Diana Kitala

Chapter metrics overview

129 Chapter Downloads

View Full Metrics

Abstract

High rate complications and recurrences in reconstructive surgery using in situ synthetic/polypropylene (PP) meshes have driven to a new concept based on mesenchymal stem cells (MSCs) for homeostasis repair in pelvic floor disorders (PFD). Prevention and therapy with MSCs are up to date analyzed on small and large animal models, less in women trials. Cell based-vaginal/intraurethral, or systemically introduced, tissue engineering (TE) with new generation meshes/scaffolds MSCs seeded-bone marrow, adipose tissue and recently proposed the endometrial/menstrual MSCs (eMSCs/MenSCs) for PFDs, management. Easy collected, isolated with specific markers, cultured for number harvesting, without ethic and immune compatibility issues, with unique biologic properties eMSCs/MenSCs differentiate in many cellular types—smooth muscle, and fibroblast-like cells, preserving cell shape, and phenotype, without oncogenic risks, and collagen, elastin fibers; eMSCs/MenSCsare appropriate for PFDs management, respecting good protocols for human safety. The quick appeared regenerative effect-mediated by angiogenesis, apoptosis inhibition, cell proliferation, no chronic inflammation and low/no foreign body reactions, less thick collagen fibers, and fibrosis improve connective/neuromuscular tissues; less pelvic structures stiffness with more elasticity are advantages for new meshes/scaffolds generation in TE. Human eSMCs/MenSCs deliver bioactive factors by their exosomes/microvesicles/secretome for paracrine effects to injury site, facilitating in vivo tissue repair.

Keywords

  • pelvic floor disorders
  • endometrial/menstrual mesenchymal stem cells
  • cell based therapy
  • tissue engineering
  • new generation meshes/scaffolds

1. Introduction

Pelvic Floor Disorders (PFDs) are a continuous gynecologist’s challenge, reason for this paper to review their correction attempts, by better knowledge of pathophysiology, pelvic connective tissue structure and biology with the new concepts of regenerative medicine by mesenchymal stem cells (MSCs) and/or their bioactive mollecules, sometimes crucial. All these objectives are after the era of reconstructive surgery with native tissue or in situ synthetic/polypropylen (PP), non-absorbable meshes for fascia repair. The surgical procedures have immediate, and subsequent (within 5 years) complications (10–30% rate) [1], disorder’s recurrence or a new disorder, and host reactions to mesh—as a foreign body reaction (FBR), with fibrosis, mainly to transvaginal type, explaining abnormal functions after interventions. FDA (USA) had two public health notifications, warning since 2010 against vaginal mesh use in POP, because complications, recurrences, and litigations [2]; PP mesh use is prohibited for transvaginal POP surgery in Australia, New Zealand, USA, and UK [3]. Stem cells, as endometrial/menstrual mesenchymal stem cells (eMSCs/MenSCs) with their remarkable unique biological properties are considered a new potential tool for PFDs prevention and therapy, in a properly response to pathophysiology. MSCs are key regulatory components in the regenerating stem cell local microenvironment termed “stem cell niche” or for MSCs culture conditions, per se or by their secretome, facts that positively influence altered pelvic floor connective tissue, and contribute to tissue homeostasis restoration [4]. Allogeneic and autologus MSCS/MenSCs are proposed for these aims, being easy collected, isolated, purified with known perivascular surface markers for pericytes [5], and with the novel single perivascular marker Sushi Domain Containing 2 (SUSD2) for purifying eMSCs [6], and maintaining cells clonogenicity, reduced by flow cytometry used for their sorting [5, 6]. Different to other MSCs culture expandation imposing presence in culture medium of some constituents of their secretome [7, 8], for eMSCs and ERCs one adds transforming growth factor-β receptor inhibitor (TGF-β R inhibitor), that limits cells spontaneous differentiation to fibroblasts, and maintains the undifferentiated cells status in days following administration, to ensure efficacy [9]. eMSCs and ERCs have no ethic issue, or incompatibility risk, no need of immunosuppressive adjuvant drug, they intervene in repair and regeneration by new blood vessels formation, modulating host immune system, reducing chronic inflammation, FBRs, and fibrosis, with no scar as endometrium regenerates after each menstruation from the menarcha to menopause, under a normal estrogen/progesterone balance, and in postmenopause when on menopausal hormone therapy (MHT). One proposes cell based therapy-eMSCs/MenSCs direct placement in vaginal walls or intraurethral, or systemic administration [10, 11] to prevent postpartum POP or SUI, and tissue engineering (TE)/tissue grafts with eMSCc/MenSCs seeded in a new meshes/scaffold generation preferable biodegradable, obtained by a variety of technologies, as knitting (like old ones) from alternative synthetic and natural polymers, electrospinning, and three-dimensional printing [12], and a composite mesh of polyamide plus a gelatin layer [13], instead of PP meshes, with no ectopic tissue formation, or malignant tumor [14]. There are many small and large animal models, and few human clinical studies on MSCs for tissue restoration and repair when PFDs.

Advertisement

2. Contemporary burden of women’s pelvic floor disorders

PFDs named also Pelvic Floor Dysfunctions represent a women old pathology, with high incidence and prevalence in the last 70 years, associated to worldwide lifespan increase, independently to their different definitions and diagnosis criteria, last updated in 2010 by the International Urogynecological Association and International Continence Society [15], with over 250 definitions for clinical categories and subclassifications, and alphanumeric code for each definition. PFDs is an umbrella term for a group of clinical conditions caused by pelvic floor supportive tissue weakening, sometimes degenerative, occurring independently or simultaneously, connected to genetics, childbirth and aging, and it includes pelvic organs prolapse (POP) into vagina, as a hernia in the endopelvic fascia [16], and alterations in sensory or emptying abnormalities of the lower urinary and gastrointestinal tracts: urinary incontinence (UI) with clinical manifestations of stress urinary incontinence (SUI), overactive bladder syndrome (ORB), detrusor instability, urinary retention, and fecal incontinence, and sexual dysfunctions. PFDs are diagnosed in middle-aged women as stress urinary incontinence (SUI)—incidence peak around 46 years, and in elderly women when a second SUI peak between 70–71 years, being described in young women, also, around 18 years, with annual risk of 3.8 and 3.9 per 1000 women at that bimodal peaks; the risk for pelvic organ prolapse (POP) is increasing progressively from 50 to 60 years in Swedish population [17], to ages of 71 and 73 years, when annual risk is 4.3 per 1000 North American women evaluated for lifetime risks for SUI, or POP, or both surgery between 2007 and 2011 [18], and higher in institutionalized, and when dementia [19]. The recent Swedish national register-based cohort study [20] estimated a 12–19% lifetime risk for surgery for POP, similar to the earliear 19%-for Australian women [21]. PFDs have serious negative influences on women’s quality of life at any age [22]. SUI and POP are highly related to childbirth-associated pelvic floor injury [23], less influenced by route of delivery (vaginal, cesarean section), sometimes inherited, as a familial disorder, associated to other hereditary conditions—joint hypermobility and even Ehlers–Danlos syndrome, a mutation in the gene for collagen III [24, 25]. One recommends the extension of familial conditions lower than first degree generation relationship [26] for familial risk reduction and prolapse prevention, many cases being more frequently in postmenopause, without any pregnancy/delivery, when the association of ovarian aging hypoestrogenism, plus vasomotor syndrome, depressive mood, chronic constipation and cough, obesity, characteristics for this life period, are increasing PDFs medical and financial burden [27].

2.1 Synthetic mesh unfavorable results in surgery for pelvic floor disorders

In front of PFDs burden, one tried to correct the supportive tissue of cardinal, utero-sacral ligaments, levatorani muscle, and urethral sphincter damaged by gestation, parturition, and aging initially by non-surgical interventions: physical exercises (after delivery), pessaries—first choice for POP symptomatic women [28], or as a tool for decision of mid-urethral slim mesh, when one decides POP surgery, to avoid over and under-treatment if vaginal mesh is used for an nonexistent SUI or a urinary leakage when surgery for POP [29], pharmacological therapies, laser, nonablative monopolar radiofrequency [30], and surgery, when all these failure; either vaginal or abdominal route, or both with native tissue repair, through open surgery or laparoscopy, or reconstructive surgery with synthetic, monofilament PP mesh respecting the principles for different PFDs manifestations, classified in anterior and posterior compartment, and vaginal vault disorders. It was evident that the addition of mesh as reinforcement to vaginal walls provides better prolapse correction, compared with colporrhaphy using native tissue alone, by both objective and subjective criteria [31], as it was demonstrated for abdominal wall defects/hernia repair [32]. Unfortunately medical staff and patients wishes for pelvic floor normal functions restoration with PP meshes were not accomplished, or the results were not better than after native tissue repair, being reported high risks—immediate, subsequent (within first 5 years) readmissions for later postoperative complications, such as intractable pain, or mesh erosion or extrusion into the bladder, bowel or vagina [33], requiring surgical excision in ≥10% [28], or deterioration of vaginal biomechanical properties by high stiffness mesh implanted for prolapse [34], and further recurrences of previous disorder, or a de novo one, as incontinence surgery-incidence of 9.9% after surgery for POP without occult SUI (apical and non-apical) [35] or further prolapse surgery, sacro-spinous fixation with synthetic mesh being a risk factor for POP recurrence [36], andtransvaginal PP mesh having a higher risk than vaginal vault sacro–colpopexy [37, 38]. The population-based cohort study from Scotland [39] compared the primary outcome—immediate postoperative complications and subsequent (within 5 years) readmissions for later postoperative complications, further incontinence surgery, or further prolapse surgery in women older than 20 years, after first, single PP mesh (retropubic mesh, or single prolapse mesh procedure) to non-mesh procedures during 20 years. Immediate complications were lower after mesh procedures [adjusted relative risk (aRR) 0.44 (95%CI 0.36–055) and subsequent prolapse surgery [adjusted incidence rate ratio (air) 0.30 (0.24–0.39)], and a similar risk for further incontinence surgery [0.90 (0.73–1.11)], and later complications [1.12 (0.98–1.27)]. Anterior compartment prolapse PP mesh repair was associated to a similar risk of immediate complications as non-mesh surgery [aRR 0.93 (95%CI 0.49–1.79)], but with an increased risk of further incontinence [air 3.20 (2.06–4.96)], and prolapse surgery [1.69 (1.29–2.20)], and a substantially higher risk of later complications [3.15 (2.46–4.04)]. Posterior compartment mesh repair was associated to a similarly increased risk of repeat prolapse surgery and later complications as non-mesh surgery. Vaginal vault prolapse had similar outcome when vaginal and, separately, abdominal mesh repair were compared with vaginal non-mesh repair. Both vaginal and abdominal mesh procedures for vaginal vault prolapse repair are associated with similar effectiveness and complication rates to non-mesh repair.

The synthetic meshes used for pelvic floor reinforcement or reconstruction may provide the necessary mechanical support for damaged tissue, but implants’ biological actions interfere with host biology, inducing the growth of a fibrous tissue layer, as an additional physical support, but the scar may contract the mesh, and surrounding tissue up to 60% [40], Mesh parameters influence host tissues: microstructure-porosity (permeability for host’s cells, mainly immune cells, fibroblasts, macrophages, metabolites, oxygen at repair place, and also bacteria effects, by chronic inflammation), fiber filament type (monofilament: polyamide and polyetheretherketone monofilaments or multifilaments; synthetic/natural/composite), and diameter, mesh weight (light/ultralight/heavy), mechanical properties as stiffness, and elasticity, chemical properties, materials biodegradability, and integration in host organ by new blood vessels formation, facts that contributed to the new concept of stem cells in cells based therapy, and tissue engineering (TE) for PFDs prevention or repair [13, 41].

Advertisement

3. A new concept of stem cell use in prevention and therapy of women’s pelvic floor disorders

Tissue engineering is a new option in the field of pelvic floor repair when soft tissue reinforcement or reconstruction, or normal function are necessary. MSCs and/or their secretome are proposed after many studies regarding pelvic tissue biology, materials properties associated to stem cells designed to restore the anatomical functions, to provide a real pelvic floor mechanical support after damage, usually to be like a hammock, and to offer both the lost stiffness when under tension and the flexibility under bending [42].

3.1 Tissue homeostasis/remodeling behind repair in pelvic floor disorders

Pelvic floor—a complex resistance piece, keeps pelvic organs within the body, still allowing passage through urethra, vagina and anal canal, around which are designated striated muscles—levatorani, with its three fascicles, and superficialperinealmuscles or urogenital diaphragm [43], forming a functional neuromuscular unit, and fibrous connective tissue, generating endopelvic fascia, cardinal and utero-sacral suspensory ligaments, and vaginal dense fibromuscular-connective tissue. Pelvic connective tissue maintains the position of organs adjacent to vagina, and the close anatomical relationship among vagina, bladder, and rectum may contribute to the emergence of anatomical-functional failure of adjacent organs/systems, in PFDs [44], according to their normal different stiffness/elasticity. Animal models indicate that molecular changes in tissue composition, mainly protein content, coincide to altered biomechanical properties, in PFDs, mainly in POP, as one cites in Australian [45], European [46], North American analyses [47, 48]. Human and mouse pelvic floor provided similarities of pathological changes, centered on deep and superficial muscles, ligaments, and connective tissue, mainly on vaginal walls. Pelvic floor connective tissue contains stromal cells and a very complex extracellular matrix (ECM). The balance between ECM synthesis and degradation during women/females life is a key in pelvic floor properties, and the vaginal structures are strongly influenced during women, small and large animals life, ovine models demonstrating architectural and functionaldifferences according the reproductive status. The Australian ovine model [45] revealed the lowest total collagen content in virgin vaginal tissue, in contrast to parous tissue with highest total collagen and lowest elastin content with concomitant high maximum stress, in contrast to pregnant sheep with lowest collagen and highest elastin contents, and thickest smooth muscle layer and low maximum stress, and poor dimensional recovery following repetitive gestational loading. The vaginal tissue is anisotropic with some biomechanical properties—loading pressure, deformation rates, resistance to rupture, which were tested in ewes [49], and in vaginal specimens collected during surgery for POP [50] compared to specimens from cadavers without noticed PFDs (non-pelvic organ prolapse)—the first experimental study providing vaginal tissue mechanical behavior. The results highlight the non-linear relationship between stress (force per unit of surface) and strain, the vagina being hyperelastic and supporting very large deformation before rupture appearance, as in labor, and fetal expulsion. The vaginal walls tissue is stiffer in patients with POP than non-POP [51]. Comparison of biomechanical properties of the crucial organs of pelvic support [52], showed significant differences at large strain levels: vagina is more rigid, and less extendible than rectum, which, is more rigid than the bladder. The anterior and posterior vaginal walls have different stiffness, and the bladder tissue was anisotropic at large strain levels, facts very important for tissue repair: region with dysfunction/disorder, or procedure type.

ECM molecules are arranged in a matrix/scaffold, surrounding stromal cells (fibroblasts, myofibroblasts, smooth muscle) that synthesize collagen, as tropocollagen and elastin, as tropoelastin to form the fibers complex network, plus proteoglycans, and matricellular proteins, enzymes, according to their genes. All forming the complex network of pelvic floor support, recently updated [53]. EMC contains:

  • tropocollagen, self-assembled into fibrils, aggregating to form a collagen type I (for tensile strength) and type III collagen fiber (for elastic properties, and increased collagen III reduces mechanical strength [54]; collagen form a cross-linked network intertwined with elastin—the elastic fibers core component, secreted by elastogenic cells as the monomer tropoelastin, and undergoes self aggregation, cross-linking and deposition on to microfibrils assemble into insoluble elastin polymers. A microfibril scaffold-primarily formed by the protein fibrilin-1, is required for elastic fibers formation [53]. Collagen and elastin fibers are surrounded by a viscous substance of proteoglycans-consisting of a core protein to which one or more glycosaminoglycan (GAG) chains—ashyaluronan or hyaluronic acid, heparan, dermatan sulfate and the small leucine-rich repeat proteoglycans (SLRPs)-decorin, lumican and fibromodulin [17] are covalently attached; SLRPs cover the surface of collagen fibers, contributing to fiber optimal formation [55]. Proteoglycans have key roles in controlling gradients and availability of potent growth factors, chemokines, cytokines, and morphogens, very important in tissue’s homeostasis, mechanical strength, development, and repair. One or more proteoglycans are cell surface or transmembrane receptors for adhesion molecules in all mammalian extracellular matrices [56], contributing to progenitor stem cells microenvironment/niche [57, 58].

  • matricellular proteins or elastic fibers associated [59]: fibrilin-1, fibrilin-2, fibulin-3, -4, -5, are involved in elastic fibers synthesis and assembly. Fibulin-5 is a pivotal molecule with dual functions involving MMP-9 enzyme regulation, and in tethering polymerized monomeric form of elastin to surrounding cells in vaginal wall, and positively regulating coacervation, but negatively regulating maturation of coacervated elastin in vitro [60]. When pre pregnancy POP fibulin-5 knockout mice increases in severity after vaginal delivery [61, 62, 63].

  • adhesion molecules: fibronectin; integrins—α, β or α-β integrins interaction with fibulin-5 is essential in vascular development, but dispensable for fibronectin fibrils assembly.

  • enzymes:

    • matrix proteases as matrix metalloproteinase (MMP)-2, MMP-9 involved indisruption of collagen and elastin fibers, and particularly increased in POP, and in postmenopause comparative to premenopauseal asymptomatic cases; estrogens withdrawal or antiestrogenic therapy upregulates MMP-9; and TIMP-1, TIMP-2

    • lysyl oxidase-like–1 (LOXl-1)—predominantly catalyzes elastin cross-linking, its inhibition associated to increased MMP-9 led to subclinical POP [27, 64], because tropoelastin accumulation, according to the theory of antielastase-elastase inbalance in mice lacking LOXl-1, and the lack of deposit with normal elastic fibers in the uterine tract, and an abnormal postpartum heal of elastin, In LOXL1 knockout mice, smooth muscle cells stiffness and cells adhesion are altered, being proved the interplay between smooth muscle mechanics and ECM remodeling, mainly in postpartum [65, 66].

  • water, very important for all body composition.

    The content, aspect and cross-link of collagen and elastic fibers, matricellular proteins, proteoglycans, and enzymes specially MMPs, LOXl-1 are negatively changed when POP; elastic and mechanical strength are decreased during gestation, and with age, beingconceivable that a loss of elastic fiber–associated proteins in pelvic floor connective tissues with aging, may disrupt the optimal balance between synthesis and degradation of vaginal elastic fibers, and lead to POP, fact associated to the critical negative proteases role in POP progression [48]. The fibers amount is less interested initially after vaginal delivery; their histomorphology is first changed, regarding length (shorter), and cross-linking in net-work [61], fibers density decreases later, by aging [67]. The quantification of collagen and elastic fibers shows a more important decrease of elastic fibers in superficial epithelial layers near vaginal epithelium, and less in the deepness of pelvic cavity, around muscles, and thin, irregular and disrupted collagen bundles, higher levels of collagen type III in the vaginal wall, and fragmentation of collagen fibers [68], being appreciated that epithelial-stromal interactions, and fibulin-5-integrin interactions—that suppress ROS generation, are critical in regulation of MMP-9 in mice vaginal wall [48], with an increased level of MMP-2, -9 in advanced prolapse [69, 70]. It is sure that such molecular changes are not corrected by surgical techniques with native tissue or with PP mesh, and the procedures of tissue engineering by different MSCs types, and/or their secretome may change pelvic floor future histomorphology and functionality with normal/near normal connective tissue appearance, that will be discussed in Section 2.5.

3.2 Genetics, gestational and postmenopausal influences on pelvic floor connective tissue disorders

Genes, sexual steroid hormones with their receptors, ligands and co-activators modulate pelvic floor structures, and volumes entire women’s life. One analyses Homeobox genes (HOXA-11involved in utero sacral ligaments fibroblasts proliferation and p53 regulation [71]), gene encoding LOXl1-generating a primarily failure of elastin postpartum healing in knockout mice, the decrease gene signal for production of three SLRPs-decorin, lumican and fibromodulin, which are collagen fiber assembly regulators with affected collagen fibrilogenesis and collagen fibrils shape, and impairment in elastic fiber assembly by down regulation of fibulin-5 in POP [17]. Genes encoding fibulin-5, fibulin-3, Upii-sv40t—involved in elastin fiber structure, are analyzed in PFDs associated to knockout mice aging [27].

Pregnancy induces adaptations in pelvic floor structures for vaginal delivery to withstand deformations with minimum damages, but vaginal delivery leads to floor disorders, damaging nerves, connective tissue, pelvic smooth and striated muscles. Pelvic connective tissue reduced stiffness and elasticity is essential, being demonstrated that the load carrying response (other than the functional response to the pelvic organs) of each fascia component, pelvic organ, smooth muscle, and ligament are assumed to be isotropic, hyperelastic, and incompressible [72]. There are parallel gestational changes in levatorani muscle: sarcomerogenesis, fiber elongation, and an increased ECM collagen content, with muscular stiffness [73], to avoid sarcomere hyperelongation resulted from mechanical strains imposed by vaginal delivery. Sometimes delivery related strains lead to acute sarcomere hyperelongation, and pregnancy pelvic floor muscles (PFMs) adaptation is exceeded [74]; with pelvic floor muscles (PFMs) avulsions discovered postpartum [73]. Human parturition needs PFMs elongation of 300% in resting muscle length to achieve fetal vaginal delivery, as computational models revealed [75]. An instrumental vaginal delivery with forceps may induce important damages of levator ani muscle visible at 3D/4D postpartum ultrasound [76], and one considers that the majority of vaginal deliveries are followed by subclinical damages. The postpartum pelvic floor repair is different from cervix uteri repair, with loss of pregestational EMC composition restoration. Each vaginal delivery, in special genes and familial heredity conditions, and also without these risk factors, may contribute to EMC damages, with changes of pelvic floor shape and in biochemical structure in pregnancy, and post delivery versus nullipara [77], gestation has the greatest impact on vaginal tissue composition and biomechanical properties proved in animal models (mice, sheep), and women. Important differences of pelvic floor EMC structures are described [45] between virgin, pregnant and parous females regarding total collagen, collagen III/I + III ratios, GAGs, and elastin, and in passive biomechanical properties-compliance and elasticity, and maximum stress and strain, with permanent strain following cyclic loading after each gestation. Vaginal tissue of virgin sheep had the lowest total collagen content and permanent strain, and parous tissue had the highest total collagen, and lowest elastin content with concomitant high maximum stress in contrast to pregnant sheep, that had the highest elastin and lowest collagen contents, and thickest smooth muscle layer, situation associated with low maximum stress, and poor dimensional recovery following repetitive loading. Vaginal biomechanical properties do not recover after pregnancy to those of virgins [44], and tensile strength appears to be linked to vaginal content: total collagen, elastin, and smooth muscles show a direct influence on tissue compliance—reduced after ovine consecutive pregnancies, different to rectum and bladder compliance which are stiffer than vaginal walls after many deliveries [78]. Vaginal distensibility pregnancy-induced and along vaginal delivery by tissue vaginal pressure is not recovered in late postpartum rats [79]. It was demonstrated in mice vaginal culture, the POP appearance and progression after each pregnancy and vaginal delivery, caused by a combination of inhibited elastin linking with tropoelastin accumulation [47], because inability to initiate damaged tissue necessary clear and replacement, with poor elastin properly self repair after each delivery, through abnormal enzymatic actions of MMP-2 (decrease)—TMP-4 (rise) [48], after an initial total elastin amount preservation.

Ageing associated hypoestrogenism is worsening pelvic floor condition. Sexual steroid hormones have receptors in all pelvic organs, not only in genitalia (Table 1) [80]. Uterine prolapse, but not SUI is diagnosed in nuliparous postmenopausal women.

ERsP4RsARs
Vagina+++
Urethra+
Urethral sphincter+
Periurethal venous system+
Bladder trigon+++
Pelvic floor
Pubo cervical muscle
Levator Ani muscle
Cardinal ligaments
Utero-sacral ligaments
Periurethral Fascia
Perivaginal tissue
+
?(+/−)
+
+ (α, β)
+ (α, β)
+
+
+

Table 1.

Sexual steroid hormones receptors distribution in pelvic floor structures.

Adapted from Rechberger and Skorupski [80] (creative common: License CC BY4.0 for adapt). ERs: estrogen receptors; P4Rs: progesterone receptors; ARs: androgens receptors.

Aging is associated to intrinsic aging stem cells-meaning self renewal reducing, through their genes aging [81] in the general dysfunctional frailty syndrome, where pro-inflammatory cytokines-TNF-α, IL-6 and C-reactive, are increased [82], and one may delay aging effects by menopausal hormone therapy (MHT), started from perimenopause for urogenital aging, with local estriol to reduce vaginal atrophy, and some symptoms of bladder aging [83, 84], and it is adjuvant in pre and long term postoperative care, associated to systemic MHT, around age of 50′ as new protocols advice, and different MSCs, types treatments, for frailty delay to safe health and function of organs/tissues, and one speaks about safe proper frailty treatment with MSCs, to increase health and function of organs/tissues [85, 86].

3.3 Mesenchymal stem/stromal cells for pelvic tissue repair and regeneration

High number surgical gynecological procedures proposed along 100 years showed limitations, low adequacy to PFDs pathophysiology, no restoring organs’ normal positions and functions, and better understanding by three dimensional digital models combining DeLancey JO’s theory [16] to Petros P integral theory of continence [interdependence between pelvic organ support systems, linking ligament fascia lesions, and clinical expression, with less critics after TVT (tension free vaginal tape) technique] [87], and tissue structure continuing to deteriorate by aging after correction, being far to be restored by surgery either by native tissue or by PP meshes. MSCs, and their secretome, are discussed since more than 10 years, specially after techniques for their potency enhancement by specific culture systems, including three-dimensional culture conditions, or their priming preconditioning with some molecules of their secretome [7, 65]. MSCs represent a pathophysiologic correction, and limitation of PDFs progression [88]. Tissue engineering is a new option to restore and maintain micturition normality via direct effects on damaged or dysfunctional tissues, or pelvic floor repair when soft tissue reinforcement is necessary [89, 90], to improve outcomes in POP management [91]. MSCs also referred as mesenchymal stromal cells belong to the pool of progenitor and adult stem cells (ASCs) family, from all postnatal organs and tissues, with specific properties for each one, ensuring the capacity of renewal after damage, and in aging [7]. Collected and isolated from various anatomical sites, more or less easy accessible, few ethics-related issues, MSCs are actually easily separated/purified [92], with specific markers, and cultured. Some consider multipotent MSCs to have a limited self-renewal capacity [93], in a specific microenvironment termed “stem cells niche”, first described by Schofield R [94] as “adult stem cell niche hypothesis”, which is reconsidered to be more dynamically than originally appreciated, with a bilateral influence from healthy or damaged tissue to MSCs, mainly by immunological and inflammatory signals in conjunction to MSCs’ paracrine effects. Others [95] consider that MSCs unique properties-high proliferative ability, self-renewal, differentiation to mesodermal lineages, appropriate to their location, are supporting tissue regeneration in physiologic and pathologic conditions, and are contributing to tissue homeostasis. MSCs are key regulatory components in the regenerating stem cell niche, by the increase of their own compound, or increasing physiologic cells turn-over [96] to support tissue regeneration after injury, or are activated in injured tissue, where they are inactive [97], or are attracting supporting cells to niche [4], or are activating tissue’s own cells to facilitate repair [98], capabilities that are different according to tissue type. These biological properties determined the change of “stem” cell nomination to “stromal” for a more appropriate connotation [7], and earlier Caplan AI [99] proposed the name of “Medicinal Signaling Cells” for a more accurate presentation: when systemically administered MSCs home in on sites of injury/disease, and exhibit a paracrine action, by secreting bioactive molecules as regulatory and growth factors, chemokines, cytokines, nucleic acids, packaged into extracellular vesicles or MSCs-derived exosomes, with trophic and immunomodulatory actions, reflecting that MSCs make therapeutic drugs in situ that are medicinal, important for tissue repair. MSCs fate in a tissue is influenced by local microenvironment or niche fixed compartment, where ASCs are in a dormant state (G0) through signaling pathways inhibitory for growth and differentiation, often involving transforming growth factor-ß (TGF-ß), and bone morphogenetic protein (BMP) family members [57], being anchored to niche cells by adhesion molecules—cadherins, integrins [100] during stem cells’ periods of inactivity, and niche cells differentiation signals to resident stem cells [58]. The bioactive molecules produced through MSCs homing when systemic administrated, or when are added in cultures to potentiate MSCs action, as MSCs primers [65], are considered more important than cell engraftment and replacement. MSCs and their bioactive molecules have proangiogenic [101], antifibrotic, anti-inflammatory, and pro-inflammatory actions, which sustain proliferation [102], and stimulate effect of resident progenitor cells, in relationship to disease/organ/tissue type [103]. The damaged, ischemic tissue activates MSCs after injury, through different inflammatory signals (hypoxia, proinflammatory cytokines as IL-1β, IFN-γ, TNF-α, lipopolysaccharide) from host innate immune system and leads microenvironment to coordinate the production of immunomodulatory factors to sustain inflammation progression and rapid remove of allogenic MSCs, and production of growth factors to stimulate endothelial cells, fibroblasts, and tissue progenitor cells’ differentiation from MSCs niche, all contributing to tissue repair, in an orderly action by angiogenesis, EMC remodeling, and functional tissue restoration [7], MSCs have the ability to home to injured tissues to exert their paracrine actions when systemically administered, a very attractive feature for this chapter discussions (Figure 1) [104].

Figure 1.

MSCs role in damaged connective tissue repair. MSCs activation after tissue injury. The damaged tissue activates MSCs after injury through different inflammatory signals (hypoxia, cytokines asIL-1β, IFN-γ, TNF-α, LPS). MSCs activation leads microenvironment to coordinate the production of immunomodulatory factors, that sustain inflammation progression, and of growth factors to stimulate endothelial cells, fibroblasts, and tissue progenitor cells to differentiate, and all contribute to tissue repair, in an orderly action by angiogenesis, EMC remodeling, and functional tissue restoration. Adapted from Miceli et al. [7]. It is an open access article, distributed under the terms and conditions of the creative commons attribution (CCBY), licensee MDPI, Basel, Switzerland.

MSCs do not impose immunosuppresion, being immune-privileged due to their low expression of CD40, CD80, CD86, and major histocompatibility complex I (MHC I), and the lack of MHC II [92], or because they are immune evasive [105].

Advertisement

4. Endometrial and menstrual mesenchymal stem cells for prevention and therapy of pelvic floor disorders

MSCs from different tissues exhibit many common characteristics, their biological activity and some markers are different and depend on tissue origin: bone marrow (obtained by aspirate), adipose tissue (obtained by liposuction), placenta (for maternal MSCs), and umbilical cord, amniotic membranes and liquid (for fetal MSCs) collected at birth. All these MSCs have some limitations, as in vitro expansion [106] for their rarity in original tissue, invasive methods to harvest bone marrow aspirates, donor aging affects MSCs proliferative capacity [86], and the necessity to add their secretome’s active molecules in culture medium [7, 8, 107].

4.1 Unique characteristics of endometrial mesenchymal stromal/stem cells and menstrual mesenchymal stromal cells

In the last 15–20 years, a MSCs subpopulation of stem/progenitor/regenerative cells has been identified and characterized in human endometrium (eMSCs) and in menstrual blood (MenMSCs or ERCs), comparable to bone marrow and adipose tissue MSCs [95], but with unique biologic characteristics [1, 108], and knowledge plus new technique capabilities made them a very promising MSCs category in autologous and allogeneic cellular therapy, and TE. Uterine fragments of shedding endometrial tissue with their remarkable cells turn-over—like hematopoietic bone marrow, intestinal epithelium, epidermis, contribute to endometrial repair and renewal without scar, and gene profiling has demonstrated that the lysed stroma is enriched in genes involved in EMC dynamics, biosynthesis and degradation [109, 110], very promising in pelvic floor connective tissue repair/restoration, and other endometrial fragments from menstrual blood contain MenSCs. The concept of endometrial renewing after each menstruation (~400 cycles in woman’s life) by endometrial stem/progenitor cells located perivascular [6], in the basalis of endometrial glands near myometrium was first hypothetized by Prianishnikov VA, 1978 [111], reloaded by Padykula HA, 1989 [112], and it is continued in Australia at the Department of Obstetrics—Gynecology, Monash University and Centre for Women’s Health Research (Melbourrne, Victoria) by a team led by Gargett CE [113], who presented the first direct evidence that human endometrium contains rare populations of epithelial (0.22%), mesenchymal/stromal-eMSCs (1.25%), and endothelial progenitor cells, which exhibit the adult stem cells behavior in vitro, as clonogenicity, and later their differentiation potential (reviewed in 2016) (Figure 2) [115].

Figure 2.

Schematic perivascular localisation of human eMSCs. Co-expressing CD146 and PDGFRβ/CD140b and SUSD2+ eMSC in the endometrial basalis and functionalis layer, indicating eMSC will be shed into menstrual blood. Reprinted from Gargett and Masuda [114] with license permission: 535199193277 for Copyright Oxford University Press, 2022, July.

One may add a small side population (SP) cells which enriches endometrial stem cells fractions, according to their identity and differentiation potential [116]. eMSCs are responsible for cyclic regeneration of human, mice, and ovine endometrium [117, 118, 119, 120]. Other research group [121] demonstrated that the low number of human endometrial stromal/stem cells seems to belong to the family of MSCs, by possessing the minimal criteria of MSCs assessment [122] clonogenity, self-renewal, plastic adherence in culture, high proliferative potential and capacity and ability to differentiate into at least one type of mature functional progeny, but eMSCs have multilineage differentiation capacity [123]. eMSCs have proliferative capability to undergo 30 populations doubling before reaching senescence, generating 6.5 × 1011 cells (Table 2) [124].

  • MSCs must be plastic-adherent when maintained in standard culture conditions.

  • MSCs must express CD105, CD73 & CD90, and lack expression of CD45, CD34, CD14 or CD11b, CD79α or CD19 & HLA-DR surface molecules.

  • MSCs must differentiate to osteoblasts, adipocytes and chondroblasts in vitro

Table 2.

Minimal criteria for defining multipotent MSCs.

International Society for Cellular Therapy position statement [122].

Taylor HS [125] at Yale University (USA) presented bone marrow (BM) as an exogenous source of eMSCs, which appear histologically as epithelial and stromal endometrial cells, expressing appropriate markers of endometrial cell differentiation, and cyclic mobilization of BM-derived stem cells is considered a normal physiologic process [126]. Menstrual blood, an usual waste tissue, but a “bio-waste” as recently reconsidered [127] (endometrial functionalis layer shed during menstruation) is an easy obtained source of MSCs, with no ethic issue, and isolated, cultured similarly to bone marrow aspirated. Menstrual stem cells (MenSCs), named also endometrial regenerative cells (ERCs) by the team from Bio-Communications Research Institute (Wichita, USA), who first isolated and cultured them [128], have the classic properties, and pattern of MSCs surface markers, are multipotent [129], retain a stable karyotype in culture [130], proved at more than 68 doublings without any karyotype or functional abnormalities [128, 131]. ECRs have similar capabilities for tissue repair and restoration as eMSCs, according to their secretome that ensure cells paracrine actions on endometrium (after menstruation, and Asherman syndrome) [132], ovaries [133], and on different organ [134, 135]. There are many controversies on eMSCs and ERCs makers [108], being demonstrated that endometrial/menstrual MSCs clones express MCSs markers [ITGB1 (CD29), CD44, NT5E (CD73), THY1 (CD90), ENG (CD105), PDGFRB (CD140B), MCAM (CD146)], but not endothelial or hemopoietic markers PECAM1 (CD31), CD34, PTPRC (CD45), and the pan-leukocyte marker CD45 [128]. The Australian team used these markers for eMSCs isolation and culture [124], plus two perivascular cell surface markers—CD146, and platelet-derived growth factor-receptor β (PDGF-Rβ) or CD140B, and have determined eMSCs location near blood vessels in human endometrium [114, 136, 137], blood vessel wall is considered the eMSCs niche, as it is for all MSCs [138, 139]. These markers were used in association to Sushi Domain Containing 2 (SUSD2) or W5C5, a special marker for eMSCs and MenSCs isolation, proposed as a novel single marker for purifying eMSCs, and to reconstitute endometrial stromal tissue in vivo from endometrial biopsies [6]. MenSCs need a selective marker enrichment to be consistent and efficacious as eMSCs obtained by endometrial biopsy. It is known that perivascular MSCs or pericytes are rare cells, difficult to harvest from adult tissues, and necessitate substantial ex-vivo culture expansion to achieve a sufficient number of potent cells, and prolonged culture of MSCs determine a spontaneous differentiation to fibroblasts, which limits culture expansion, and these significant limits challenged the special add in culture medium of a novel small molecule-transforming growth factor-β receptor inhibitor, namely A83-01, that limits the inconvenient and maintains eMSCs and other MSCs undifferentiated in the days following administration and ensure the therapeutic efficacy of a small proportion (2%) of cells which are estimated to remain in vivo in the days following administration [991, 114140]. MenSCs secretome needs a special attention for its exceptional therapeutic effects, due to extracellular vesicles (EV) [135], including microvesicles, exosomes and apoptotic bodies transporting bioactive molecules. A total of 895 molecules are identified in exosomes [141], as micro RNA, lipids, growth factors (vascular endothelial growth factor, insulin-like growth factor-1, hepatocyte growth factor), chemokines, cytokines as regulators of immune response in different tissues [142], which can be isolated from menstrual blood as are MenSCs isolated [143]. The human MenSCs transcriptome and methylome profiles showed their most distinctive expression and epigenetic signature compared to human bone marrow and adipose MSCs [144]. MenSCs trandifferentiation capacity is extensively discussed, associated to their possibility to differentiate into mesodermal lineage (including chondrogenic, osteogenic, adipogenic, and cardiomyogenic fate), endodermal lineage (hepatocyte), and ectodermal lineage (neural and glial) [145], processes that varies considerably between each type, and it is different when one compares them to bone marrow and adipocyte MSCs [128]. Recently there were proved the beneficial effects of MenSCs and their secretome in pulmonary healing in severe acute adults lung cells injury (ARDS) from COVID, by increasing number of CD4 lymphocytes, reducing expression of inflammatory markers (C Reactive Proteine, ferritine, LDH), absorption of bilateral pleural exudates, better than other MSCs types (BM, adipose tissue) when systemic transplanted [146], having also the advantage of easy collection in emergency.

4.2 Potential application of endometrial and menstrual mesenchymal stem cells in pelvic floor disorders prevention and therapy

eMSCs with autologous and allogeneic origins are easily procured from endometrial biopsies—during reproductive years, under contraceptives, in postmenopause [115], without anesthesia or from menstrual blood (source that can be repeatedly used at every menstrual cycle, much easier obtained vs. other sources of ASC). with remarkable differentiation capabilities, plus paracrine actions from their secretome, proved by preclinical and some phase III trials—much discussed and criticized in USA [147], because industry commercial enthusiasm [148]. FDA (USA) approved in 2011 clinical trials with ERCs, which can be used for PFDs prevention and therapy. Actually eMSCs are not accessible for human trials all over the world, or there are no legally approved banks for eMSCs/MenMSCs, these cells being used in some countries only for academic/scientific health centers or not-for profit public institutions. European Medicine Agency has a “hospital exemption” clause, with existence of many unknowns/controversies on eMSCs/MenMSCs-based therapies in human (allogeneic or autologous), as they are used in animal models. Actually one must consider that amalgamation of highly specialized disciplines such as tissue engineering, stem cell therapy and personalized medicine provide important approaches and tools to respond to these challenges in PFDs prevention and therapy, as there were found regulatory approval and deployment for disorders with unmet medical needs [149].

4.2.1 Endometrial/menstrual mesenchymal stem cells based for pelvic floor disorders prevention and therapy

Cell therapy is an emerging field in clinical practice, bone marrow, adipose tissue being subjects for trials in chronic and degenerative disease. Basic evidences provide the preventive role of MSCs autologous cell based therapy in rats SUI by local-urethral administration [150], which is a minimally invasive procedure, and intravenous (i.v.) route [10]. MSCs are homing in damaged pelvic organs, where they are attracted by cytokines [151] and chemokines [152]. After i.v. administration, post vaginal distension (VD)—a model for childbirth injury, GFP-labeled cells were depicted at 4 to 10 days in urethra, vagina, rectum, and levator ani muscle of sacrificed animals, with significantly more MSCs homing at 4 days versus after sham VD, and reduction of GFP intensity at 10 days after VD [153]. It is discussed PFDs prevention with MSCs in high risk women by genetic predisposition, with postpartum SUI and/or POP, and the possibility to induce homing a long time after injury or to increase homing after an acute injury via stem cells genetic modification to express a greater number of homing ligands [154] or an electrical stimulation to the paravaginal region, which induces neural stem cells migration [155], or by MSCs paracrine effects in damaged tissues [growth factors as Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF), angiopoietin-2, and Platelet-Derived Growth Factor (PDGF-BB)], which were proved to be in a rate of about 10–100,000 times more for MenMSCs than the control mesenchymal cell lines derived from umbilical cord blood, and by stimulating metalloproteinases involved in elastin postpartum [128]. Post infusion febrile reaction is the sole adverse event associated to bone marrow MSCs i.v. [156]. One discusses the i.v. dose differentiation between small (rodents), large (ovine) animals, and human: 50 million MSCs/kg for rodents, and 1–2 million cells/kg and never more than 12 million cells/kg for human. MSCs pulmonary entrapment, with their quick phagocytisation, by lung tissue macrophages [157], and the genetic instability and tumorigenicity [156], not valuable for eMSCs/MenMSCs [14]. ERCs presenting more than 68 doublings without any karyotype or functional abnormalities [128, 131].

Australia has programs based on techniques to purify eMSCs by magnetic beads and special markers, for production of large number of cells under Good Manufacturing Practice (GMP) conditions to offer women’s own eMSCs when they need for PFDs, for TE. Actually there are only animal studies, no human trial on eMSCs, /MenSCs for PFDs, being discussed other MSCs types. The Cochrane Database Syst Rev. (2017) [158] found in Cochrane Incontinence Group Specialized Trials Register only one small RCT for SUI, on injection of autologous MSCs with fat origin vs placebo, terminated early because of safety concerns, and afterwards there are mentioned some clinical trials for SUI treated with autologous muscle derived MSCs compared to placebo [159], or transurethral and periurethral intrasphincteric injections of cellular suspension for SUI, with limited accuracy of results [160], or a preliminary randomized study of stem cells from adipose tissue implanted for fecal incontinence [161].

4.3 Tissue engineering with endometrial and menstrual mesenchymal stromal cells for pelvic floor disorders

Tissue engineering (TE) for pelvic floor disorders treatment combines principles of eMSCs/MenSCs biology with materials science (scaffolds, meshes for pelvic implantation), and biomedical engineering [162]. The most discussed PFDs beneficiary of eMSCs therapy are POP and SUI—de novo, persistent or recurrent after failure of surgery with native tissue or reconstruction. There are necessary multidisciplinary trained teams. and special protocols, as are presenting Ichim T (2008) CEO at Medistem Laboratory (San Diego, USA), and Gargett CE (2013)—head of the Australian Stem Cell Centre.

The key to safe and efficacious TE in PFDs was the generation of some tissue substitute by materials with nano-architecture/nanofiber technology [163] or 3D printing, mimicking EMC pelvic floor topography, mainly vaginal EMC, or to induce favorable tissue mechanical responses, or to add some EMC constituents (as tropoelastin-the elastin core in EMC network, besides collagen, lost in POP) [164], for production of mesh/scaffold new generation, which allows entrapment, and persistence of seeded eMSCs/MenSCs up to 14 days after implant [41]. eMSCs are in vitro optimized in serum free conditions—fibronectine is the optimal substrate for human eMSCs attachment [91], and eMSCs transcriptome reveals improved potential for cell based therapy after adding TGF-β receptor inhibitor in culture medium (to prolong their undifferentiated status after implantation, by eMSCs apoptosis, and senescence prevention, and maintaining the percentage of SUSD2+ cells to more than 90% for all samples) [165]. Another proposal for eMSCs tissue repair efficacy augmentation is the add of a protective delivery system, as a compatible bio-hydrogel carrier that encapsulates eMSCs in mesh/scaffold and improves cells retention at site from host immune system actions to rapid their remove, due to loss of vascular niches [107], and by their encapsulation in hydrogel MSCs can promote endogenous cellular repair [140]. There are many composite meshes produced from different materials: nondegradable polyamide meshes, as those of polyamide/gelatin seeded with 100,000 human or ovine MenSCs/cm2, which stimulate angiogenesis, host synthesis type 1 and type III collagen, lower leukocytes infiltrate at 90 days postimplantation (when tested on rats) [166], andthe new biomimetic tissue generation of degradable nano/microstructured meshes [167], meshes obtained through new technologies of electrospinning and 3 D bioprinted endometrial stem cells on an aloe vera–alginate (AV-ALG) injectable hydrogel or on melt electrospun poly epsilon-caprolactone mesh, with the largest open pore diameter and the lowest thickness that promotes eMSCs encapsulated in the hydrogel attachment, which reduces FBRs associated to eMSCs same action [168, 169]. The meshes designed with nanoscale fibers using electrospinning techniques promote cell–cell and cell-biomaterial interactions, being appreciated in Australia [91, 167170], and Nederland studies [171] that biometric properties of this nanostructured mesh can improve the integration, overcome erosion, and offer good outcomes in POP reconstructive surgery. The mesh type added to eMSCs have different persistence time after implantation, the natural ones have the shortest “life” duration after implant; the non-degradable polyamide/gelatin mesh plus autologuse MSC persisted 3 days in immunocompetent mice, 1–2 weeks in immunocompromised rodent model when xenogenic human eMSCs, and 90 days in ovine; the longest duration is at 3 D printed mesh [168]. The Chinese study [12] shows that a composite mesh based on synthetic and natural polymers seems to provide the best combination for an ideal pelvic floor mesh material, because natural polymers can provide ligands for cell adhesion and growth factors that promote tissue remodeling, while synthetic polymers provide mechanical strength. New scaffolds proposed for POP provide a three-dimensional environment, and are mimicking EMC network, specially by their micro/nanoscale architecture [172], offering a larger area for EMC constituent proteins, and more binding sites for cell membrane receptors, and adhesion molecules, growth factors, genes, immunomodulatory agents, and external stimuli (electrical, or magnetic pulses), which are delivered simultaneously to target sites after scaffold implantation for promotion of new healthy tissue [173]. No ideal mesh/scaffold for PFDs exists to day, and one may choose the mesh/scaffold according to patient history, and implants’ properties, such as material type: natural (as purified human/animal collagen, chitosan, gelatin, elastin, fibrin, silk, and fibronectin for human eSMCs [174, 175], or synthetic, or by the old criteria used for PP meshes, as pore sizes, mesh’s weight, and the host tissue response to mesh/scaffold material or to eMSCS/ERCs, understanding that reaction to implant materials are crucial for balance between their own elasticity and stiffness, vaginal tissue capacity of strain, which are absent when PP meshes are used. Novel blends of electrospun synthetic and natural polymers combined with eMSC in new generation of implants show that this approach promotes host cell infiltration and slows biomaterial degradation that has potential to strengthen the vaginal wall during healing [164, 165], actioning like intrinsic ECM, but with some limitation regarding small pore size of electrospun nanofiber meshes and toxic organic solvents used for their production [12]. /Human eMSCs/MenSCs modulate host tissue response to implanted materials, by stimulating tissue proper stem cells proliferation, their own high proliferation rate [128], and scaffolds’ eMSCs infiltration, and constituents of their secretome as growth factors, enzymes as MMPs—important for elastin postpartum recovery [128], influence mesh mechanical behavior after implantation [166], by fibrosis and FBRs reducing, when nondegradable polyamide mesh implant, through influencing macrophage polarization switching from an M1 to M2 phenotype, as in rodent and ovine models [166, 176, 177]. eMSCs seeded in degradable nano/microstructured meshes improve mesh tissue integration, eMSCs are entraped over 6 weeks in vivo, by cells with immunomodulatory effects, and by increasing local angiogenesis reduce FBRs to mesh implanted in mice with POP [166, 167, 169], and induce an up-regulation of M2 markers—as CD206 and Arg1, Mrc1, and Il10 genes in host tissue macrophages, parallel to reduction of cellular infiltration and secretion of inflammatory cytokines Il-1β and Tnf-α [175].

The Australian researchers [140] appreciate that tissue engineered mesh inserted transvaginally in large animal models will aid the validation of these constructs prior to clinical translation by assessing their integration with host tissue, and FBRs through histological analysis, immunoassays and gene profiling. Research has commenced with the completion of multiple xenogenic small and large animals studies assessing eMSC/PAG constructs, as mentioned above [166]. These animal models will be crucial in further assessing the efficacy of locally delivered eMSC and further determination of their action mechanisms. Based on these findings, multiple heterologous small and large animal studies are underway to assess the efficacy of other biomimetic degradable materials such as PLCL and 3D-PCL meshes seeded with eMSC/MenSCs.

Advertisement

5. Future proposals for stem cell prevention and therapy in pelvic floor disorders

The Australian research teams recognize some unknown data about eMSCs/MenSCs, mediation on cellular migration and recruitment by their paracrine effects, how eMSCs mediate M2 immunomodulatory responses during the FBR after implantation of bioengineered constructs. One discusses eMSCs secretome constituents as future associated to new generation implants for tissue engineering in PFDs. The Canadian and North American and Chinese researchers recognize the high financial burden for the studies and introduction in human clinical practice according to legislation issues which are incomplete resolved in Europe and North America, in comparison to Australia and Japan.

Advertisement

Conflict of interest

None.

References

  1. 1. Emersson JS, Gargett EC. Endometrial mesenchymal stem cells as a cell based therapy for pelvic organ prolapse. World Journal of Stem Cells. 2016;8(5):202-215. DOI: 10.4252/wjsc.v8.i5.202
  2. 2. Macowski SJ, Jurnalov C, Phelps JY. Use of vaginal mesh in the face of recent FDA warning and litigation. American Journal of Obstetrics & Gynecology. 2010;203(2):103.e1-103.e4
  3. 3. Holt E. US FDA rules manufacturers to stop selling mesh devices. Lancet. 2019;393:1686. DOI: 10.1016/S0140-6736(19)30938-9
  4. 4. Wosczyna MN, Konishi CT, Perez Carbajal EE, Wang TT, Rando TA, et al. Mesenchymal stromal cells are required for regeneration and homeostatic maintenance of skeletal muscle. Cell Reports. 2019;27:2029-2035.e5. DOI: 10.1016/j.celrep.2019.04.074
  5. 5. Schwab KE, Gargett CE. Co-expression of two perivascular cell markers isolates mesenchymal stem-like cells from human endometrium. Human Reproduction. 2007;22(11):2903-2911
  6. 6. Masuda H. A novel marker of human endometrial mesenchymal stem like cells. Cell Transplantation. 2012;21(10):2201-2214. DOI: 10.3727/096368911X37362
  7. 7. Miceli V, Bulati M, Iannolo G, Zito G, Gallo A, Conaldi PG. Therapeutic properties of mesenchymal stromal/stem cells: the need of cell priming for cell-free therapies in regenerative medicine. International Journal of Molecular Sciences. 2021;22(2):763. DOI: 10.3390/ijms22020763
  8. 8. Alvites R, Branquinho M, Sousa CA, Lopes B, Sousa P, Mauricio AC. Mesenchymal stem/stromal cells and their paracrine activity-immunomodulation mechanisms and how to influence the therapeutic potential. Pharmaceutics. 2022;14(2):381. DOI: 10.3390/pharmaceutics14020381
  9. 9. Gurung S, Werkmeister JA, Gargett CE. Inhibition of transforming growth factor-beta receptor signaling promotes culture expansion of undifferentiated human endometrial mesenchymal stem/stromal cells. Scientific Reports. 2015;5:15042. DOI: 10.1038/srep15042
  10. 10. Cruz M, Dissaranan C, Cotleur A, Kiedrowski M, Damaser M, et al. Pelvic organ distribution of mesenchymal stem cells injected intravenously after simulated childbirth injury in female rats. Obstetrics and Gynecology International. 2012;2012:612946. DOI: 10.1155/2012/612946
  11. 11. Ben Menachem-Zidon O, Gropp M, Ben Shushan E, Reubinoff B, Shveiky D. Systemically transplanted mesenchymal stem cells induce vascular-like structure formation in a rat model of vaginal injury. PLoS One. 2019;14(6):e0218081
  12. 12. Yuan M, Hu M, Dai F, Fan Y, Cheng Y, et al. Application of synthetic and natural polymers in surgical mesh for pelvic floor reconstruction. Materials & Design. 2021;209:109984
  13. 13. Edwards SL, Werkmeister JA, Rosamilia A, Ramshaw JAM, White JF, et al. Characterisation of clinical and newly fabricated meshes for pelvic organ prolapse repair. Journal of the Mechanical Behavior of Biomedical Materials. 2013;23:53-61
  14. 14. von Bahr L, Batsis I, Moll G, Hagg M, Le Blanc K, et al. Analysis of tissue following mesenchymal stromal cell therapy in humans indicates limited long term engraftment and no ectopic tissue formation. Stem Cells. 2012;30(7):1575-1578. DOI: 10.1002/stem.1118
  15. 15. Haylen BT, de Ridder D, Freeman RM, Swift SE, Berghmans B, et al. An international Urogynecological association (IUGA)/international continence society (ICS) joint report on the terminology for female pelvic floor dysfunction. Neurourology and Urodynamics. 2010;29:4-20
  16. 16. DeLancey JO. Anatomy and biomechanics of genital prolapse. Clinical Obstetrics and Gynecology. 1993;36:897-909
  17. 17. Westergren Söderberg M, Byström B, Kalamajski S, Malmstörm A, Ordeberg G-E. Gene expression of small leucine-rich repeat proteoglycans and fibulin are decreased in pelvic organ prolapse. Molecular Human Reproduction. 2009;15(4):251-257
  18. 18. Wu MJ, Matthews AC, Conover MM, Pate V, Jonsson Funk M. Lifetime risk of stress urinary incontinence or pelvic organ prolapse surgery. Obstetrics and Gynecology. 2014;123(6):1201-1206. DOI: 10.1097/AOG.0000000000000286
  19. 19. National Institute of health consensus: Consensus development conference. Urinary incontinence in adults. Journal of the American Geriatrics Society. 1990;38:265-276
  20. 20. Nussler E, Granäsen G, Bixo M, Löfgren M. Long term outcome after routine surgery for pelvic organ prolapse. A national register-based cohort study. International Urogynecology Journal. 2022. DOI: 10.1007/s00192-022-05156-y
  21. 21. Smith FJ, Holman CD, Moorin RE, Tsokos N. Lifetime risk of undergoing surgery for pelvic organ prolapsed. Obstetrics and Gynecology. 2010;116(5):1096-1100. DOI: 10.1097?AOG.0b01363181f3729
  22. 22. Dieter AA, Wilkins MF, Wu JM. Epidemiological trends and future care needs for pelvic floor disorders. Current Opinion in Obstetrics and Gynecology. 2015;27:380-384
  23. 23. Dooley Y et al. Urinary incontinence prevalence: Results from the National Health and nutrition examination survey. The Journal of Urology. 2008;179(2):656-671
  24. 24. McIntosh LJ, Mallett VT, Frahm JD, Richardson DA, Evans MI. Gynecologic disorders in women with ehlers-danlos syndrome. Journal of the Society for Gynecologic Investigation. 1995;2:559-564
  25. 25. Norton PA, Baker JE, Sharp HC, Warenski JC. Genitourinary prolapse and joint hypermobility in women. Obstetrics and Gynecology. 1995;85:225-228
  26. 26. Allen-Brady K, Bortolini MAT, Damaser MS. Mouse knockout models for pelvic organ prolapse, a systematic review. International Urogynecology Journal. 2022. DOI: 10.1007/s00192-021-05066-5
  27. 27. Subak LL, Waetjen LE, van den Eeden S, Thom DH, Brown JS, et al. Cost of pelvic organ prolapse surgery in the United States. Obstetrics and Gynecology 2001;98(4):646-651. DOI: 10.1016/S0029-7844(01)01472-7
  28. 28. Van Geelen JM, Dwyer PL. Where to for pelvic organ prolapsed treatment after FDA pronouncements? A systematic review of the recent literature. International Urogynecology Journal. 2013;5:707-718. DOI: 10.1007/s00192-012-2025-3
  29. 29. Goessens E, Deriemaeker H, Cammu H. The use of vaginal pessary to decide whether a mid urethral sling should be added to prolapsed surgery. The Journal of Urology. 2020;203(3):593-603. DOI: 10.1097/JU.0000000000000575
  30. 30. Dobrokhotova EY, Nagieva T, Kareva EN. Focused radiofrequency treatment of postpartum vaginal relaxation syndrome. Obstetrics Gynecology and Reproduction. 2020;14(4):437-446
  31. 31. Altman D, Vayrynen T, Engl ME, Axelsen S, Falconer C. For Nordic transvaginal mesh group: Anterior colporrhaphy versus transvaginal mesh for pelvic-organ prolapse. New England Journal of Medicine. 2011;364(19):1826-1836. DOI: 10.1056/NEJMoa1009521
  32. 32. Chapple CR, MacNeil S. The use of implanted materials for treating women with pelvic organ prolapse and stress urinary incontinence. Current Opinion in Urology. 2019;29(4):431-436. DOI: 10.1097/MOU.0000000000000619
  33. 33. Abhari ER, Izett-Kay LM, Morris LH, Cartwright R, Snelling JBS. Host-biomaterial interactions in mesh complications after pelvic floor reconstructive surgery. Nature Reviews Urology. 2021;18(12):725-738. DOI: 10.1038/s41585-021-00511-y
  34. 34. Feola A, Abramowitch S, Jallah Z, Stein S, Moalli P, et al. Deterioration in biomechanical properties of the vagina following implantation of a high-stiffness prolapse mesh. BJOG. 2013;120:224-232
  35. 35. Alas AN, Chinthakanan O, Espaillat L, Davila GW, Aguilar VC, et al. De novo stress urinary incontinence after pelvic organ prolapse surgery in women without occult incontinence. International Urogynecology Journal. 2017;28(4):583-590. DOI: 10.1007/s00192-016-3149-7
  36. 36. Weemhoff M, Vergeldt TF, Notten K, Serroyen J, Roumen FJ, et al. Avulsion of puborectalis muscle and other risk factors for cystocele recurrence: A 2-year follow-up study. International Urogynecology Journal. 2012;23(1):65-71. DOI: 10.1007/s00192-011-1524-y
  37. 37. Quiroz LH, Gutman RE, Shippey S, Cundiff GW, Handa VL, et al. Abdominal sacrocolpopexy: Anatomic outcomes and complications with Pelvicol, autologous and synthetic graft materials. American Journal of Obstetrics and Gynecology. 2008;198(5):557. e1-557. e5
  38. 38. Maher C, Feiner B, Baessler K, Adams EJ, Hagen S, Glazener CM. Surgical management of pelvic organ prolapse in women. Cochrane Database of Systematic Reviews. 2013;4:CD004014. DOI: 10.1002/14651858.CD004014.pub5
  39. 39. Morling JR, Mc Allister AD, Agur W, Fischbacher MC, Wood R, et al. Adverse events after first, single, mesh and non-mesh surgical procedures for stress urinary incontinence and pelvic organ prolapse in Scotland, 1997-2016: A population-based cohort study. Lancet. 2017;389(10069):629-640
  40. 40. Pascal G, Hernandez Gascon B, Rodriguez M, Bellon JM, et al. The long-term behavior of lightweight and heavyweight meshes used to repair abdominal wall defects is determined by the host tissue repair process provoked by the mesh. Surgery. 2012;152:886-895
  41. 41. Ulrich D, Edwards SL, Su K, Tan KS, White JF, et al. Human endometrial mesenchymal stem cells modulate the tissue response and mechanical behaviour of polyamide mesh implants for pelvic organ prolapse repair. Tissue Engineering. 2013;20:785-798
  42. 42. Vashaghian M, Zaat SJ, Smit TH, Roovers JP. Biomimetic implants for pelvic floor repair. Neurourology and Urodynamics. 2018;37:566-580
  43. 43. Thompson P. The myology of the pelvic floor, Newton: McCorquodale, 1899, cited by Raizada V, Mittal KM. Pelvic floor anatomy and applied physiology. Gastroenterology Clinics of North America. 2008;37(3):493-vii. DOI: 101016jgec.2008.06.003
  44. 44. Rynkevic R, Martins P, Hympanova L, Deprest J, et al. Biomechanical and morphological properties of the multiparous ovine vagina and effect of subsequent pregnancy. Journal of Biomechanics. 2017;57:94-102. DOI: 10.1016/j.biomech.2017.03.023
  45. 45. Ulrich D, Edwards SL, Su K, White JE, Gargett CE, et al. Influence of reproductive status on tissue composition and biomechanical properties of ovine vagina. PLoS One. 2014;9(4):e93172. DOI: 10.1371/journal;pone.0093172
  46. 46. de Landsheere L, Brieu M, Blacher S, Munaut C, Cosson M, et al. Elastin density. Link between histological and biomechanical properties of vaginal tissue in women with pelvic organ prolapse? International Urogynecology Journal. Apr 2016;27(4):629-635. DOI: 10.1007/s00192
  47. 47. Jameson S, Swaminathan G, Dahal S, Court B, Damaser MS, et al. Elastin homeostasis is altered with pelvic organ prolapsed in cultures of vaginal cells from lysiloxydase like-1 knockout mice model physiological reports. 2020;8(11):e14436
  48. 48. Dahal S, Kuang M, Rietsch A, Damaser MS, et al. Quantitative morphometry of elastic fibers in pelvic organ prolapse. Annals of Biomedical Engineering. 2021;49(8):1909-1922. DOI: 10.1007/s10439-021-02760-9
  49. 49. Rubod C, Boukerrou M, Brieu M, Dubois P, Cosson M. Biomechanical properties of vaginal tissue. Part 1: New experimental protocol. The Journal of Urology. 2007;178:320-325
  50. 50. Rubod C, Boukerrou M, Brieu M, Jean-Charles C, Dubois P, et al. Biomechanical properties of vaginal tissue: Preliminary results. International Urogynecology Journal. 2008;19:811-816
  51. 51. Jean-Charles C, Rubod C, Brieu M, Boukerrou M, Fasel J, et al. Biomechanical properties of prolapsed or non-prolapsed vaginal tissue: Impact on genital prolapse surgery. International Urogynecology Journal. 2010;21:1535-1538
  52. 52. Rubod C, Brieu M, Cosson M, Rivaux G, Clay J-C, et al. Biomechanical properties of human pelvic organs. The Journal of Urology. 2012;79(968):e917-e922
  53. 53. Shin SJ, Yanagisawa H. Recent updates on the molecular network of elastic fiber formation. Essays in Biochemistry. 2019;63(3):395-376. DOI: 10.1042/EBC20180052
  54. 54. Hulmes DJS. Building collagen molecules, fibrils, and suprafibrillar structures. Journal of Structural Biology. 2002;137:2-10. DOI: 10.1006/jsbi.2002.4450
  55. 55. Svensson L, Narlid I, Oldberg A. Fibromodulin and lumican bind to the same region on collagen type I fibrils. FEBS Letters. 2000;470:178-182
  56. 56. Couchman JR, Pataki CA. An introduction to proteoglycans and their localization. Journal of Histochemistry & Cytochemistry. 2012;60:885-897
  57. 57. Li L, Xie T. Stem cell niche: Structure and function. Annual Review of Cell and Developmental Biology. 2005;21:605-631
  58. 58. Moore KA, Lemischka IR. Stem cells and their niches. Science. 2006;311:1880-1885
  59. 59. de Vega S, Iwamoto T, Yamada Y. Fibulins: Multiple roles in matrix structures and tissue functions. Cellular and Molecular Life Sciences. 2009;66(11-12):1890-1902. DOI: 10.1007/s00018-009-8632-6
  60. 60. Yanagisawa H, Davis EC. Unraveling the mechanism of elastic fiber assembly: The roles of short fibulins. The International Journal of Biochemistry & Cell Biology. 2010;42(7):1084-1093. DOI: 10.1016/j.biocel.2010.03.009
  61. 61. Drewes PG, Yanasgisawa H, Starcher B, Hornstra I, Word RA, et al. Pelvic organ prolapse in fibulin-5 knockout mice: Pregnancy-induced changes in elastic fiber homeostasis in mouse vagina. The American Journal of Pathology. 2007;170(2):578-589. DOI: 10.2353/ajpath.2007.060662
  62. 62. Rahn DD, Ruff MD, Brown SA, Tibbals HF, Word RA. Biomechanical properties of the vaginal wall: Effect of pregnancy, elastic fiber deficiency, and pelvic organ prolapse. American Journal of Obstetrics and Gynecology. 2008;198(590):e591-e596
  63. 63. Rahn DD, Acevedo FJ, Roshanravan S, Keller PW, Word RA, et al. Failure of pelvic organ support in mice deficient in fibulin-3. The American Journal of Pathology. 2009;174(1):206-215. DOI: 10.2353/ajpath.2009.080212
  64. 64. Liu X, Zhao Y, Gao J, Pawlyk B, Li T, et al. Elastic fiber homeostasis requires lysyl oxidase-like 1 protein. Nature Genetics. 2004;36(2):178-182. DOI: 10.1038/ng1297
  65. 65. Ferreira JR, Teixeira GQ , Santos SG, Barbosa MA, Goncalves RM, et al. Mesenchymal stromal cell secretome: Influencing therapeutic potential by cellular pre-conditioning. Frontiers in Immunology. 2018;9:2837. DOI: 10.3389/fimmu.2018.02837
  66. 66. Svensson K, Bortolini MAT, Damaser MS. Mouse knockout models for pelvic organ prolapse, a systematic review. International Urogynecology Journal. 2022. DOI: 10.1007/s00192-021-05066-5
  67. 67. Bruel A, Ortoft G, Oxlund H. Inhibition of cross-links in collagen is associated with reduced stiffness of the aorta in young rats. Atherosclerosis. 1998;140(1):135-145. DOI: 10.1016/S0021-9150(98)00130-
  68. 68. Edwall L, Carlström K, Fianu Jonasson A. Markers of collagen synthesis and degradation in urogenital tissue and serum from women with and without uterovaginal prolapse. Molecular Human Reproduction. 2008;14(3):193-197
  69. 69. Moalli PA, Shand SH, Zyczynski HM, Gordy SC, Meyn LA. Remodeling of vaginal connective tissue in patients with prolapse. Obstetrics and Gynecology. 2005;106(5 pt 1):953-963
  70. 70. Phillips CH, Anthony F, Benyon C, Monga AK. Collagen metabolism in the uterosacral ligaments and vaginal skin of women with uterine prolapse. BJOG : An International Journal of Obstetrics and Gynaecology. 2006;113(1):39-46
  71. 71. Connell KA, Guess MK, Chen HW, Lynch T, Taylor HS, et al. HOXA11 promotes fibroblast proliferation and regulates p53 in uterosacral ligaments. Reproductive Sciences. 2009;16(7):694-700
  72. 72. Bhattarai A, Staat M. Modeling of soft connective tissue to investigate female pelvic floor dysfunction. Computational and Mathematical Methods in Medicine. 2018:9518076
  73. 73. Catanzarite T, Bremner S, Barlow C, Bou-Malham L, Alperin M, et al. Pelvic muscles’ mechanical response to strains in the absence and presence of pregnancy-induced adaptations in a rat model. American Journal of Obstetrics & Gynecology. 2018;210(5):512.e1-512.e9. DOI: 10.1016/j.ajog.2018.02.001
  74. 74. Bartolini MA, Drutz HP, Lovatsis D, Alarab M. Vaginal delivery and pelvic floor dysfunction, current evidence and implications to future research. International Urogynecology Journal. 2010;21(8):1025-1030
  75. 75. Lien KC, Mooney B, DeLancey JO, Aston-Miller JA. Levatorani muscle stretch induced by simulated vaginal birth. Obstetrics and Gynecology. 2004;103(1):31-40
  76. 76. Kroffta L, Otcenasek M, Kasikova E, Feyereisl J. Pubococcgygeus-puborectalis trauma after forceps delivery-evaluation of levatorani muscle with 3D/4D ultrasound. International Urogynecology Journal and Pelvic Floor Dysfunction. 2009;20:1175-1181
  77. 77. Rautzang RM, Rostaminia G, Moalli AP, Abramovitch S. Pelvic floor shape variations during pregnancy and after vaginal delivery. Computer Methods and Programs in Biomedicine. 2020;194:105516. DOI: 10.1016/J.cmpb.2020.105516
  78. 78. Rynkevic R. The effect of consecutive pregnancies on the ovine pelvic soft tissue. Link between biomechanical and histological components. Annals of Anatomy. 2019;222:166-172. DOI: 10.1016/j.aanat.2018.12.002
  79. 79. Alperin M, Feola A, Duerr R, Moalli P, Abramavowitch S. Pregancy and delivery-induced biomechanical changes in rat vagina persist postpartum. International Urogynecology Journal. 2010;21:1169-1174
  80. 80. Rechberger T, Skorupski P. The controversies regarding the role of estrogens in urogynecology. Folia Histochemica et Cytobiologica. 2007;45(Supp. 1):S:17-S:21. DOI: 10.2478/4476
  81. 81. Yu KR, Kang KS. Aging-related genes in mesenchymal stem cells: A mini review. Gerontology. 2013;59:557-563. DOI: 10.1159/000353857
  82. 82. Schulman IH, Balkan W, Hare JM. Mesenchymal stem cell therapy for aging frailty. Frontiers in Nutrition. 2018;5:108. DOI: 10.3389/fnut.2018.00108
  83. 83. Hillard T. The postmenopausal bladder. Menopause International. 2010;16(2):74-80. DOI: doi; 10.1258/mi.2010.010020
  84. 84. Robinson D, Toozs Hobson P, Cardozo L. The effect of hormones on the lower urinary tract. Menopause International. 2013;19(1):155-162. DOI: doi; 10.1177/1754045511398
  85. 85. Sousa-Victor P, Gutarra S, Garcia-Prat L, Rodriguez-Ubreva J, Serrano AL, et al. Geriatric muscle stem cells switch reversible quiescence into senescence. Nature. 2014;506:316-321. DOI: 10.1038/nature13013
  86. 86. Beane OS, Fonseca VC, Cooper LL, et al. Impact of aging on the regenerative properties of bone marrow, muscle-, and adipose-derived mesenchymal stem/stromal cells. PLoS One. 2014;9:e115963
  87. 87. Petros P, Woodman JP. The integral theory of continence. International Urogynecology Journal. 2008;19(1):35-40. DOI: 10.1007/s00192-007-0475-9
  88. 88. Tran C, Damaser SM. The potential role of stem cells in the treatment of urinary incontinence. Therapeutic Advances in Urology. 2015;7(1):22-40
  89. 89. Furuta A, Jankowski R, Honda M, Pruchnic R, Chancellor M, et al. State of the art of where we are at using stem cells for stress urinary incontinence. Neurourology and Urodynamics. 2007;26:966-971
  90. 90. Wang H, Chuang Y, Chancellor M. Development of cellular therapy for the treatment of stress urinary incontinence. International Urogynecology Journal. 2011;22:1075-1083
  91. 91. Gargett CE, Gurung S, Darzi S, Werkmeister JA, Mukherjee S. Tissue engineering approaches for treating pelvic organ prolapse using a novel source of stem/stromal cells and new materials. Current Opinion in Urology. 2019;29:450-457. DOI: doi; 10.1097/mou.0000000000000634
  92. 92. Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringden O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Experimental Hematology. 2003;31:890-896. DOI: 10.1016/S0301-472X(03)00110-3
  93. 93. Zhao Q , Gregory CA, Lee RH, Reger RL, McNeill E, et al. MSCs derived from iPSCs with a modified protocol are tumor-tropic but have much less potential to promote tumors than bone marrow MSCs. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:530-535. DOI: 10.1073/pnas.1423008112
  94. 94. Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells. 1978;4:7-25
  95. 95. Darzi S, Werkmeister JA, Deane JA, Gargett CE. Identification and characterization of human endometrial mesenchymal stem/stromal cells and their potential for cellular therapy. Stem Cells Translational Medicine. 2016;5:1127-1132. DOI: 10.5966/sctm.2015-0190
  96. 96. Barker N. Adult intestinal stem cells: Critical drivers of epithelial homeostasis and regeneration. Nature Reviews. Molecular Cell Biology. 2014;15:19-33. DOI: 10.1038/nrm3721
  97. 97. Brack AS, Rando TA. Tissue-specific stem cells: Lessons from the skeletal muscle satellite cell. Cell Stem Cell. 2012;10:504-514. DOI: 10.1016/j.stem.2012.04.001
  98. 98. Joe AW, Yi L, Natarajan A, Le Grand F, Rossi FM, et al. Muscle injury activates resident fibro/adipogenic progenitors that facilitate myogenesis. Nature Cell Biology. 2010;12:153-163. DOI: 10.1038/ncb2015
  99. 99. Caplan AI. Mesenchymal stem cells: Time to change the name! Stem Cells Translational Medicine. 2017;6(6):1445-1451. DOI: 10.1002/sctm.17-0051
  100. 100. Wilson A, Trumpp A. Bone-marrow haematopoietic-stem-cell niches. Nature Reviews. Immunology. 2006;6:93-106
  101. 101. Maacha S, Sidahmed H, Jacob S, Gentilcore G, Cugno C, et al. Paracrine mechanisms of mesenchymal stromal cells in angiogenesis. Stem Cells International. 2020:4356359. DOI: 10.1155/2020/4356359
  102. 102. Fu X, Liu G, Halim A, Ju Y, Song AG, et al. Mesenchymal stem cell migration and tissue repair. Cell. 2019;8:784. DOI: 10.3390/cells8080784
  103. 103. Xi J, Yan X, Zhou J, Yue W, Pei X. Mesenchymal stem cells intissue repairing and regeneration: Progress and future. Burns & Trauma. 2013;1:13-20
  104. 104. Karp JM, LengTeo GS. Mesenchymal stem cell homing: The devil is in the details. Cell Stem Cell. 2009;4:206-216
  105. 105. Ankrum JA, Ong JF, Karp JM. Mesenchymal stem cells: Immune evasive, not immune privileged. Nature Biotechnology. 2014;32(3):252-260
  106. 106. Rastegar F, Shenaq D, Huang J, et al. Mesenchymal stem cells: Molecular characteristics and clinical applications. World Journal of Stem Cells. 2010;2:67-80
  107. 107. Caplan AI. MSCs: The sentinel and safe-guards of injury. Journal of Cellular Physiology. 2016;231:1413-1416. DOI: 10.1002/jcp.25255
  108. 108. Sanchez-Mata A, Gonzales-Muňoz E. Understanding menstrual blood-derived stromal/stem cells: Definition and properties. Are we rushing into their therapeutic applications? Science. 2021;24(12):103501
  109. 109. Evans J, Kaitu'u-Lino T, Salamonsen LA. Extracellular matrix dynamics in scar-free endometrial repair: Perspectives from mouse in vivo and human in vitro studies. Biology of Reproduction. 2011;85:511-523
  110. 110. Henriet P, Gaide Chevronnay HP, Marbaix E. The endocrine and paracrine control of menstruation. Molecular and Cellular Endocrinology. 2012;358:197-207
  111. 111. Prianishnikov VA. On the concept of stem cell and a model of functional-morphological structure of the endometrium. Contraception. 1978;18(3):213-223
  112. 112. Padykula HA, Coles LG, Kaiserman-Abramof IR, et al. The basalis of the primate endometrium: A bifunctional germinal compartment. Biology of Reproduction. 1989;40:681-690
  113. 113. Chan RW, Schwab KE, Gargett CE. Clonogenicity of human endometrial epithelial and stromal cells. Biology of Reproduction. 2004;70:1738-1750
  114. 114. Gargett CE, Masuda H. Adult stem cells in the endometrium. Molecular Human Reproduction. 2010;16:818-834. DOI: 10.1093/molehr/gaq061
  115. 115. Gargett CE, Schwab KE, Deane JA. Endometrial stem/progenitor cells: The first 10 years. Human Reproduction Update. 2016;22:137-163
  116. 116. Kato K, Yoshimoto M, Kato K, Adachi S, Wake N, et al. Characterization of side-population cells in human normal endometrium. Human Reproduction. 2007;22:1214-1223
  117. 117. Schwab KE, Chan RW, Gargett CE. Putative stem cell activity of human endometrial epithelial and stromal cells during the menstrual cycle. Fertility and Sterility. 2005;84(Suppl. 2):1124-1130
  118. 118. Chan RW, Gargett CE. Identification of label-retaining cells in mouse endometrium. Stem Cells. 2006;24:1529-1538
  119. 119. Cervello I, Martinez-Conejero JA, Horcajadas JA, Pellicer A, Simón C. Identification, characterization and co-localization of label-retaining cell population in mouse endometrium with typical undifferentiated markers. Human Reproduction. 2007;22(1):45-51
  120. 120. Letouzey V, Tan KS, Deane JA, Ulrich D, Gargett CE, et al. Isolation and characterisation of mesenchymal stem/stromal cells in the ovine endometrium. PLoS One. 2015;10:e0127531
  121. 121. Dimitrov R, Timeva T, Kyurkchiev D, Stamenova M, Kyurkchiev S, et al. Characterization of clonogenic stromal cells isolated from human endometrium. Reproduction. 2008;135:551-558
  122. 122. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Horwitz E, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315-317
  123. 123. Wolff FE, Wolff BA, Du H, Taylor SH. Demonstration of multipotent stem cells in the adult human endometrium by in vitro chondrogenesis. Reproductive Sciences. 2007;14(6):524-533
  124. 124. Gargett CE, Schwab KE, Zillwood RM, Nguyen HP, Wu D. Isolation and culture of epithelial progenitors and mesenchymal stem cells from human endometrium. Biology of Reproduction. 2009;80:1136-1145
  125. 125. Taylor HS. Endometrial cells derived from donor stem cells in bone marrow transplant recipients. JAMA. 2004;292:81-85
  126. 126. Du H, Taylor SH. Stem cells and female reproduction. Reproductive Sciences. 2009;16(2):126-139. DOI: 10.1177/1933719108329956
  127. 127. Najafi-Ghalehlou N, Feizkhah A, Mobayen M, Pourmohammadi-Bejarpasi Z, Roudkenar MH, et al. Plumping up cushion of human biowaste in regenerative medicine, novel insights into a state-of the art reserve arsenal. Stem Cell Reviews and Reports. 2022
  128. 128. Meng X, Ichim ET, Zhong J, Rogers A, Riordan HN, et al. Endometrial regenerative cells: A novel stem cell population. Journal of Translational Medicine. 2007;5:57. DOI: 10.1186/1479-5876-5- 57
  129. 129. Ulrich D, Muralitharan R, Gargett CE. Toward the use of endometrial and menstrual blood mesenchymal stem cells for cell-based therapies. Expert Opinion on Biological Therapy. 2013;13(10):1387-1400. DOI: 10.157/147125982013.826187
  130. 130. Darzi S, Zarnani AH, Jeddi-Tehrani M, Entezami K, Kazemnejad S, et al. Osteogenic differentiation of stem cells derived from menstrual blood versus bone marrow in the presence of human platelet releasate. Tissue Engineering. 2012;18:1720-1728
  131. 131. Patel A, Silva F. Menstrual blood stromal cells: The potential for regenerative medicine. Regenerative Medicine. 2008;3(4):43-44
  132. 132. Zhang S, Li P, Yuan Z, Tan J. Platelet-rich plasma improves therapeutic effects of menstrual blood-derived stromal cells in rat model of intrauterine adhesion. Stem Cell Research & Therapy. 2019;10:61
  133. 133. Feng P, Li P, Tan J. Human menstrual blood-derived stromal cells promote recovery of premature ovarian insufficiency via regulating the ECM-dependent FAK/AKT signaling. Stem Cell Reviews and Reports. 2019;15:241-255
  134. 134. Chen L, Qu J, Cheng T, Chen X, Xiang C. Menstrual blood-derived stem cells: Toward therapeutic mechanisms, novel strategies, and future perspectives in the treatment of diseases. Stem Cell Research & Therapy. 2019;10:406
  135. 135. Dalirfardouei R, Jamialahmadi K, Jafarian AH, Mahdipour E. Promising effects of exosomes isolated from menstrual blood-derived mesenchymal stem cell on wound-healing process in diabetic mouse model. Journal of Tissue Engineering and Regenerative Medicine. 2019;13:555-568
  136. 136. Schwab KE, Hutchinson P, Gargett CE. Identification of surface markers for prospective isolation of human endometrial stromal colony-forming cells. Human Reproduction. 2008;23:934-943
  137. 137. Crişan M, Corselli M, Chen CWW, Péault B. Perivascular cells for regenerative medicine. Journal of Cellular and Molecular Medicine. 2012;16(12):2851-2860. DOI: 10.1111/j.1582-4934.2012.01617.x
  138. 138. Zhu X, Yu F, Yan G, Hu Y, Sun H, Ding L. Human endometrial perivascular stem cells exhibit a limited potential to regernerate endometrium after xenotransplantation. Human Reproduction. 2021;36(1):145-159. DOI: 10.1093/humrep/deaa261
  139. 139. Gurung S, Ulrich D, Sturm M, Rosamilia A, Werkmeister JA, Gargett CE. Comparing the effect of TGF-β receptor inhibition on human perivascular mesenchymal stromal cells derived from endometrium, bone marrow and adipose tissues. Journal of Personalized Medicine. 2020;10:261. DOI: 10.3390/jpm10040261
  140. 140. Hennes DMZB, Rosamilia A, Werkmeister JA, Gargett CE, Mukherjee S. Endometrial SUSD2+ mesenchymal stem/stromal cells in tissue engineering: Advances in novel cellular constructs for pelvic organ prolapse. Journal of Personalized Medicine. 2021;11(9):840. DOI: 10.3390/jpm11090840
  141. 141. Marinaro F, Gomez-Serrano M, Jorge I, Silla-Castro JC, Casado JG, et al. Unraveling the molecular signature of extracellular vesicles from endometrial-derived mesenchymal stem cells: Potential modulatory effects and therapeutic applications. Frontiers in Bioengineering and Biotechnology. 2019;7:431
  142. 142. Marinaro F, Macias-Garcia B, Sanchez-Margallo FM, Blazquez R, Vazquez J, et al. Extracellular vesicles derived from endometrial human mesenchymal stem cells enhance embryo yield and quality in an aged murine modeldagger. Biology of Reproduction. 2019;100:1180-1192
  143. 143. Dalirfardouei R, Jamialahmadi K, Mahdipour E. A feasible method for the isolation of mesenchymal stem cells from menstrual blood and their exosomes. Tissue & Cell. 2018;55:53-62
  144. 144. Lopez-Caraballo L, Martorell-Marugan J, Carmona-Saez P, Gonzalez-Munoz E. Analysis of menstrual blood stromal cells reveals SOX15 triggers oocyte-based human cell reprogramming. iScience. 2020;23:101376
  145. 145. Bozorgmehr M, Gurung S, Darzi S, Nikoo S, Kazemnejad S, Zarnani AH, et al. Endometrial and menstrual blood mesenchymal stem/stromal cells: Biological properties and clinical application. Frontiers in Cell and Development Biology. 2020;8:497
  146. 146. Tang L, Jiang Y, Zhu M, Chen L, Zhon X, Zhou C, et al. Clinical study using mesenchymal stem cells for the treatment of patients with severe COVID-19. Frontiers in Medicine. 2020;14(5):664-673
  147. 147. Bianco P, Cao X, Frenette PS, Mao JJ, Robey PG, Simmons PJ, et al. The meaning, the sense and the significance: Translating the science of mesenchymal stem cells into medicine. Nature Medicine. 2013;19:35-42
  148. 148. Turner L, Knoepfler P. Selling stem cells in the USA: Assessing the direct-to-consumer industry. Cell Stem Cell. 2016;19:154-157
  149. 149. Galipeau J, Sensebe L. Mesenchymal stromal cells: Clinical challenges and therapeutic opportunities. Cell Stem Cell. 2018;22(6):824-833
  150. 150. Herschorn S, Carr L, Birch C, et al. Autologous muscle derived cells as therapy fa or stress urinary incontinence: A randomized trial. Neurology and Urodynamics. 2010;12(2):307-312
  151. 151. Woo L, Hijaz A, Kuang M, Penn M, Damaser M, Rackley R. Over expression of stem cell homing cytokines in urogenital organs following vaginal distention. The Journal of Urology. 2007;177:1568-1572
  152. 152. Wood H, Kuang M, Woo L, Hijaz A, Butler R, Penn M, et al. Cytokine expression after vaginal distention of different durations in virgin Sprague-Dawley rats. The Journal of Urology. 2008;180:753-759
  153. 153. Sarkar D, Vemula PK, Zhao W, Gupta A, Karp M, et al. Engineered mesenchymal stem cells with self assembled vesicles for systemic cell targeting. Biomaterials. 2010;31(19):5266-5274
  154. 154. Li L, El-Hayek YH, Lin B, et al. Direct current electrical field guide neuronal stem/progenitor cell migration. Stem Cells. 2008;26(8):2193-2200
  155. 155. Lalu MM, Mcintyre L, Pugliese C, Fergusson D, Stewart DJ, et al. Safety of cell therapy with mesenchymal stromal cells (SafeCell): A systematic review and meta-analysis of clinical trials. PLoS One. 2012;2012(7):e47559
  156. 156. Nemeth K, Leelahavanichkul A, Yuen PS, Mayer B, Mezey E, et al. Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nature Medicine. 2009;15:42-49
  157. 157. Sensebe L, Fleury-Cappellesso S. Biodistribution of mesenchymal stem/stromal cells in a preclinical setting. Stem Cells International. 2013:678063. DOI: 10.1155/2013/678063
  158. 158. Kirchin V, Page T, Keegan EP, Om Atiemo K, Aluko P, et al. Urethral injection therapy for urinary incontinence in women. Cochrane Database of Systematic Reviews. 2017;7(7):CD003881. DOI: 10.1002/14651858. CD003881.pub4
  159. 159. Jankowski RJ, Tu LM, Carlson C, Robert M, Carr L, et al. A double-blind, randomized, placebo-controlled clinical trial evaluating the safety and efficacy of autologous muscle derived cells in female subjects with stress urinary incontinence. International Urology and Nephrology. 2018;50(12):2153-2165
  160. 160. Burdzinska A, Dybowski B, Zarvchta-Wiśniewska W, Paczek L, et al. Limited accuracy of transurethral and periurethralintrasphincteric injections of cellular suspension. Neurourology and Urodynamics. 2018;37(5):1612-1622
  161. 161. Sarveazad A, Newstead LG, Mirzaer R, Taghi-Joghataer M, Mahjoubi BL, et al. A new method for treating fecal incontinence by implanting stem cells derived from human adipose tissue: Preliminary findings of a randomized double-blind clinical trial. Stem Cell Research & Therapy. 2017;8(1):40
  162. 162. Atala A. Tissue engineering of human bladder. British Medical Bulletin. 2011;97:81-104. DOI: 10.1093/bmb/ldr003
  163. 163. Mukherjee S, Venugopal JR, Ravichandran R, Rmalingam M, Ramakrishna S, et al. Nanofiber technology for controlling stem cell functions and tissue engineering. Micro and Nanotechnologies in Engineering Stem Cells and Tissues. 2013;1:27-51. DOI: 10.1002/9781118574775.ch2
  164. 164. Aghaei-Ghareh-Bolagh B, Mukherjee S, Lockley KM, Gargett CE, Weiss AS, et al. A novel tropoelastin-based resorbable surgical mesh for pelvic organ prolapse repair. Materials Today Bio. 2020;8:100081. DOI: doi.org/10.1016/j.mtbio.2020.100081
  165. 165. Gurung S, Williams S, Deane JA, Westmeister JA, Gargett CE. The transcriptome of human endometrial mesenchymal stem cells under TGF betaR inhibition reveals improved potential for cell-based therapies. Frontiers in Cell and Development Biology. 2018;6:164. DOI: 10.3389/fcell.2018.00164
  166. 166. Ulrich D, Edwards SL, Su K, Tan KS, Gargett CE, et al. Human endometrial mesenchymal stem cells modulate the tissue response and mechanical behavior of polyamide mesh implants for pelvic organ prolapse repair. Tissue Engineering. Part A. 2014;20:785-798. DOI: 10.1089/ten.TEA.2013.0170
  167. 167. Mukherjee S, Darzi S, Rosamilia A, Kadam V, Werkmeister JA, et al. Blended nanostructured degradable mesh with endometrial mesenchymal stem cells promotes tissue integration and anti-inflammatory response in vivo for pelvic floor application. Biomacromolecules. 2019;20:454-468. DOI: 10.1021/acs.biomac.8b01661
  168. 168. Paul K, Darzi S, Mcphee G, Del Borgo MP, Gargett CE, et al. 3D bioprinted endometrial stem cells on melt electrospun poly epsilon-caprolactone mesh for pelvic floor application promote anti-inflammatory responses in mice. Acta Biomaterialia. 2019;97:162-176. DOI: 10.1016/j.actbio.2019.08
  169. 169. Paul K, Darzi S, Werkmeister JA, Gargett CE, Mukherjee S. Emerging nano/micro-structured degradable polymeric meshes for pelvic floor reconstruction. Nanomaterials (Basel). 2020;10(6):1120. DOI: 10.3390/nano10061120
  170. 170. Mukherjee S, Darzi S, Paul K, Cousins FL, Werkmeister JA, Gargett CE. Electrospun nanofiber meshes with endometrial MSCs modulate foreign body response by increased angiogenesis, matrix synthesis, and anti-inflammatory gene expression in mice: Implication in pelvic floor. Frontiers in Pharmacology. 2020;11:353. DOI: 10.3389/fphar.2020.00353
  171. 171. Vashaghian M, Ruiz-Zapata AM, Kerhof MH, Smit TH, et al. Toward a new generation of pelvic floor implants with electrospunnanofibrous matrices. A feasibility study. Neurourology and Urodynamics. 2017;36(3):565-575. DOI: 10.1002/nau.22969
  172. 172. Sridharan I, Ma Y, Kim T, Kobak W, Wang R, et al. Structural and mechanical profiles of native collagen fibers in vaginal wall connective tissue. Biomaterials. 2012;33:1520-1527. DOI: 10.1016/j.biomaterials.2011.11.005
  173. 173. Muzzio N, Moya S, Romero G. Multifunctional scaffolds and synergistic strategies in tissue engineering and regenerative medicine. Pharmaceutics. 2021;13(6):792. DOI: 10.3390/pharmaceutics13060792
  174. 174. Rajaraman G, White J, Tan KS, Ulrich D, Gargett CE, et al. Optimization and scale-up culture of human endometrial multipotent mesenchymal stromal cells: Potential for clinical application. Tissue Engineering. Part C, Methods. 2013;19:80-92
  175. 175. Su K, Edwards SL, Tan KS, White JF, Gargett CE, Werkmeister JA, et al. Induction of endometrial mesenchymal stem cells into tissue-forming cells suitable for fascial repair. Acta Biomaterialia. 2014;10:5012-5020
  176. 176. Darzi S, Deane JA, Nold CA, Edwards SE, Mukherjee S, et al. Endometrial mesenchymal stem/stromal cells modulate the macrophage response to implanted polyamide/gelatin composite mesh in immunocompromised and immunocompetent mice. Scientific Reports. 2018;8:6554. DOI: 10.1038/s41598-018-24919-6
  177. 177. Emmerson SJ, Mukherjee S, Melendez-Munoz J, Cousins F, Edwards SL, Karjalainen P, et al. Composite mesh design for delivery of autologous mesenchymal stem cells influences mesh integration, exposure and biocompatibility in an ovine model of pelvic organ prolapse. Biomaterials. 2019;225:119495. DOI: 10.1016/j.biomaterials.2019.119495

Written By

Manuela Cristina Russu

Submitted: 05 August 2022 Reviewed: 12 September 2022 Published: 29 October 2022