Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomedicines ; 11(10)2023 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-37892992

RESUMEN

We investigated whether anogenital distance (AGD) is associated with adenomyosis, endometriosis and uterine leiomyomas (UL, also called uterine fibroids). We recruited 81 women with UL, 105 with ovarian endometrioma (OE), 116 with adenomyosis, 28 with both adenomyosis and UL, and 100 control subjects with other acquired gynecological conditions but not endometriosis, adenomyosis, UL, or polycystic ovarian syndrome. Measurements from the anterior clitoral surface to the center of the anus (AGDAC), from the tip of the clitoris to the center of the anus (AGDACt), and from the posterior fourchette to the center of the anus (AGDAF) were made in all subjects. Multiple regression was performed to estimate the association between AGDs and presence of OE, adenomyosis, and UL while controlling for possible confounding factors. We found that, compared with controls, women with OE and adenomyosis, but not UL, had significantly shorter AGDAF, but not AGDAC. However, the amount of variance that could be explained by the disease status is rather moderate, suggesting that factors other than disease status, bodyweight and height were also responsible for AGD. Thus, prenatal exposure to reduced levels of androgen may increase the risk of developing endometriosis and adenomyosis. However, other factors may also contribute to the pathogenesis of endometriosis and adenomyosis.

2.
J Clin Endocrinol Metab ; 105(11)2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32813013

RESUMEN

CONTEXT: While fibrosis in endometriosis has recently loomed prominently, the sources of myofibroblasts, the principal effector cell in fibrotic diseases, remain largely obscure. Mesothelial cells (MCs) can be converted into myofibroblasts through mesothelial-mesenchymal transition (MMT) in many fibrotic diseases and adhesion. OBJECTIVE: To evaluate whether MCs contribute to the progression and fibrogenesis in endometriosis through MMT. SETTING, DESIGN, PATIENTS, INTERVENTION, AND MAIN OUTCOME MEASURES: Dual immunofluorescence staining and immunohistochemistry using antibodies against calretinin, Wilms' tumor-1 (WT-1), and α-smooth muscle actin (α-SMA) were performed on lesion samples from 30 patients each with ovarian endometrioma (OE) and deep endometriosis (DE), and 30 normal endometrial (NE) tissue samples. Human pleural and peritoneal MCs were co-cultured with activated platelets or control medium with and without neutralization of transforming growth factor ß1 (TGF-ß1) and/or platelet-derived growth factor receptor (PDGFR) and their morphology, proliferation, and expression levels of genes and proteins known to be involved in MMT were evaluated, along with their migratory and invasive propensity, contractility, and collagen production. RESULTS: The number of calretinin/WT-1 and α-SMA dual-positive fibroblasts in OE/DE lesions was significantly higher than NE samples. The extent of lesional fibrosis correlated positively with the lesional α-SMA staining levels. Human MCs co-cultured with activated platelets acquire a morphology suggestive of MMT, concomitant with increased proliferation, loss of calretinin expression, and marked increase in expression of mesenchymal markers. These changes coincided with functional differentiation as reflected by increased migratory and invasive capacity, contractility, and collagen production. Neutralization of TGF-ß1 and PDGFR signaling abolished platelet-induced MMT in MCs. CONCLUSIONS: MCs contribute to lesional progression and fibrosis through platelet-induced MMT.


Asunto(s)
Plaquetas/metabolismo , Transdiferenciación Celular/fisiología , Endometriosis/patología , Miofibroblastos/patología , Enfermedades del Ovario/patología , Enfermedades Peritoneales/patología , Adulto , Calbindina 2/metabolismo , Línea Celular , Progresión de la Enfermedad , Endometriosis/metabolismo , Femenino , Fibrosis/metabolismo , Fibrosis/patología , Humanos , Persona de Mediana Edad , Miofibroblastos/metabolismo , Enfermedades del Ovario/metabolismo , Enfermedades Peritoneales/metabolismo , Proteínas WT1/metabolismo , Adulto Joven
3.
Reprod Biomed Online ; 41(3): 373-384, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32651107

RESUMEN

RESEARCH QUESTION: Does sodium tanshinone IIA sulfonate (STS) induce cellular senescence in endometriotic lesions and thus restrict lesional development and fibrogenesis in a recently established mouse model of deep endometriosis? DESIGN: Prospective randomized animal experiment in which deep endometriosis was induced in female Balb/C mice, which were then randomly divided into three groups (low-dose STS, high-dose STS and inert vehicle control) and received treatment for 2 weeks. All mice were then sacrificed and their lesions excised and harvested. Lesion weight was quantified and all lesion samples were subjected to histochemical analysis of the extent of lesional fibrosis by Masson trichrome staining, and of cellular senescence by senescence-associated ß-galactosidase (SA-ß-gal), along with immunohistochemistry analyses of p53, CCN1, activate Salvador 1 (Sav1), hyaluronan synthase 2 (HAS2), survivin, granulocyte-macrophage colony-stimulating factor (GM-CSF) and CD163-positive M2 macrophages. Plasma P-selectin and hyaluronic acid levels were also quantified. Hotplate testing was also administered before the induction, then before and after treatment. RESULTS: STS treatment resulted in significantly reduced lesion weight, stalled lesional fibrogenesis and improved hyperalgesia, seemingly through the induction of cellular senescence by activating p53, Sav1 and CCN1 while suppressing HAS2, survivin and GM-CSF, resulting in increased apoptosis and reduced lesional infiltration of alternatively activated macrophages. In addition, STS treatment significantly reduced the plasma concentration of P-selectin and hyaluronic acid, possibly leading to reduced lesional platelet aggregation. CONCLUSIONS: STS appears to be a promising compound for treating endometriosis. The results suggest that senescence may restrict lesional progression and fibrogenesis, and targeting the senescence pathway may have desirable therapeutic potential.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Endometriosis/patología , Endometrio/efectos de los fármacos , Fenantrenos/farmacología , Animales , Endometriosis/metabolismo , Endometrio/metabolismo , Endometrio/patología , Femenino , Galectinas/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Ácido Hialurónico/sangre , Ratones , Selectina-P/sangre , Estudios Prospectivos , Survivin/metabolismo
4.
Reprod Biomed Online ; 41(3): 500-517, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32709523

RESUMEN

RESEARCH QUESTION: Do endometriotic lesions undergo endothelial-mesenchymal transition (EndoMT)? DESIGN: Lesion samples from 30 patients with ovarian endometriomas and deep endometriosis, and control endometrial tissue samples from 30 women without endometriosis, were analysed. In-vitro experimentation using the human umbilical vein endothelial cell (HUVEC) line were conducted. Immunofluorescence staining and immunohistochemistry analysis using antibodies against endothelial cell and mesenchymal cell markers were conducted. The HUVEC cells were co-cultured with activated platelets or control medium with and without neutralization of TGF-ß1 PDGFR, or both. Their morphology, proliferation and expression levels of genes and proteins known to be involved in EndoMT were evaluated, along with their migratory and invasive propensity, contractility and collagen production capability. RESULTS: The proportion of CD31 and FSP-1 dual-positive cells in FSP-1+ fibroblasts was 74.7% (±5.4%) in ovarian endometrioma lesions, significantly higher than that in deep endometriosis lesions (26.8% ± 26.0%; P = 5.7 × 10-5), and was zero in normal endometrium. The extent of lesional fibrosis correlated positively with staining levels of the lesional mesenchymal markers FSP-1 and α-SMA (r = 0.91; P < 2.2 × 10-16, r = 0.81; P = 5.8 × 10-15, respectively). Human endothelial cells co-cultured with activated platelets acquire a morphology suggestive of EndoMT, concomitant with increased proliferation, loss of CD31 but marked increase in expression of mesenchymal markers. Morphological and gene and protein expression changes are accompanied by functional differentiation reflected by increased migratory and invasive capacity, contractility and collagen production. Neutralization of TGF-ß1 and PDGFR signalling abolished platelet-induced EndoMT in human endothelial cells. CONCLUSIONS: Multiple sources of myofibroblasts exist in endometriotic lesions, and implicates platelets, EndoMT, or both, as potential therapeutic targets for treating endometriosis.


Asunto(s)
Plaquetas , Endometriosis/patología , Transición Epitelial-Mesenquimal/fisiología , Fibroblastos/patología , Enfermedades del Ovario/patología , Ovario/patología , Enfermedades Peritoneales/patología , Adulto , Transdiferenciación Celular/fisiología , Células Endoteliales/patología , Femenino , Fibrosis/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Persona de Mediana Edad , Adulto Joven
5.
Sci Rep ; 9(1): 2698, 2019 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-30804432

RESUMEN

Endometriotic lesions are known to be hyperinnervated, especially in lesions of deep endometriosis (DE), which are frequently in close proximity to various nerve plexuses. DE lesions typically have higher fibromuscular content than that of ovarian endometriomas (OE) lesions, but the underlying reason remains elusive. Aside from their traditional role of pain transduction, however, whether or not sensory nerves play any role in the development of endometriosis is unclear. Here, we show that, thorough their respective receptors neurokinin receptor 1 (NK1R), calcitonin receptor like receptor (CRLR), and receptor activity modifying protein 1 (RAMP-1), neuropeptides substance P (SP) and calcitonin gene related peptide (CGRP) induce epithelial-mesenchymal transition (EMT), fibroblast-to-myofibroblast transdifferentiation (FMT) and further turn stromal cells into smooth muscle cells (SMCs) in endometriotic lesions, resulting ultimately in fibrosis. We show that SP and CGRP, or the rat dorsal root ganglia (DRG) supernatant, through the induction of NK1R and CGRP/CRLR/RAMP-1 signaling pathways, promoted EMT, FMT and SMM in endometriosis, resulting in increased migratory and invasive propensity, cell contractility, production of collagen, and eventually to fibrosis. Neutralization of NK1R and/or CGRP/CRLR/RAMP-1 abrogated these processes. Extended exposure of endometriotic stromal cells to SP and/or CGRP or the DRG supernatant induced increased expression of α-SMA, desmin, oxytocin receptor, and smooth muscle myosin heavy-chain. Finally, we show that DE lesions had significantly higher nerve fiber density, increased staining levels of α-SMA, NK1R, CRLR, and RAMP-1, concomitant with higher lesional fibrotic content than that of OE lesions. The extent of lesional fibrosis correlated positively with the staining levels of NK1R, CRLR, and RAMP-1, as well as the nerve fiber density in lesions. Thus, this study provides another piece of evidence that sensory nerves play an important role in promoting the development and fibrogenesis of endometriosis. It explains as why DE frequently have higher fibromuscular content than that of OE, highlights the importance of lesional microenvironment in shaping the lesional fate, gives more credence to the idea that ectopic endometrium is fundamentally wounds that go through repeated tissue injury and repair, and should shed much needed light into the pathophysiology of endometriosis.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/farmacología , Endometriosis/metabolismo , Sustancia P/farmacología , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Inmunohistoquímica , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Neuroquinina-1/metabolismo
6.
Hum Reprod ; 34(3): 452-468, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30689856

RESUMEN

STUDY QUESTION: Do sensory nerves play any role in the development of endometriosis? SUMMARY ANSWER: Sensory nerves participate in all major steps (epithelial-mesenchymal transition (EMT), fibroblast-to-myofibroblast transdifferentiation (FMT) and smooth muscle metaplasia (SMM)) in the development and fibrogenesis of endometriotic lesions. WHAT IS KNOWN ALREADY: Endometriotic lesions are known to be hyperinnervated due to neurogenesis resulting from neutrophins secreted by endometriotic lesions and possibly platelets. These neutrophins seem to preferentially favour production of sensory neurons at the expense of sympathetic neurons. STUDY DESIGN, SIZE, DURATION: Three independent, yet complementary, prospective, randomized mouse experimentations were conducted. A total of 143 female Balb/C mice and 24 female immunodeficient nude Balb/C mice were used. The mice were sacrificed 2 or 4 weeks after the induction of endometriosis. PARTICIPANTS/MATERIALS, SETTING, METHODS: In Experiment 1, 21 mice were randomly divided into three groups of equal size for sympathetic denervation, sensory denervation and controls. Denervation was carried out chemically. In Experiment 2, 24 nude mice were randomly divided into three equal-sized groups: the BEFORE and AFTER groups that respectively received surgical denervation 3 days before or after the induction of endometriosis by subcutaneous grafting of human endometriotic tissues, and the Control group that received a sham surgery without denervation 3 days before induction. For Experiments 1 and 2, all mice were sacrificed two weeks after induction of endometriosis. In Experiment 3, substance P (SP) and aprepitant, a potent and selective neurokinin 1 receptor (NK1R) antagonist, were used to activate and inhibit the NK1R signalling pathway, respectively. A total of 32 mice were randomly divided into four groups of equal sizes: control (CTL), SP, Before-Induction and After-Induction. One day before the induction of endometriosis, mice in CTL, SP and Before-Induction groups were infused with sterile saline, SP and aprepitant, respectively, via Alzet osmotic pumps. Two weeks after the induction, the After-induction group was infused with aprepitant in similar fashion. All mice were sacrificed four weeks after the induction of endometriosis. In all three experiments, the bodyweight and hotplate latency were evaluated before induction and sacrifice. In addition, all lesions were excised, weighed and processed for quantification and immunohistochemistry analysis of markers for EMT, FMT and SMM, and the extent of fibrosis was evaluated by Masson trichrome staining. MAIN RESULTS AND THE ROLE OF CHANCE: In Experiment 1, chemical denervation of sympathetic and sensory nerves reduced the lesion weight by 43.2% (±23.1%) and 68.7% (±20.3%), respectively, as compared with controls. In particular, sensory denervation led to significantly greater reduction in lesion weight than sympathetic denervation. Sensory denervation also resulted in significantly improved hyperalgesia as compared with controls. In contrast, sympathetic denervation yielded only transient improvement in hyperalgesia. Both sympathetic and sensory denervation resulted in lower immunoreactivity against markers of proliferation and fibrosis, especially sensory denervation.In Experiment 2, surgical denervation before or after induction of endometriosis also decelerated the development of endometriosis, as manifested by significantly reduced lesion weight and extent of lesional fibrosis, along with improved hyperalgesia.In Experiment 3, NK1R activation by SP infusion accelerated lesional development, as evidenced by significantly increased lesional weight, more thorough progression of EMT, FMT, SMM, exaggerated lesional fibrosis and deteriorated hyperalgesia. In contrast, NK1R antagonism decelerated lesional development and improved hyperalgesia. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: This study is limited by the use of histologic and immunohistochemistry analyses only and the lack of molecular data. WIDER IMPLICATIONS OF THE FINDINGS: Since sensory nerves are known to be important in wound healing and fibrogenesis, our findings also give more credence to the notion that endometriotic lesions are wounds undergoing repeated tissue injury and repair. As such, sensory nerves or the NK1R signalling pathway in particular may be potential targets for intervention. STUDY FUNDING/COMPETING INTEREST(S): This research was supported by Grants 81471434 (SWG), 81530040 (SWG), 81771553 (SWG), 81671436 (XSL) and 81871144 (XSL) from the National Natural Science Foundation of China and an Excellence in Centres of Clinical Medicine grant (2017ZZ01016) from the Science and Technology Commission of Shanghai Municipality. None of the authors have anything to disclose.


Asunto(s)
Endometriosis/fisiopatología , Neuropéptidos/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Plaquetas/metabolismo , Diferenciación Celular , Proliferación Celular , Desnervación , Femenino , Fibroblastos/metabolismo , Humanos , Hiperalgesia/fisiopatología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Músculo Liso/metabolismo , Mioblastos/metabolismo , Distribución Aleatoria , Receptores de Neuroquinina-1/metabolismo , Transducción de Señal
7.
Hum Reprod ; 34(2): 235-247, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30561644

RESUMEN

STUDY QUESTION: Is it possible to establish a mouse model of deep endometriosis (DE)? SUMMARY ANSWER: A mouse DE model that is macroscopically and microscopically similar to nodular lesions in humans can be constructed in as short as 3 weeks by intraperitoneal injection of uterine fragments along with the infusion of substance P (SP) and/or calcitonin gene-related peptide (CGRP). WHAT IS KNOWN ALREADY: Although a baboon DE model was reported 5 years ago, its prohibitive cost and demand for facilities and expertise associated with the use of non-human primates put its use out of reach for most laboratories. STUDY DESIGN, SIZE, DURATION: A total of 48 female Balb/C mice were used for this study. Among them, 16 were randomly selected as donors that contributed uterine fragments, and the remaining 32 were recipient mice. The mice with induced endometriosis were followed up for 3-4 weeks. PARTICIPANTS/MATERIALS, SETTING, METHODS: One day before the induction of endometriosis by intraperitoneal injection of uterine fragments, osmotic pumps were inserted into equal groups of recipient mice to infuse either sterile saline, SP, CGRP, or both SP and CGRP. The hotplate test was administrated to all mice at the baseline and before and after induction of endometriosis. Four (3 for the SP+CGRP group) weeks after induction, all mice were sacrificed. Their endometriotic lesions were excised, weighed and processed for histopathologic examination, and histochemistry, immunohistochemistry and immunofluorescence analyses of markers of proliferation, angiogenesis, epithelial-mesenchymal transition (EMT), fibroblast-to-myofibroblast transdifferentiation (FMT), smooth muscle metaplasia (SMM), mesothelial-mesenchymal transition (MMT) and endothelial-mesenchymal transition (EndoMT) were done. The extent of lesional fibrosis was evaluated by Masson trichrome staining. To further evaluate surrounding organ/tissue invasion, the peritoneal areas adhesive to the lesions were excised for immunohistochemical analysis. MAIN RESULTS AND THE ROLE OF CHANCE: Endometriotic lesions in mice treated with SP and/or CGRP satisfied all requirements for DE, i.e. presence of endometrial epithelial and stromal cells, abundance of fibromuscular content, and encapsulation in surrounding tissues or organs. The lesion weight in the CGRP, SP and SP+CGRP groups was 1.62, 2.14 and 2.18-fold, respectively, heavier than that of control group. Concomitantly, the SP, CGRP and SP+CGRP groups had significantly shorter hotplate latency than that of control group. Lesions in mice treated with SP and/or CGRP, especially with SP+CGRP, exhibited characteristics consistent with EMT, FMT, SMM and extensive fibrosis, along with signs of MMT and EndoMT. Lesional invasion into surrounding tissues/organs was found to be 25.0, 75.0 and 87.5% in mice treated with CGRP, SP and SP+CGRP, but none in control mice. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: This study is limited by the use of histologic and immunohistochemistry analyses only and lacks molecular data. WIDER IMPLICATIONS OF THE FINDINGS: The establishment of a mouse DE model supports the idea that endometriotic lesions are wounds undergoing repeated tissue injury and repair and underscores the importance of microenvironments in shaping the lesions' destiny. In addition, signs consistent with MMT and EndoMT indicate that there may be more culpable factors that still remain unidentified and should be pursued in the future. Moreover, the close correlation between the extent of lesional fibrosis and markers of EMT, MMT, EndoMT, FMT and SMM as shown here should facilitate our understanding of the molecular mechanisms underlying the DE pathophysiology. Since this DE model is based on a biologically plausible and evidence-backed theory, it should shed much needed insight into the molecular mechanisms underlying the pathophysiology of DE. STUDY FUNDING/COMPETING INTEREST(S): This research was supported by Grants 81471434 (S.W.G.), 81530040 (S.W.G.), 81771553 (S.W.G.), 81671436 (X.S.L.) and 81871144 (X.S.L.) from the National Natural Science Foundation of China. None of the authors has any conflict of interest to disclose.


Asunto(s)
Modelos Animales de Enfermedad , Endometriosis/patología , Enfermedades Peritoneales/patología , Animales , Péptido Relacionado con Gen de Calcitonina/administración & dosificación , Transdiferenciación Celular , Endometrio/citología , Endometrio/patología , Transición Epitelial-Mesenquimal , Femenino , Fibrosis , Humanos , Inyecciones Intraperitoneales , Ratones , Miofibroblastos , Peritoneo/citología , Peritoneo/patología , Sustancia P/administración & dosificación
8.
Reprod Sci ; 24(6): 829-835, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27688244

RESUMEN

Hyperinnervation in endometriosis is now well documented, but so far only a few neurotrophins have been identified. Since endometriotic stromal cells secrete thromboxane A2 (TXA2), we sought to determine whether TXA2, derived from endometriotic stromal cells, induces neurite outgrowth. Using primary sensory neurons derived from rat dorsal root ganglia (DRG) and ectopic endometrial stromal cells (EESCs) derived from human ovarian endometrioma tissues, we treated the primary neurons with different concentrations of U-46619, a stable TXA2 mimetic, and performed a neuronal growth assay. The primary neurons were also cocultured with a vehicle, nerve growth factor (NGF, serving as a positive control), the supernatant of EESC culture medium, or the supernatant of EESCs pretreated with ozagrel, a thromboxane synthase inhibitor, and a neuronal growth assay was performed. The total neurite length was evaluated through immunofluorescence microscopy. We found that U-46619 significantly increased the neurite outgrowth in DRG neurons in a concentration-dependent fashion ( P < .001). It also increased the number of neurite ends in a concentration-dependent fashion. Ozagrel treatment alone had no effect on the neurite growth ( P > .05), and the treatment with the supernatant of EESCs induced neurite outgrowth just as potently as that treated with NGF (positive control; P > .05). Remarkably, treatment with the EESC supernatant increased the neurite outgrowth by nearly 3-fold as compared with the control ( P < .01), but the pretreatment with ozagrel abolished the stimulatory effect of the EESC by 31.3% ( P < .05). These findings indicate that EESCs potently induce neurite outgrowth, and endometriosis-derived TXA2 is responsible, at least in part, for this neurotrophic effect.


Asunto(s)
Endometriosis/metabolismo , Neuritas/efectos de los fármacos , Proyección Neuronal/efectos de los fármacos , Enfermedades del Ovario/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Tromboxano A2/farmacología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Relación Dosis-Respuesta a Droga , Femenino , Ganglios Espinales/efectos de los fármacos , Humanos , Factor de Crecimiento Nervioso/farmacología , Ratas
9.
Eur J Obstet Gynecol Reprod Biol ; 209: 14-24, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27418559

RESUMEN

One of major objectives in treating endometriosis is to alleviate pain since dysmenorrhea and other types of pain top the list of complaints from women with endometriosis who seek medical attention. Indeed, endometriosis-associated pain (EAP) is the most debilitating of the disease that negatively impacts on the quality of life in affected women, contributing significantly to the burden of disease and adding to the substantial personal and societal costs. Unfortunately, the mechanisms underlying the EAP are still poorly understood. In the last two decades, one active research field in endometriosis is the investigation on the distribution and genesis of nerve fibers in eutopic and ectopic endometrium, and the attempt to use endometrial nerve fiber density for diagnostic purpose. Since EAP presumably starts with the terminal sensory nerves, in or around endometriotic lesions, that transduce noxious mediators to the central nervous system (CNS) which ultimately perceives pain, this field of research holds the promise to elucidate the molecular mechanisms underlying the EAP, thus opening new avenues for novel diagnostics and therapeutics. In this review, we shall first briefly provide some basic facts on nerve fibers, and then provide an overview of some major findings in this filed while also note some conflicting results and expose areas in need of further research. We point out that since recently accumulated evidence suggests that endometriotic lesions are wounds undergoing repeated tissue injury and repair, the relationship between endometriotic lesions and nerve fibers is not simply unidirectional, i.e. lesions promote hyperinnervations. Rather, it is bidirectional, i.e. endometriotic lesions and nerve fibers engage active cross-talks, resulting in the development of endometriosis and pain. That is, nerve fibers and endometriotic lesions are actually partners in crime in inflicting pains in women with endometriosis, aided and abetted possibly by other culprits, some yet to be identified. We provide a list of possible perpetrators likely to be involved in this crime. Finally, we discuss possible implications when viewing the relationship from this vista.


Asunto(s)
Endometriosis/patología , Endometrio/patología , Fibras Nerviosas/patología , Dolor Pélvico/patología , Calidad de Vida , Células Receptoras Sensoriales/patología , Endometriosis/complicaciones , Femenino , Humanos , Dolor Pélvico/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...