Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38565267

RESUMEN

Angiogenesis, the formation of new blood vessels from the preexistent microvasculature, is an essential component of wound repair and tumor growth. Nonsteroidal anti-inflammatory drugs that suppress prostanoid biosynthesis are known to suppress the incidence and progression of malignancies including colorectal cancers, and also to delay the wound healing. However, the precise mechanisms are not fully elucidated. Accumulated results obtained from prostanoid receptor knockout mice indicate that a prostaglandin E-type receptor signaling EP3 in the host microenvironment is critical in tumor angiogenesis inducing vascular endothelial growth factor A (VEGF-A). Further, lymphangiogenesis was also enhanced by EP signaling via VEGF-C/D inductions in pathological settings. These indicate the importance of EP receptor to facilitate angiogenesis and lymphangiogenesis in vivo. Prostanoids act beyond their commonly understood activities in smooth muscle contraction and vasoactivity, both of which are quick responses elicited within several seconds on stimulations. Prostanoid receptor signaling will be a potential therapeutic target for disease conditions related to angiogenesis and lymphangiogenesis.

2.
In Vivo ; 38(1): 160-173, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38148085

RESUMEN

BACKGROUND/AIM: Intestinal lymphatic vessels (lacteals) play a critical role in the absorption and transport of dietary lipids into the circulation. Calcitonin gene-related peptide and receptor activity-modifying protein 1 (RAMP1) are involved in lymphatic vessel growth. This study aimed to examine the role of RAMP1 signaling in lacteal morphology and function in response to a high-fat diet (HFD). MATERIALS AND METHODS: RAMP1 deficient (RAMP1-/-) or wild-type (WT) mice were fed a normal diet (ND) or HFD for 8 weeks. RESULTS: RAMP1-/- mice fed a HFD had increased body weights compared to WT mice fed a HFD, which was associated with high levels of total cholesterol, triglycerides, and glucose. HFD-fed RAMP1-/- mice had shorter and wider lacteals than HFD-fed WT mice. HFD-fed RAMP1-/- mice had lower levels of lymphatic endothelial cell gene markers including vascular endothelial growth factor receptor 3 (VEGFR3) and lymphatic vascular growth factor VEGF-C than HFD-fed WT mice. The concentration of an absorbed lipid tracer in HFD-fed RAMP1-/- mice was higher than that in HFD-fed WT mice. The zipper-like continuous junctions were predominant in HFD-fed WT mice, while the button-like discontinuous junctions were predominant in HFD-fed RAMP1-/- mice. CONCLUSION: Deletion of RAMP1 signaling suppressed lacteal growth and VEGF-C/VEGFR3 expression but accelerated the uptake and transport of dietary fats through discontinuous junctions of lacteals, leading to excessive obesity. Specific activation of RAMP1 signaling may represent a target for the therapeutic management of diet-induced obesity.


Asunto(s)
Vasos Linfáticos , Factor C de Crecimiento Endotelial Vascular , Ratones , Animales , Factor A de Crecimiento Endotelial Vascular , Proteína 1 Modificadora de la Actividad de Receptores/metabolismo , Obesidad/genética , Obesidad/metabolismo , Vasos Linfáticos/metabolismo , Grasas de la Dieta , Dieta Alta en Grasa/efectos adversos , Ratones Endogámicos C57BL , Ratones Noqueados
3.
Mol Med Rep ; 28(4)2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37654213

RESUMEN

Endometriosis is highly dependent on angiogenesis and lymphangiogenesis. Prostaglandin E2, an arachidonic acid metabolite, has been shown to promote the formation of new blood and lymphatic vessels. However, the role of another arachidonic acid metabolite, thromboxane A2 (TXA2) in angiogenesis and lymphangiogenesis during endometriosis remains largely unexplored. Using a murine model of ectopic endometrial transplantation, fragments from the endometrium of WT donor mice were transplanted into the peritoneal walls of recipient WT mice (WT→WT), resulting in an increase in both the area and density of blood and lymphatic vessels. Upon transplantation of endometrial tissue from thromboxane prostanoid (TP) receptor (TXA2 receptor)­deficient (TP­/­) mice into TP­/­ mice (TP­/­â†’TP­/­), an increase in implant growth, angiogenesis, and lymphangiogenesis were observed along with upregulation of pro­angiogenic and lymphangiogenic factors, including vascular endothelial growth factors (VEGFs). Similar results were obtained using a thromboxane synthase (TXS) inhibitor in WT→WT mice. Furthermore, TP­/­â†’TP­/­ mice had a higher number of F4/80+ cells than that of WT→WT mice, with increased expression of genes related to the anti­inflammatory macrophage phenotype in endometrial lesions. In cultured bone marrow (BM)­derived macrophages, the levels of VEGF­A, VEGF­C, and VEGF­D decreased in a TP­dependent manner. Furthermore, TP signaling affected the polarization of cultured BM­derived macrophages to the anti­inflammatory phenotype. These findings imply that inhibition of TP signaling promotes endometrial implant growth and neovascularization.


Asunto(s)
Endometriosis , Prostaglandinas , Receptores de Tromboxano A2 y Prostaglandina H2 , Animales , Femenino , Ratones , Ácido Araquidónico , Dinoprostona , Neovascularización Patológica/genética , Tromboxanos , Receptores de Tromboxano A2 y Prostaglandina H2/antagonistas & inhibidores , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo
4.
Mol Biol Rep ; 50(10): 7981-7993, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37540456

RESUMEN

BACKGROUND: Accumulating evidence suggests that prostaglandin E2, an arachidonic acid (AA) metabolite, enhances lymphangiogenesis in response to inflammation. However, thromboxane A2 (TXA2), another AA metabolite, is not well known. Thus, this study aimed to determine the role of thromboxane prostanoid (TP) signaling in lymphangiogenesis in secondary lymphedema. METHODS AND RESULTS: Lymphedema was induced by the ablation of lymphatic vessels in mouse tails. Compared with wild-type mice, tail lymphedema in Tp-deficient mice was enhanced, which was associated with suppressed lymphangiogenesis as indicated by decreased lymphatic vessel area and pro-lymphangiogenesis-stimulating factors. Numerous macrophages were found in the tail tissues of Tp-deficient mice. Furthermore, the deletion of TP in macrophages increased tail edema and decreased lymphangiogenesis and pro-lymphangiogenic cytokines, which was accompanied by increased numbers of macrophages and gene expression related to a pro-inflammatory macrophage phenotype in tail tissues. In vivo microscopic studies revealed fluorescent dye leakage in the lymphatic vessels in the wounded tissues. CONCLUSIONS: The results suggest that TP signaling in macrophages promotes lymphangiogenesis and prevents tail lymphedema. TP signaling may be a therapeutic target for improving lymphedema-related symptoms by enhancing lymphangiogenesis.


Asunto(s)
Vasos Linfáticos , Linfedema , Ratones , Animales , Linfangiogénesis , Prostaglandinas/metabolismo , Tromboxanos/metabolismo , Vasos Linfáticos/metabolismo , Macrófagos/metabolismo , Linfedema/genética , Linfedema/metabolismo
5.
Biomed Pharmacother ; 163: 114831, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37150029

RESUMEN

Thromboxane (TX) and prostaglandins are metabolites of arachidonic acid, a twenty-carbon unsaturated fatty acid, and have a variety of actions that are exerted via specific receptors. Angiogenesis is defined as the formation of new blood vessels from pre-existing vascular beds and is a critical component of pathological conditions, including inflammation and cancer. Lymphatic vessels play crucial roles in the regulation of interstitial fluid, immune surveillance, and the absorption of dietary fat from the intestine; and they are also involved in the pathogenesis of various diseases. Similar to angiogenesis, lymphangiogenesis, the formation of new lymphatic vessels, is a critical component of pathological conditions. The TP-dependent accumulation of platelets in microvessels has been reported to enhance angiogenesis under pathological conditions. Although the roles of some growth factors and cytokines in angiogenesis and lymphangiogenesis have been well characterized, accumulating evidence suggests that TX induces the production of proangiogenic and prolymphangiogenic factors through the activation of adenylate cyclase, and upregulates angiogenesis and lymphangiogenesis under disease conditions. In this review, we discuss the role of TX as a regulator of angiogenesis and lymphangiogenesis, and its emerging importance as a therapeutic target.


Asunto(s)
Vasos Linfáticos , Neoplasias , Humanos , Linfangiogénesis , Tromboxanos , Vasos Linfáticos/metabolismo , Neoplasias/patología , Inflamación/patología
6.
Front Cell Dev Biol ; 11: 1171317, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37082623

RESUMEN

The liver displays a remarkable regenerative capacity in response to acute liver injury. In addition to the proliferation of hepatocytes during liver regeneration, non-parenchymal cells, including liver macrophages, liver sinusoidal endothelial cells (LSECs), and hepatic stellate cells (HSCs) play critical roles in liver repair and regeneration. Liver ischemia-reperfusion injury (IRI) is a major cause of increased liver damage during liver resection, transplantation, and trauma. Impaired liver repair increases postoperative morbidity and mortality of patients who underwent liver surgery. Successful liver repair and regeneration after liver IRI requires coordinated interplay and synergic actions between hepatic resident cells and recruited cell components. However, the underlying mechanisms of liver repair after liver IRI are not well understood. Recent technological advances have revealed the heterogeneity of each liver cell component in the steady state and diseased livers. In this review, we describe the progress in the biology of liver non-parenchymal cells obtained from novel technological advances. We address the functional role of each cell component in response to liver IRI and the interactions between diverse immune repertoires and non-hematopoietic cell populations during the course of liver repair after liver IRI. We also discuss how these findings can help in the design of novel therapeutic approaches. Growing insights into the cellular interactions during liver IRI would enhance the pathology of liver IRI understanding comprehensively and further develop the strategies for improvement of liver repair.

7.
Cardiovasc Res ; 119(5): 1218-1233, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-35986688

RESUMEN

AIMS: Microsomal prostaglandin E synthase-1 (mPGES-1)/prostaglandin E2 (PGE2) induces angiogenesis through the prostaglandin E2 receptor (EP1-4). Among immune cells, regulatory T cells (Tregs), which inhibit immune responses, have been implicated in angiogenesis, and PGE2 is known to modulate the function and differentiation of Tregs. We hypothesized that mPGES-1/PGE2-EP signalling could contribute to recovery from ischaemic conditions by promoting the accumulation of Tregs. METHODS AND RESULTS: Wild-type (WT), mPGES-1-deficient (mPges-1-/-), and EP4 receptor-deficient (Ep4-/-) male mice, 6-8 weeks old, were used. Hindlimb ischaemia was induced by femoral artery ligation. Recovery from ischaemia was suppressed in mPges-1-/- mice and compared with WT mice. The number of accumulated forkhead box protein P3 (FoxP3)+ cells in ischaemic muscle tissue was decreased in mPges-1-/- mice compared with that in WT mice. Expression levels of transforming growth factor-ß (TGF-ß) and stromal cell derived factor-1 (SDF-1) in ischaemic tissue were also suppressed in mPges-1-/- mice. The number of accumulated FoxP3+ cells and blood flow recovery were suppressed when Tregs were depleted by injecting antibody against folate receptor 4 in WT mice but not in mPges-1-/- mice. Recovery from ischaemia was significantly suppressed in Ep4-/- mice compared with that in WT mice. Furthermore, mRNA levels of Foxp3 and Tgf-ß were suppressed in Ep4-/- mice. Moreover, the number of accumulated FoxP3+ cells in ischaemic tissue was diminished in Ep4-/- mice compared with that in Ep4+/+ mice. CONCLUSION: These findings suggested that mPGES-1/PGE2 induced neovascularization from ischaemia via EP4 by promoting the accumulation of Tregs. Highly selective EP4 agonists could be useful for the treatment of peripheral artery disease.


Asunto(s)
Dinoprostona , Linfocitos T Reguladores , Ratones , Masculino , Animales , Prostaglandina-E Sintasas/genética , Prostaglandina-E Sintasas/metabolismo , Dinoprostona/metabolismo , Dinoprostona/farmacología , Linfocitos T Reguladores/metabolismo , Ratones Noqueados , Isquemia/genética , Factor de Crecimiento Transformador beta , Factores de Transcripción Forkhead/genética
8.
In Vivo ; 36(6): 2604-2614, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36309359

RESUMEN

BACKGROUND/AIM: Invariant natural killer T (iNKT) cells are involved in the initiation and resolution of inflammatory responses. We previously reported that activated iNKT cells facilitate liver repair after hepatic ischemia reperfusion (I/R) injury by accelerating macrophage polarization during the early phase of hepatic I/R injury. Upon activation with α-galactosylceramide (α-GalCer), iNKT cell numbers transiently decrease before increasing within 72 h of stimulation. In the present study, we examined the role of expanded hepatic iNKT cells in the late phase of hepatic I/R injury. MATERIALS AND METHODS: iNKT cells were activated by intraperitoneal injection of α-GalCer in male C57/BL6 mice at the induction of hepatic ischemia followed by reperfusion. RESULTS: Numbers of activated hepatic iNKT cells immediately diminished after hepatic I/R and reached minimal levels at 24 h and 48 h post-reperfusion. Numbers of hepatic iNKT cells then increased at 72 h and 96 h post-reperfusion to levels approximately 2-fold higher than in mice that underwent a sham operation. Liver repair as demonstrated by decreased necrotic area and increased expression of proliferating cell nuclear antigen (PCNA) was enhanced in α-GalCer-treated mice at 96 h post-reperfusion. Interleukin (IL)-13 production by proliferating iNKT cells was observed at 96 h post-reperfusion, which was associated with enhanced liver repair and increased numbers of reparative macrophages. CONCLUSION: Repopulation of hepatic iNKT cells promotes liver repair by stimulating macrophage phenotype switching in the late phase of hepatic I/R injury.


Asunto(s)
Células T Asesinas Naturales , Daño por Reperfusión , Masculino , Ratones , Animales , Células T Asesinas Naturales/metabolismo , Ratones Endogámicos C57BL , Hígado/metabolismo , Daño por Reperfusión/metabolismo , Isquemia
9.
In Vivo ; 36(5): 2061-2073, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36099134

RESUMEN

BACKGROUND/AIM: Microsomal prostaglandin E synthase-1 (mPGES-1) is an enzyme, which catalyzes the final step of prostaglandin E2 (PGE2) synthesis. PGE2 in involved in wound-induced angiogenesis. Regulatory T cells (Tregs) regulate not only immune tolerance but also tissue repair and angiogenesis. We examined whether the mPGES-1/PGE2 axis contributes to wound-induced angiogenesis and granulation tissue formation through Treg accumulation. MATERIALS AND METHODS: The dorsal subcutaneous tissues of male mPGES-1-deficient (mPGES-1-/-) and C57BL/6 wild-type (WT) mice were implanted with polyurethane sponge disks. Angiogenesis was estimated by determining the wet weight of sponge tissues and the expression of proangiogenic factors including CD31, vascular endothelial growth factor (VEGF), and transforming growth factor ß (TGF-ß) in granulation tissues. RESULTS: Angiogenesis was suppressed in mPGES-1-/- mice compared with WT mice, which was associated with attenuated forkhead box P3 (Foxp3) expression and Foxp3+ Treg accumulation. The number of cells double-positive for Foxp3/TGFß and Foxp3/VEGF were lower in mPGES-1-/- mice than in WT mice. Neutralizing Tregs with antibodies (Abs) against CD25 or folate receptor 4 (FR4) inhibited the Foxp3+ Treg angiogenesis and accumulation in WT mice, but not in mPGES-1-/- mice. The topical application of PGE2 into the implanted sponge enhanced angiogenesis and accumulation of Tregs expressing TGFß and VEGF in WT and mPGES-1-/- mice. CONCLUSION: Tregs producing TGFß and VEGF accumulate in wounds and contribute to angiogenesis through mPGES-1-derived PGE2 mPGES-1 induction may control angiogenesis in skin wounds by recruiting Tregs.


Asunto(s)
Prostaglandina-E Sintasas/metabolismo , Linfocitos T Reguladores , Factor A de Crecimiento Endotelial Vascular , Animales , Dinoprostona/metabolismo , Factores de Transcripción Forkhead , Tejido de Granulación , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/genética , Prostaglandina-E Sintasas/genética , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta , Factor A de Crecimiento Endotelial Vascular/genética
10.
Am J Biol Anthropol ; 177(1): 39-47, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-36787753

RESUMEN

OBJECTIVES: Efforts have been made to mathematically reconstruct the brain morphology from human fossil crania to clarify the evolutionary changes in the brain that are associated with the emergence of human cognitive ability. However, because conventional reconstruction methods are based solely on the endocranial shape, deep brain structures cannot be estimated with sufficient accuracy. Our study aims to investigate the possible morphological correspondence between the cranial and deep brain morphologies based on humans and African great apes, with the goal of a more precise reconstruction of fossil brains. MATERIALS AND METHODS: Midsagittal endocranial and deep brain landmarks were obtained from magnetic resonance images of humans and three species of African great apes. The average midsagittal endocranial profile of all four species was calculated after Procrustes registration. The spatial deformation function from each of the endocranial profiles to the average endocranial profile was defined, and the brain landmarks enclosed in the endocranium were transformed using the deformation function to evaluate the interspecific variabilities of the positions of the brain landmarks on the average endocranial profile. RESULTS: The interspecific differences in the shape-normalized positions of the corpus callosum, anterior commissure, thalamus center, and brainstem were approximately within the range of 2% of the human cranial length, indicating that the interspecific variabilities of the positions of these deep brain structures were relatively small among the four species. DISCUSSION: Such an invariant relationship of the deep brain structure and the endocranium that encloses the brain can potentially be utilized to reconstruct the brains of fossil hominins.


Asunto(s)
Hominidae , Animales , Humanos , Hominidae/anatomía & histología , Cráneo/anatomía & histología , Encéfalo/diagnóstico por imagen , Tronco Encefálico , Cuerpo Calloso
11.
Pharmacol Ther ; 232: 108011, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34614423

RESUMEN

Lymphatic vessels have crucial roles in the regulation of interstitial fluids, immune surveillance, and the absorption of dietary fat in the intestine. Lymphatic function is also closely related to the pathogenesis of various disease states such as inflammation, lymphedema, endometriosis, liver dysfunction, and tumor metastasis. Lymphangiogenesis, the formation of new lymphatic vessels from pre-existing lymphatic vessels, is a critical determinant in the above conditions. Although the effect of growth factors on lymphangiogenesis is well-characterized, and biologically active lipids are known to affect smooth muscle contractility and vasoaction, there is accumulating evidence that biologically active lipids are also important inducers of growth factors and cytokines that regulate lymphangiogenesis. This review discusses recent advances in our understanding of biologically active lipids, including arachidonic acid metabolites, sphingosine 1-phosphate, and lysophosphatidic acid, as regulators of lymphangiogenesis, and the emerging importance of the lymphangiogenesis as a therapeutic target.


Asunto(s)
Vasos Linfáticos , Linfedema , Femenino , Humanos , Inflamación , Lípidos , Linfangiogénesis/fisiología , Vasos Linfáticos/patología , Linfedema/patología
12.
Int J Exp Pathol ; 103(1): 4-12, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34655121

RESUMEN

The process of gastric ulcer healing includes cell migration, proliferation, angiogenesis and re-epithelialization. Platelets contain angiogenesis stimulating factors that induce angiogenesis. Thromboxane A2 (TXA2 ) not only induces platelet activity but also angiogenesis. This study investigated the role of TXA2 in gastric ulcer healing using TXA2 receptor knockout (TPKO) mice. Gastric ulcer healing was suppressed by treatment with the TXA2 synthase inhibitor OKY-046 and the TXA2 receptor antagonist S-1452 compared with vehicle-treated mice. TPKO showed delayed gastric ulcer healing compared with wild-type mice (WT). The number of microvessels and CD31 expression were lower in TPKO than in WT mice, and TPKO suppressed the expression of transforming growth factor beta (TGF-ß) and vascular endothelial growth factor A (VEGF-A) in areas around gastric ulcers. Immunofluorescence assays showed that TGF-ß and VEGF-A co-localized with platelets. Gastric ulcer healing was significantly reduced in WT mice transplanted with TPKO compared with WT bone marrow. These results suggested that TP signalling on platelets facilitates gastric ulcer healing through TGF-ß and VEGF-A.


Asunto(s)
Neovascularización Patológica/metabolismo , Úlcera Gástrica/tratamiento farmacológico , Tromboxanos/farmacología , Cicatrización de Heridas/efectos de los fármacos , Animales , Ratones Endogámicos C57BL , Activación Plaquetaria/efectos de los fármacos , Prostaglandinas/farmacología , Receptores de Tromboxanos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Úlcera Gástrica/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Sci Rep ; 11(1): 20889, 2021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34686756

RESUMEN

Investigating the morphological differences of the calcaneus in humans and great apes is crucial for reconstructing locomotor repertories of fossil hominins. However, morphological variations in the calcaneus of the great apes (chimpanzees, gorillas, and orangutans) have not been sufficiently studied. This study aims to clarify variations in calcaneal morphology among great apes based on three-dimensional geometric morphometrics. A total of 556 landmarks and semilandmarks were placed on the calcaneal surface to calculate the principal components of shape variations among specimens. Clear interspecific differences in calcaneal morphology were extracted, corresponding to the degree of arboreality of the three species. The most arboreal orangutans possessed comparatively more slender calcaneal tuberosity and deeper pivot region of the cuboid articular surface than chimpanzees and gorillas. However, the most terrestrial gorillas exhibited longer lever arm of the triceps surae muscle, larger peroneal trochlea, more concave plantar surface, more inverted calcaneal tuberosity, more everted cuboid articular surface, and more prominent plantar process than the orangutans and chimpanzees. These interspecific differences possibly reflect the functional adaptation of the calcaneus to locomotor behavior in great apes. Such information might be useful for inferring foot functions and reconstructing the locomotion of fossil hominoids and hominids.


Asunto(s)
Calcáneo/anatomía & histología , Gorilla gorilla/anatomía & histología , Pan troglodytes/anatomía & histología , Adaptación Fisiológica/fisiología , Animales , Fósiles , Hominidae/anatomía & histología , Humanos , Locomoción/fisiología
14.
Front Immunol ; 12: 754106, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34691073

RESUMEN

Macrophage polarization is critical for liver tissue repair following acute liver injury. However, the underlying mechanisms of macrophage phenotype switching are not well defined. Invariant natural killer T (iNKT) cells orchestrate tissue inflammation and tissue repair by regulating cytokine production. Herein, we examined whether iNKT cells played an important role in liver repair after hepatic ischemia-reperfusion (I/R) injury by affecting macrophage polarization. To this end, we subjected male C57BL/6 mice to hepatic I/R injury, and mice received an intraperitoneal (ip) injection of α-galactosylceramide (α-GalCer) or vehicle. Compared with that of the vehicle, α-GalCer administration resulted in the promotion of liver repair accompanied by acceleration of macrophage differentiation and by increases in the numbers of Ly6Chigh pro-inflammatory macrophages and Ly6Clow reparative macrophages. iNKT cells activated with α-GalCer produced interleukin (IL)-4 and interferon (IFN)-γ. Treatment with anti-IL-4 antibodies delayed liver repair, which was associated with an increased number of Ly6Chigh macrophages and a decreased number of Ly6Clow macrophages. Treatment with anti-IFN-γ antibodies promoted liver repair, associated with reduced the number of Ly6Chigh macrophages, but did not change the number of Ly6Clow macrophages. Bone marrow-derived macrophages up-regulated the expression of genes related to both a pro-inflammatory and a reparative phenotype when co-cultured with activated iNKT cells. Anti-IL-4 antibodies increased the levels of pro-inflammatory macrophage-related genes and decreased those of reparative macrophage-related genes in cultured macrophages, while anti-IFN-γ antibodies reversed the polarization of macrophages. Cd1d-deficient mice showed delayed liver repair and suppressed macrophage switching, compared with that in wild-type mice. These results suggest that the activation of iNKT cells by α-GalCer facilitated liver repair after hepatic I/R injury by both IL-4-and IFN-γ-mediated acceleration of macrophage polarization. Therefore, the activation of iNKT cells may represent a therapeutic tool for liver repair after hepatic I/R injury.


Asunto(s)
Galactosilceramidas/farmacología , Regeneración Hepática/fisiología , Hígado/inmunología , Activación de Macrófagos , Células T Asesinas Naturales/inmunología , Animales , Antígenos CD1d/genética , Antígenos CD1d/inmunología , Células Cultivadas , Técnicas de Cocultivo , Interferón gamma/antagonistas & inhibidores , Interferón gamma/biosíntesis , Interleucina-4/antagonistas & inhibidores , Interleucina-4/biosíntesis , Hígado/irrigación sanguínea , Regeneración Hepática/inmunología , Activación de Linfocitos/efectos de los fármacos , Macrófagos/clasificación , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células T Asesinas Naturales/efectos de los fármacos , Células T Asesinas Naturales/metabolismo , Daño por Reperfusión
15.
In Vivo ; 35(5): 2577-2587, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34410945

RESUMEN

BACKGROUND/AIM: Although the pathology of sinusoidal obstruction syndrome (SOS) is characterized by damage to liver sinusoidal endothelial cells (LSECs), the processes underlying LSEC repair are incompletely understood. The angiopoietin (Ang)/Tie system contributes to angiogenesis. The present study aimed to examine the processes of LSEC repair and the involvement of the Ang/Tie pathway in LSEC recovery. MATERIALS AND METHODS: Experimentally, SOS was induced by intraperitoneal injection of monocrotaline (MCT) to C57/BL6 mice. RESULTS: Levels of LSEC markers were up-regulated during the repair phase of MCT-induced hepatotoxicity. The damaged LSECs recovered from the injury by expanding LSECs expressing lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1) in the peri-central area of MCT-injured livers, while LSECs in the same area of uninjured livers lacked LYVE-1 expression. Bone marrow (BM)-derived cells did not incorporate into the restored LSECs. Tie2 expression was related to LSEC recovery in MCT-injured liver tissue. CONCLUSION: The resident LSECs neighboring uninjured tissue replace damaged LSECs in MCT-injured livers. Tie2 is involved in LSEC recovery from MCT-induced hepatotoxicity.


Asunto(s)
Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Monocrotalina , Animales , Células Endoteliales , Hepatocitos , Hígado , Ratones , Monocrotalina/toxicidad
16.
Inflamm Regen ; 41(1): 16, 2021 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-34082837

RESUMEN

Vascular endothelial growth factor (VEGF) is not only an important factor for angiogenesis but also lung development and homeostasis. VEGF-A binds three tyrosine kinase (TK) receptors VEGFR1-3. Idiopathic pulmonary fibrosis (IPF) is one of the poor prognoses of lung diseases. The relationship of VEGF and IPF remains to be clarified. Treatment with nintedanib used for the treatment of IPF reduced fibroblast proliferation, inhibited TK receptors, platelet-derived growth factor receptor (PDGFR), fibroblast growth factor receptor (FGFR), and VEGFR. Because the effect of that treatment is still not satisfactory, the emergence of new therapeutic agents is needed. This review describes the enhancement of pulmonary fibrosis by VEGFR1-TK signal and suggests that the blocking of the VEGFR1-TK signal may be useful for the treatment of pulmonary fibrosis.

17.
Exp Ther Med ; 22(1): 668, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33986833

RESUMEN

Monocrotaline (MCT) administration induces liver injury in rodents that mimics the pathology of human sinusoidal obstruction syndrome. MCT-induced SOS models are used to investigate the mechanism of injury and optimize treatment strategies. However, the processes underlying liver repair are largely unknown. Specifically, the role of macrophages, the key drivers of liver repair, has not been elucidated. The current study aimed to examine the role of macrophages in the repair of MCT-induced liver injury in male C57/BL6 mice. Maximal liver injury occurred at 48 h post-MCT treatment, followed by repair at 120 h post-treatment. Immunofluorescence analysis revealed that CD68+ macrophages were recruited to the injured regions after MCT treatment. This was associated with the decreased expression of genes related to a pro-inflammatory macrophage phenotype and the increased expression of those associated with a reparative macrophage phenotype during the repair phase. The results also revealed that stromal cell-derived factor-1 (SDF-1) and its receptor C-X-C chemokine receptor-4 (CXCR4) were upregulated, and CD68+ macrophages were co-localized with CXCR4 expression. Treatment of mice with AMD3100, a CXCR4 antagonist, delayed liver repair and increased the expression of genes related to a pro-inflammatory macrophage phenotype. In contrast, SDF-1 treatment stimulated liver repair and increased the expression of genes related to a reparative macrophage phenotype. The results suggested that macrophages accumulate in the liver and repair damaged tissue after MCT treatment, and that the SDF-1-CXCR4 axis is involved in this process.

18.
Toxicol Lett ; 335: 71-81, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-33122006

RESUMEN

Destruction of liver sinusoidal endothelial cells (LSECs) is an initial event in sinusoidal obstruction syndrome (SOS) that leads to accumulation of platelets in the liver. Herein, we explored the role of platelets during progression of experimental SOS induced by monocrotaline (MCT) in mice. Depletion of platelets using an anti-CD41 antibody or anti-thrombocyte serum exacerbated MCT-induced liver injury in C57BL/6 mice, as indicated by an increase in the alanine transaminase (ALT) level, which was associated with hemorrhagic necrosis. Thrombocytosis induced by thrombopoietin (TPO) or the TPO receptor agonist romiplostim (ROM) attenuated MCT-induced liver injury, as evidenced by lower levels of ALT and mRNA encoding matrix metalloproteinase (MMP) 9, and higher levels of mRNA encoding vascular endothelial growth factor receptor (VEGFR) 2 and VEGFR3. The level of activated hepatic platelets was higher in TPO- and ROM-treated mice than in saline-treated mice. Co-culture with a high number of platelets increased the viability of LSECs and their mRNA levels of CD31, VEGFR2, and VEGFR3, and decreased their mRNA level of MMP9. The level of VEGF-A was increased in the culture medium of LSECs co-cultured with platelets. These results indicate that platelets attenuate MCT-induced liver injury by minimizing damage to LSECs.


Asunto(s)
Plaquetas/efectos de los fármacos , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/sangre , Enfermedad Veno-Oclusiva Hepática/sangre , Enfermedad Veno-Oclusiva Hepática/inducido químicamente , Monocrotalina/toxicidad , Trombocitosis/sangre , Animales , Plaquetas/citología , Supervivencia Celular/efectos de los fármacos , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/prevención & control , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Enfermedad Veno-Oclusiva Hepática/prevención & control , Pruebas de Función Hepática , Masculino , Ratones Endogámicos C57BL , Recuento de Plaquetas , Receptores Fc , Receptores de Trombopoyetina/agonistas , Proteínas Recombinantes de Fusión/farmacología , Trombocitosis/inducido químicamente , Trombopoyetina/farmacología
19.
Biomed Pharmacother ; 121: 109581, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31715374

RESUMEN

BACKGROUND: Accumulation of myeloid-derived suppressor cells (MDSCs) to tumors is related to cancer prognosis. We investigated the contribution of host stromal microsomal prostaglandin E synthase-1 (mPGES-1) to the accumulation of MDSCs in metastasized lungs of prostate cancer in mice. MATERIAL AND METHODS: Eight-week-old male C57Bl/6 wild type (WT) mice and mPGES-1 knock out mice (mPGES-1KO) were injected with RM9 murine prostate cancer cell line (5 × 106 cells/mL). Lung metastasis was evaluated by the number of colonies, the weight of the lung, and the number of MDSCs (CD11b+Gr1+ cells) in the lung. RESULTS: Intravenous injections of RM9, a murine prostate cancer cell line to WT mice revealed that lung metastasis and accumulation of MDCSs were suppressed with treatments with a Gr1 antibody, a COX-2 inhibitor, and an mPGES-1 inhibitor. Lung metastasis and accumulation of CD11b+Gr1+MDSCs were suppressed in mPGES-1KO mice. The mRNA level of stromal cell-derived factor-1 (SDF-1) in the lung and the number of accumulated SDF-1-expressing CD11b+Gr1+ MDSCs were elevated at an early stage in lung metastasis of C-X-C chemokine receptor type 4 (CXCR4)-expressing RM9 in an mPGES-1-dependent manner. The number of CXCR4-expressing CD11b+Gr1+MDSCs in WT mice was higher than that in mPGES-1KO mice. RM9 lung metastasis and accumulation of CD11b+Gr1+MDSCs were suppressed by CXCR4 antibody in WT mice but not in mPGES-1KO. WT mice transplanted with mPGES-1 KO bone marrow (BM) showed a significant reduction in lung metastasis and accumulation of CD11b+Gr1+MDSCs. CONCLUSION: These results suggest that mPGES-1 enhances tumor metastasis by inducing accumulation of BM-derived MDSCs. Selective mPGES-1 inhibitors might, therefore, represent valuable therapeutic tools for the suppression of tumor metastasis.


Asunto(s)
Neoplasias Pulmonares/patología , Células Supresoras de Origen Mieloide/metabolismo , Prostaglandina-E Sintasas/genética , Animales , Antígenos Ly/metabolismo , Células de la Médula Ósea/citología , Antígeno CD11b/metabolismo , Quimiocina CXCL12/metabolismo , Neoplasias Pulmonares/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , ARN Mensajero/metabolismo , Receptores CXCR4/metabolismo
20.
Biomed Pharmacother ; 117: 109067, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31176171

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a lethal lung disease with a poor prognosis. Fibroblast proliferation amplifies extracellular matrix deposition and increases angiogenesis. Vascular endothelial growth factor (VEGF) is one of the most potent angiogenic factors. VEGF interacts with VEGF receptors (VEGFR1 and VEGFR2). A previous study showed that VEGFR1 tyrosine kinase (TK) signaling induced blood flow recovery mediated by bone marrow (BM)-derived stem cells. We hypothesized that VEGFR1-TK signaling might be related to pulmonary fibrosis. MATERIAL AND METHODS: Six-week-old male C57Bl/6 wild-type (WT) mice and VEGFR1 TK knockout mice (TKKO mice) were treated with a single intratracheal injection of bleomycin (BLM; 0.1 µg in 50 µl saline) or vehicle (saline; 50 µl). Lung fibrosis was evaluated by histology, real-time PCR and ELISA for pro-fibrotic factors, and assessment of lung mechanics. RESULTS: The fibrotic area in the lung and the lung elastance were significantly reduced in TKKO mice (P < 0.01). The expression of the fibrosis-related factors type I collagen, S100A4, and transforming growth factor (TGF)-ß was also significantly reduced in TKKO mice on day 21 after BLM injection. TKKO mice also had significantly lower levels of stromal cell-derived factor (SDF)-1 in the lungs and plasma on days 14 and 21 after BLM treatment (P < 0.05). Moreover, the expression of C-X-C chemokine receptor type 7 (CXCR7) and CXCR4, the receptors for SDF-1, was also suppressed in TKKO mice. Immunohistochemical analysis showed that treatment with a CXCR4 antibody decreased the accumulation of VEGFR1+ cells in the lung in WT mice but not in TKKO mice. CONCLUSION: These results suggest that VEGFR1 TK signaling promotes BLM-induced pulmonary fibrosis by activating the SDF-1/CXCR4 axis in infiltrating VEGFR1+ cells.


Asunto(s)
Fibrosis Pulmonar/metabolismo , Transducción de Señal , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Bleomicina , Quimiocina CXCL12/administración & dosificación , Quimiocina CXCL12/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CXCR/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA