Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
J Neuroendocrinol ; 36(10): e13415, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38808481

RESUMEN

Ischemic stroke is a significant global health issue, ranking fifth among all causes of death and a leading cause of serious long-term disability. Ischemic stroke leads to severe outcomes, including permanent brain damage and neuronal dysfunction. Therefore, decreasing and preventing neuronal injuries caused by stroke has been the focus of therapeutic research. In recent years, many studies have shown that fluctuations in hormonal levels influence the prognosis of ischemic stroke. Thus, it is relevant to understand the role of hormones in the pathophysiological mechanisms of ischemic stroke for preventing and treating this health issue. Here, we investigate the contribution of the prolactin/vasoinhibin axis, an endocrine system regulating blood vessel growth, immune processes, and neuronal survival, to the pathophysiology of ischemic stroke. Male mice with brain overexpression of prolactin or vasoinhibin by adeno-associated virus (AAV) intracerebroventricular injection or lacking the prolactin receptor (Prlr-/-) were exposed to transient middle cerebral artery occlusion (tMCAO) for 45 min followed by 48 h of reperfusion. Overexpression of vasoinhibin or the absence of the prolactin receptor led to an increased lesion volume and decreased survival rates in mice following tMCAO, whereas overexpression of prolactin had no effect. In addition, astrocytic distribution in the penumbra was altered, glial fibrillary acidic protein and S100b mRNA expressions were reduced, and interleukin-6 mRNA expression increased in the ischemic hemisphere of mice overexpressing vasoinhibin. Of note, prolactin receptor-null mice (Prlr-/-) showed a marked increase in serum vasoinhibin levels. Furthermore, vasoinhibin decreased astrocyte numbers in mixed hippocampal neuron-glia cultures. These observations suggest that increased vasoinhibin levels may hinder astrocytes' protective reactivity. Overall, this study suggests the involvement of the prolactin/vasoinhibin axis in the pathophysiology of ischemic stroke-induced brain injury and provides insights into the impact of its dysregulation on astrocyte reactivity and lesion size. Understanding these mechanisms could help develop therapeutic interventions in ischemic stroke and other related neurological disorders.


Asunto(s)
Proteínas de Ciclo Celular , Gliosis , Prolactina , Receptores de Prolactina , Animales , Prolactina/metabolismo , Masculino , Ratones , Gliosis/patología , Gliosis/metabolismo , Receptores de Prolactina/metabolismo , Receptores de Prolactina/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Ratones Endogámicos C57BL , Astrocitos/metabolismo , Astrocitos/patología , Ratones Noqueados , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/metabolismo
2.
Eye (Lond) ; 38(3): 520-528, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37673971

RESUMEN

BACKGROUND/OBJECTIVE: The prokinetic levosulpiride elevates vasoinhibin levels in the vitreous of patients with proliferative diabetic retinopathy (PDR) suggesting clinical benefits due to the anti-vasopermeability and anti-angiogenic properties of vasoinhibin. We investigated the biological activity of levosulpiride in centre-involving diabetic macular oedema (DME). PATIENTS/METHODS: Prospective, randomized, double-blinded, dual-centre, phase 2 trial in patients with centre-involving DME orally treated with placebo (n = 17) or levosulpiride (n = 17) for 8 weeks or in patients with PDR undergoing elective pars plana vitrectomy and receiving placebo (n = 18) or levosulpiride (n = 18) orally for the 1 week before vitrectomy. RESULTS: Levosulpiride improved changes from baseline in best-corrected visual acuity (p ≤ 0.037), central foveal thickness (CFT, p ≤ 0.013), and mean macular volume (MMV, p ≤ 0.002) at weeks 4, 6, and 8 compared to placebo. At 8 weeks, the proportion of eyes gaining ≥5 ETDRS letters at 4 m (41% vs. 28%), losing ≥21 µm in CFT (55% vs. 28%), and dropping ≥0.06 mm3 in MMV (65% vs. 29%) was higher after levosulpiride than placebo. The overall grading of visual and structural parameters improved with levosulpiride (p = 0.029). Levosulpiride reduced VEGF (p = 0.025) and PlGF (p = 0.008) levels in the vitreous of PDR patients. No significant adverse side-effects were detected. CONCLUSIONS: Oral levosulpiride for 8 weeks improved visual and structural outcomes in patients with centre-involving DME by mechanisms that may include intraocular upregulation of vasoinhibin and downregulation of VEGF and PlGF. Larger clinical trials evaluating long-term efficacy and safety are warranted.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Edema Macular , Sulpirida/análogos & derivados , Humanos , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/cirugía , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Estudios Prospectivos , Inyecciones Intravítreas , Inhibidores de la Angiogénesis
3.
Endocrinology ; 165(2)2023 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-38057149

RESUMEN

Vasoinhibin, a proteolytic fragment of the hormone prolactin, inhibits blood vessel growth (angiogenesis) and permeability, stimulates the apoptosis and inflammation of endothelial cells, and promotes fibrinolysis. The antiangiogenic and antivasopermeability properties of vasoinhibin were recently traced to the HGR motif located in residues 46 to 48 (H46-G47-R48), allowing the development of potent, orally active, HGR-containing vasoinhibin analogues for therapeutic use against angiogenesis-dependent diseases. However, whether the HGR motif is also responsible for the apoptotic, inflammatory, and fibrinolytic properties of vasoinhibin has not been addressed. Here, we report that HGR-containing analogues are devoid of these properties. Instead, the incubation of human umbilical vein endothelial cells with oligopeptides containing the sequence HNLSSEM, corresponding to residues 30 to 36 of vasoinhibin, induced apoptosis, nuclear translocation of NF-κB, expression of genes encoding leukocyte adhesion molecules (VCAM1 and ICAM1) and proinflammatory cytokines (IL1B, IL6, and TNF), and adhesion of peripheral blood leukocytes. Also, intravenous or intra-articular injection of HNLSSEM-containing oligopeptides induced the expression of Vcam1, Icam1, Il1b, Il6, and Tnf in the lung, liver, kidney, eye, and joints of mice and, like vasoinhibin, these oligopeptides promoted the lysis of plasma fibrin clots by binding to plasminogen activator inhibitor-1 (PAI-1). Moreover, the inhibition of PAI-1, urokinase plasminogen activator receptor, or NF-κB prevented the apoptotic and inflammatory actions. In conclusion, the functional properties of vasoinhibin are segregated into 2 different structural determinants. Because apoptotic, inflammatory, and fibrinolytic actions may be undesirable for antiangiogenic therapy, HGR-containing vasoinhibin analogues stand as selective and safe agents for targeting pathological angiogenesis.


Asunto(s)
FN-kappa B , Inhibidor 1 de Activador Plasminogénico , Humanos , Interleucina-6 , Células Endoteliales de la Vena Umbilical Humana , Oligopéptidos
4.
Endocrinology ; 164(12)2023 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-37864848

RESUMEN

The close association between rheumatoid arthritis (RA), sex, reproductive state, and stress has long linked prolactin (PRL) to disease progression. PRL has both proinflammatory and anti-inflammatory outcomes in RA, but responsible mechanisms are not understood. Here, we show that PRL modifies in an opposite manner the proinflammatory actions of IL-1ß and TNF-α in mouse synovial fibroblasts in culture. Both IL-1ß and TNF-α upregulated the metabolic activity and the expression of proinflammatory factors (Il1b, Inos, and Il6) via the activation of the nuclear factor-κB (NF-κB) signaling pathway. However, IL-1ß increased and TNF-α decreased the levels of the long PRL receptor isoform in association with dual actions of PRL on synovial fibroblast inflammatory response. PRL reduced the proinflammatory effect and activation of NF-κB by IL-1ß but increased TNF-α-induced inflammation and NF-κB signaling. The double-faceted role of PRL against the 2 cytokines manifested also in vivo. IL-1ß or TNF-α with or without PRL were injected into the knee joints of healthy mice, and joint inflammation was monitored after 24 hours. IL-1ß and TNF-α increased the joint expression of proinflammatory factors and the infiltration of immune cells. PRL prevented the actions of IL-1ß but was either inactive or further increased the proinflammatory effect of TNF-α. We conclude that PRL exerts opposite actions on joint inflammation in males and females that depend on specific proinflammatory cytokines, the level of the PRL receptor, and the activation of NF-κB signaling. Dual actions of PRL may help balance joint inflammation in RA and provide insights for development of new treatments.


Asunto(s)
Artritis Reumatoide , Citocinas , Masculino , Femenino , Ratones , Animales , Citocinas/metabolismo , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Prolactina/farmacología , Prolactina/metabolismo , Membrana Sinovial/metabolismo , Células Cultivadas , Artritis Reumatoide/metabolismo , Inflamación/metabolismo , Fibroblastos/metabolismo
5.
J Vis Exp ; (192)2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36876927

RESUMEN

Insulin resistance is a reduced effect of insulin on its target cells, usually derived from decreased insulin receptor signaling. Insulin resistance contributes to the development of type 2 diabetes (T2D) and other obesity-derived diseases of high prevalence worldwide. Therefore, understanding the mechanisms underlying insulin resistance is of great relevance. Several models have been used to study insulin resistance both in vivo and in vitro; primary adipocytes represent an attractive option to study the mechanisms of insulin resistance and identify molecules that counteract this condition and the molecular targets of insulin-sensitizing drugs. Here, we have established an insulin resistance model using primary adipocytes in culture treated with tumor necrosis factor-α (TNF-α). Adipocyte precursor cells (APCs), isolated from collagenase-digested mouse subcutaneous adipose tissue by magnetic cell separation technology, are differentiated into primary adipocytes. Insulin resistance is then induced by treatment with TNF-α, a proinflammatory cytokine that reduces the tyrosine phosphorylation/activation of members of the insulin signaling cascade. Decreased phosphorylation of insulin receptor (IR), insulin receptor substrate (IRS-1), and protein kinase B (AKT) are quantified by western blot. This method provides an excellent tool to study the mechanisms mediating insulin resistance in adipose tissue.


Asunto(s)
Adipocitos , Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Animales , Ratones , Insulina , Receptor de Insulina , Factor de Necrosis Tumoral alfa , Diferenciación Celular , Cultivo Primario de Células
6.
Front Endocrinol (Lausanne) ; 13: 994898, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36157442

RESUMEN

Diabetic retinopathy (DR) and diabetic macular edema (DME) are major causes for visual loss in adults. Nearly half of the world's population with diabetes has some degree of DR, and DME is a major cause of visual impairment in these patients. Severe vision loss occurs because of tractional retinal detachment due to retinal neovascularization, but the most common cause of moderate vision loss occurs in DME where excessive vascular permeability leads to the exudation and accumulation of extracellular fluid and proteins in the macula. Metabolic control stands as an effective mean for controlling retinal vascular alterations in some but not all patients with diabetes, and the search of other modifiable factors affecting the risk for diabetic microvascular complications is warranted. Prolactin (PRL) and its proteolytic fragment, vasoinhibin, have emerged as endogenous regulators of retinal blood vessels. PRL acquires antiangiogenic and anti-vasopermeability properties after undergoing proteolytic cleavage to vasoinhibin, which helps restrict the vascularization of ocular organs and, upon disruption, promotes retinal vascular alterations characteristic of DR and DME. Evidence is linking PRL (and other pituitary hormones) and vasoinhibin to DR and recent preclinical and clinical evidence supports their translation into novel therapeutic approaches.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Mácula Lútea , Edema Macular , Adulto , Humanos , Edema Macular/complicaciones , Prolactina , Retina , Trastornos de la Visión
7.
Endocrinology ; 163(5)2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35305012

RESUMEN

Inflammatory arthritis defines a family of diseases influenced by reproductive hormones. Vasoinhibin, a fragment of the hormone prolactin (PRL), has antiangiogenic and proinflammatory properties. We recently showed that vasoinhibin reduces joint inflammation and bone loss in severe antigen-induced arthritis (AIA) by an indirect mechanism involving the inhibition of pannus vascularization. This unexpected finding led us to hypothesize that a severe level of inflammation in AIA obscured the direct proinflammatory action of vasoinhibin while allowing the indirect anti-inflammatory effect via its antiangiogenic properties. In agreement with this hypothesis, here we show that the intra-articular injection of an adeno-associated virus type-2 vector encoding vasoinhibin reduced joint inflammation in a severe AIA condition, but elevated joint inflammation in a mild AIA model. The proinflammatory effect, unmasked in mild AIA, resulted in joint swelling, enhanced leukocyte infiltration, and upregulation of expression of genes encoding proinflammatory mediators (Il1b, Il6, Inos, Mmp3), adhesion molecule (Icam1), and chemokines (Cxcl1, Cxcl2, Cxcl3, Ccl2). Furthermore, vasoinhibin induced the expression of proinflammatory mediators and chemokines in cultured synovial fibroblasts through nuclear factor-κB. Finally, matrix metalloproteases and cathepsin D, upregulated in the arthritic joint, cleaved PRL to vasoinhibin, and vasoinhibin levels increased in the circulation of mice subjected to AIA. We suggest that vasoinhibin is generated during inflammatory arthritis and acts on synovial fibroblasts and endothelial cells to initially promote and later inhibit inflammation, respectively. These opposite effects may work together to help keep joint inflammation under balance.


Asunto(s)
Artritis Experimental , Artritis , Animales , Artritis Experimental/genética , Células Endoteliales/metabolismo , Inflamación , Ratones , Neovascularización Patológica , Prolactina/metabolismo
9.
J Biol Chem ; 298(3): 101695, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35143839

RESUMEN

Vascular endothelial cells (ECs) form a critical interface between blood and tissues that maintains whole-body homeostasis. In COVID-19, disruption of the EC barrier results in edema, vascular inflammation, and coagulation, hallmarks of this severe disease. However, the mechanisms by which ECs are dysregulated in COVID-19 are unclear. Here, we show that the spike protein of SARS-CoV-2 alone activates the EC inflammatory phenotype in a manner dependent on integrin ⍺5ß1 signaling. Incubation of human umbilical vein ECs with whole spike protein, its receptor-binding domain, or the integrin-binding tripeptide RGD induced the nuclear translocation of NF-κB and subsequent expression of leukocyte adhesion molecules (VCAM1 and ICAM1), coagulation factors (TF and FVIII), proinflammatory cytokines (TNFα, IL-1ß, and IL-6), and ACE2, as well as the adhesion of peripheral blood leukocytes and hyperpermeability of the EC monolayer. In addition, inhibitors of integrin ⍺5ß1 activation prevented these effects. Furthermore, these vascular effects occur in vivo, as revealed by the intravenous administration of spike, which increased expression of ICAM1, VCAM1, CD45, TNFα, IL-1ß, and IL-6 in the lung, liver, kidney, and eye, and the intravitreal injection of spike, which disrupted the barrier function of retinal capillaries. We suggest that the spike protein, through its RGD motif in the receptor-binding domain, binds to integrin ⍺5ß1 in ECs to activate the NF-κB target gene expression programs responsible for vascular leakage and leukocyte adhesion. These findings uncover a new direct action of SARS-CoV-2 on EC dysfunction and introduce integrin ⍺5ß1 as a promising target for treating vascular inflammation in COVID-19.


Asunto(s)
COVID-19 , Inflamación , Integrina alfa5beta1 , FN-kappa B , Glicoproteína de la Espiga del Coronavirus , Factor de Necrosis Tumoral alfa , COVID-19/metabolismo , COVID-19/patología , COVID-19/virología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/virología , Integrina alfa5beta1/metabolismo , Interleucina-6/metabolismo , FN-kappa B/metabolismo , Oligopéptidos , SARS-CoV-2 , Transducción de Señal , Glicoproteína de la Espiga del Coronavirus/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
10.
J Neuroendocrinol ; 34(4): e13091, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35078262

RESUMEN

Excessive vasopermeability and angiogenesis compromise vision in diabetic macular oedema (DME) and diabetic retinopathy (DR). Vasoinhibin is a fragment of the hormone prolactin (PRL) that inhibits diabetes-induced retinal hypervasopermeability and ischaemia-induced retinal angiogenesis in rodents. Hyperprolactinaemia generated by the dopamine D2 receptor antagonist, levosulpiride, is associated with higher levels of vasoinhibin in the vitreous of patients with DR, implying a beneficial outcome due to vasoinhibin-mediated inhibition of retinal vascular alterations. Here, we tested whether hyperprolactinaemia induced by racemic sulpiride increases intraocular vasoinhibin levels and inhibits retinal hypervasopermeability in diabetic rats. Diabetes was generated with streptozotocin and, 4 weeks later, rats were treated for 2 weeks with sulpiride or osmotic minipumps delivering PRL. ELISA, Western blot, and Evans blue assay were used to evaluate serum PRL, retinal vasoinhibin, and retinal vasopermeability, respectively. Hyperprolactinaemia in response to sulpiride or exogenous PRL was associated with increased levels of vasoinhibin in the retina and reduced retinal hypervasopermeability. Furthermore, sulpiride decreased retinal haemorrhages in response to the intravitreal administration of vascular endothelial growth factor (VEGF). Neither sulpiride nor exogenous PRL modified blood glucose levels or bodyweight. We conclude that sulpiride-induced hyperprolactinaemia inhibits the diabetes- and VEGF-mediated increase in retinal vasopermeability by promoting the intraocular conversion of endogenous PRL to vasoinhibin. These findings support the therapeutic potential of sulpiride and its levorotatory enantiomer, levosulpiride, against DME and DR.


Asunto(s)
Diabetes Mellitus Experimental , Retinopatía Diabética , Hiperprolactinemia , Animales , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/metabolismo , Humanos , Hiperprolactinemia/inducido químicamente , Hiperprolactinemia/complicaciones , Hiperprolactinemia/metabolismo , Prolactina/metabolismo , Ratas , Retina/metabolismo , Sulpirida/metabolismo , Sulpirida/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología
11.
Angiogenesis ; 25(1): 57-70, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34097181

RESUMEN

The hormone prolactin acquires antiangiogenic and antivasopermeability properties after undergoing proteolytic cleavage to vasoinhibin, an endogenous prolactin fragment of 123 or more amino acids that inhibits the action of multiple proangiogenic factors. Preclinical and clinical evidence supports the therapeutic potential of vasoinhibin against angiogenesis-related diseases including diabetic retinopathy, peripartum cardiomyopathy, rheumatoid arthritis, and cancer. However, the use of vasoinhibin in the clinic has been limited by difficulties in its production. Here, we removed this barrier to using vasoinhibin as a therapeutic agent by showing that a short linear motif of just three residues (His46-Gly47-Arg48) (HGR) is the functional determinant of vasoinhibin. The HGR motif is conserved throughout evolution, its mutation led to vasoinhibin loss of function, and oligopeptides containing this sequence inhibited angiogenesis and vasopermeability with the same potency as whole vasoinhibin. Furthermore, the oral administration of an optimized cyclic retro-inverse vasoinhibin heptapeptide containing HGR inhibited melanoma tumor growth and vascularization in mice and exhibited equal or higher antiangiogenic potency than other antiangiogenic molecules currently used as anti-cancer drugs in the clinic. Finally, by unveiling the mechanism that obscures the HGR motif in prolactin, we anticipate the development of vasoinhibin-specific antibodies to solve the on-going challenge of measuring endogenous vasoinhibin levels for diagnostic and interventional purposes, the design of vasoinhibin antagonists for managing insufficient angiogenesis, and the identification of putative therapeutic proteins containing HGR.


Asunto(s)
Proteínas de Ciclo Celular , Retinopatía Diabética , Inhibidores de la Angiogénesis/farmacología , Animales , Ratones , Oligopéptidos/farmacología , Prolactina
12.
Mol Cell Endocrinol ; 538: 111471, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34601001

RESUMEN

Vasoinhibin is an antiangiogenic, profibrinolytic peptide generated by the proteolytic cleavage of the pituitary hormone prolactin by cathepsin D, matrix metalloproteinases, and bone morphogenetic protein-1. Vasoinhibin can also be generated when placental lactogen or growth hormone are enzymatically cleaved. Here, it is investigated whether plasmin cleaves human prolactin and placental lactogen to generate vasoinhibin-like peptides. Co-incubation of prolactin and placental lactogen with plasmin was performed and analyzed by gel electrophoresis and Western blotting. Mass spectrometric analyses were carried out for sequence validation and precise cleavage site identification. The cleavage sites responsible for the generation of the vasoinhibin-like peptides were located at K170-E171 in prolactin and R160-T161 in placental lactogen. Various genetic variants of the human prolactin and placental lactogen genes are projected to affect proteolytic generation of the vasoinhibin-like peptides. The endogenous counterparts of the vasoinhibin-like peptides generated by plasmin may represent vasoinhibin-isoforms with inhibitory effects on vasculature and coagulation.


Asunto(s)
Fibrinolisina/metabolismo , Péptidos/análisis , Lactógeno Placentario/química , Prolactina/química , Proteínas de Ciclo Celular/química , Variación Genética , Células HEK293 , Humanos , Espectrometría de Masas , Lactógeno Placentario/genética , Prolactina/genética , Proteolisis
13.
Endocrinology ; 162(12)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34418052

RESUMEN

Vasoinhibin is an endogenous prolactin (PRL) fragment with profibrinolytic, antivasopermeability, and antiangiogenic effects. The fact that blood clotting, vascular permeability, and angiogenesis are functionally linked during the wound-healing process led us to investigate whether thrombin, a major protease in tissue repair, generates vasoinhibin. Here, we have incubated human PRL with thrombin and analyzed the resulting proteolytic products by Western blot, mass spectrometry, high-performance liquid chromatography purification, recombinant production, and bioactivity. We unveil a main thrombin cleavage site at R48-G49 that rapidly (< 10 minutes) generates a 5.6-kDa fragment (residues 1-48) with full vasoinhibin activity, that is, it inhibited the proliferation, invasion, and permeability of cultured endothelial cells and promoted the lysis of a fibrin clot in plasma with a similar potency to that of a conventional 14-kDa vasoinhibin (residues 1-123). The R48-G49 cleavage site is highly conserved throughout evolution and precedes the intramolecular disulfide bond (C58-C174), thereby allowing the 5.6-kDa vasoinhibin to be released without a reduction step. Furthermore, the 5.6-kDa vasoinhibin is produced by endogenous thrombin during the clotting process. These findings uncover the smallest vasoinhibin known, add thrombin to the list of PRL-cleaving proteases generating vasoinhibin, and introduce vasoinhibin as a thrombin-activated mechanism for the regulation of hemostasis, vasopermeability, and angiogenesis in response to tissue injury.


Asunto(s)
Fragmentos de Péptidos/metabolismo , Prolactina/metabolismo , Trombina/fisiología , Células 3T3-L1 , Secuencia de Aminoácidos , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Permeabilidad Capilar/efectos de los fármacos , Bovinos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Modelos Moleculares , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Prolactina/química , Prolactina/farmacología , Proteolisis , Regeneración/efectos de los fármacos , Regeneración/fisiología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
14.
Front Endocrinol (Lausanne) ; 12: 645085, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33959096

RESUMEN

Vasoinhibin is a protein hormone with antiangiogenic, antivasodilatatory, and antivasopermeability effects generated by the proteolytic cleavage of prolactin. The discovery of its role in diabetic retinopathy and peripartum cardiomyopathy led to the evaluation of new pharmacological treatments in clinical interventional trials. However, the quantitative evaluation of vasoinhibin in biological samples from patients has not been possible due to the lack of vasoinhibin-specific antibodies. Recently, loop 1 of vasoinhibin was identified to have a different three-dimensional structure compared to PRL, and thus to contain vasoinhibin-specific epitopes. Here, we report the development of two sets of vasoinhibin-specific monoclonal antibodies against two neighboring regions of the vasoinhibin loop 1. An experimental sandwich ELISA with two monoclonal anti-vasoinhibin antibodies was developed, which had no cross-reactivity to recombinant human full-length prolactin. The ELISA had a quantitation limit of 100 ng/ml, and intra-assay- and inter-assay coefficients of variation of 12.5% and 14%, respectively. The evaluation of 15 human serum samples demonstrated concentrations of below limit of detection (n=3), below limit of quantitation (n=1) and between 0.23 µg/ml (230 ng/ml) to 605 µg/ml (n=12) in the quantifiable range. Despite the high specificity of the monoclonal-monoclonal antibody sandwiches which discriminate vasoinhibin from PRL, there might be cross-reactivities by serum proteins other than vasoinhibin. A fully established vasoinhibin ELISA may support diagnostic and therapeutic measures in vascular diseases.


Asunto(s)
Anticuerpos Monoclonales/química , Proteínas de Ciclo Celular/química , Retinopatía Diabética/terapia , Inhibidores de la Angiogénesis , Proteínas de Ciclo Celular/sangre , Retinopatía Diabética/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Límite de Detección , Conformación Molecular , Prolactina/química , Proteolisis , Proteínas Recombinantes/química , Sensibilidad y Especificidad
15.
Transl Vis Sci Technol ; 9(9): 27, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32879783

RESUMEN

Purpose: High circulating levels of the hormone prolactin (PRL) protect against experimental diabetic retinopathy (DR) due to the retinal accumulation of vasoinhibin, a PRL fragment that inhibits blood vessel permeability and growth. A phase 2 clinical trial is investigating a new therapy for DR based on elevating serum PRL levels with levosulpiride, a prokinetic dopamine D2 receptor blocker. Here, we tested whether levosulpiride-induced hyperprolactinemia elevates PRL and vasoinhibin in the vitreous of volunteer patients with proliferative DR (PDR) undergoing elective pars plana vitrectomy. Methods: Patients were randomized to receive placebo (lactose pill, orally TID; n = 19) or levosulpiride (25 mg orally TID; n = 18) for the 7 days before vitrectomy. Vitreous samples from untreated non-diabetic (n = 10) and PDR (n = 17) patients were also studied. Results: Levosulpiride elevated the systemic (101 ± 13 [SEM] vs. 9.2 ± 1.3 ng/mL, P < 0.0001) and vitreous (3.2 ± 0.4 vs. 1.5 ± 0.2 ng/mL, P < 0.0001) levels of PRL, and both levels were directly correlated (r = 0.58, P < 0.0002). The vitreous from non-diabetic patients or from PDR patients treated with levosulpiride, but not from placebo-treated PDR patients, inhibited the basic fibroblast growth factor (bFGF)- and vascular endothelial growth factor (VEGF)-induced proliferation of endothelial cells in culture. Vasoinhibin-neutralizing antibodies reduced the vitreous antiangiogenic effect. Matrix metalloproteases (MMPs) in the vitreous cleaved PRL to vasoinhibin, and their activity was higher in non-diabetic than in PDR patients. Conclusions: Levosulpiride increases the levels of PRL in the vitreous of PDR patients and promotes its MMP-mediated conversion to vasoinhibin, which can inhibit angiogenesis in DR. Translational Relevance: These findings support the potential therapeutic benefit of levosulpiride against vision loss in diabetes.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Retinopatía Diabética/tratamiento farmacológico , Células Endoteliales , Humanos , Prolactina , Sulpirida/análogos & derivados , Factor A de Crecimiento Endotelial Vascular , Cuerpo Vítreo
16.
Lab Invest ; 100(8): 1068-1079, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32341517

RESUMEN

Increased permeability and growth (angiogenesis) of blood vessels play a key role in joint swelling and pannus formation in inflammatory arthritis, a family of diseases influenced by reproductive hormones. The hormone prolactin (PRL) protects against joint inflammation, pannus formation, and bone destruction in adjuvant-induced arthritis and these effects may involve its proteolytic conversion to vasoinhibin, a PRL fragment that inhibits angiogenesis and vasopermeability. Here, we show that the intra-articular injection of an adeno-associated virus type-2 (AAV2) vector encoding vasoinhibin reduced joint inflammation, the hyperplasia, vascular density, and vasopermeability of the pannus, and the loss of bone in mice subjected to antigen-induced arthritis. In agreement, the AAV2 vasoinhibin vector reduced the expression of proinflammatory cytokines (interleukin-1ß, interleukin-6), an endothelial cell marker (platelet endothelial cell-adhesion molecule 1), and proangiogenic molecules [vascular endothelial growth factor (VEGF), VEGF receptor 2, and hypoxia-inducible factor 1α] in the arthritic joint. Also, vasoinhibin reduced the synovial vasopermeability induced by the intra-articular injection of VEGF in healthy mice. Finally, vasoinhibin signals by blocking the phosphorylation/activation of endothelial nitric oxide synthase (eNOS) at Ser1179 and the AAV2 vasoinhibin vector inhibited the enhanced phosphorylation of eNOS Ser1179 in the arthritic joint. We conclude that vasoinhibin reduces joint inflammation and bone loss in arthritis by inhibiting pannus angiogenesis and vasopermeability via the blockage of VEGF-induced eNOS activation. These findings suggest the potential therapeutic benefit of AAV2-mediated vasoinhibin gene delivery in arthritis.


Asunto(s)
Artritis Experimental/terapia , Permeabilidad Capilar/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Osteítis/prevención & control , Osteoporosis/prevención & control , Prolactina/farmacología , Animales , Antígenos , Artritis Experimental/inducido químicamente , Artritis Experimental/genética , Proteínas de Ciclo Celular/genética , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Humanos , Articulaciones/patología , Masculino , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Osteítis/genética , Osteítis/terapia , Osteoporosis/genética , Osteoporosis/terapia
17.
Trends Endocrinol Metab ; 31(4): 276-286, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32044206

RESUMEN

The pituitary hormone prolactin (PRL) regulates a variety of functions beyond reproduction. The association between physiological (pregnancy) and pathological (prolactinoma) hyperprolactinemia and metabolic alterations led to the concept of this hormone being diabetogenic. However, large cohort clinical studies have recently shown that low circulating PRL levels are associated with metabolic disease and represent a risk factor for type 2 diabetes (T2D), whereas high PRL levels are beneficial. Moreover, PRL acts on the pancreas, liver, adipose tissue, and hypothalamus to maintain and promote metabolic homeostasis. By integrating basic and clinical evidence, we hypothesize that upregulation of PRL levels is a mechanism to maintain metabolic homeostasis and, thus, propose that the range of PRL levels considered physiological should be expanded to higher values.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Dopaminérgicos/farmacología , Homeostasis/fisiología , Hiperprolactinemia/metabolismo , Obesidad/metabolismo , Prolactina/metabolismo , Prolactinoma/metabolismo , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Femenino , Homeostasis/efectos de los fármacos , Humanos , Hiperprolactinemia/sangre , Hiperprolactinemia/tratamiento farmacológico , Obesidad/sangre , Obesidad/tratamiento farmacológico , Prolactina/sangre , Prolactina/efectos de los fármacos , Prolactinoma/sangre , Prolactinoma/tratamiento farmacológico
18.
Neuroendocrinology ; 109(2): 152-164, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31091528

RESUMEN

BACKGROUND: Vasoinhibin, a protein derived from prolactin, regulates various vascular functions including endothelial cell survival. Of note, vasoinhibin is present in the central nervous system, where it triggers neuroendocrine and behavioral responses to stress. Moreover, vasoinhibin compromises nerve growth factor (NGF)-induced neurite outgrowth in primary sensory neurons of the peripheral nervous system. Nonetheless, information on the functions of vasoinhibin in developing neurons remains limited. The present study explored whether vasoinhibin affects the neurotrophic actions of NGF by measuring the cell differentiation and survival of PC12 pheochromocytoma cells. METHODS: The effects of recombinant or lentiviral vector-transduced human vasoinhibin were tested on differentiating PC12 cells. Neurite outgrowth was quantified by measuring their length and density. The MTT assay was employed to assess cell viability, and ELISA was used to quantify DNA fragmentation as an index of apoptosis. Phosphorylated Akt and ERK1/2 were analyzed by Western blotting. RESULTS: The addition of a human recombinant vasoinhibin, and the transduction of a lentiviral vector carrying a human vasoinhibin sequence, significantly reduced NGF-induced neurite outgrowth, cell survival, and phosphorylation of Akt and ERK1/2, and increased DNA fragmentation and caspase 3 activation in PC12 cells. CONCLUSIONS: Vasoinhibin downregulates NGF-induced differentiation and survival of PC12 cells, blocking tropomyosin receptor kinase A-triggered signaling pathways and increasing apoptosis. These results establish that vasoinhibin interaction with NGF and other neurotrophins may be critical in mediating pathways involved in neuronal survival and differentiation.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/patología , Proteínas de Ciclo Celular/fisiología , Diferenciación Celular , Factor de Crecimiento Nervioso/farmacología , Feocromocitoma/patología , Neoplasias de las Glándulas Suprarrenales/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células HEK293 , Humanos , Proyección Neuronal/efectos de los fármacos , Proyección Neuronal/genética , Neuronas/efectos de los fármacos , Neuronas/fisiología , Células PC12 , Feocromocitoma/genética , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transfección
19.
Protein Expr Purif ; 161: 49-56, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31051246

RESUMEN

Vasoinhibin belongs to a family of proteins with antiangiogenic properties derived by proteolytic cleavage from the hormone prolactin (PRL). Vasoinhibin isoforms range from the first 79 to the first 159 residues of PRL. In an attempt to increase the yield of recombinant vasoinhibin and avoid incorrect intra- and inter-disulfide bond formation, the cDNA sequence comprising the first 123 amino acids of human PRL, in which cysteine 58 was or not mutated to serine, was codon-optimized. The optimized constructs achieved a 6-fold increase in mRNA expression but showed no change in protein production and reduced protein secretion when expressed in human embryo kidney (HEK293T/17) cells. Limited vasoinhibin levels associated with the activation of the unfolded protein response (UPR) and endoplasmic reticulum-associated degradation (ERAD) as revealed by the upregulation of UPR (Bip, Xbp-1, and Chop) and ERAD (Hrd1, Os9, and Sel1l) target genes. Mutation to serine introduced a new N-glycosylation site and associated with increased glycosylation and release of glycosylated vasoinhibin isoforms having reduced antiangiogenic properties. We conclude that overexpression and excessive glycosylation lead to protein degradation and reduced bioactivity, respectively, negatively affecting the production of recombinant vasoinhibin in mammalian cells.


Asunto(s)
Prolactina/genética , Prolactina/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Expresión Génica , Glicosilación , Células HEK293 , Humanos , Ingeniería de Proteínas , Proteolisis , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Respuesta de Proteína Desplegada
20.
Sci Rep ; 8(1): 17111, 2018 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-30459448

RESUMEN

Vasoinhibin belongs to a family of angiogenesis inhibitors generated when the fourth α-helix (H4) of the hormone prolactin (PRL) is removed by specific proteolytic cleavage. The antiangiogenic properties are absent in uncleaved PRL, indicating that conformational changes create a new bioactive domain. However, the solution structure of vasoinhibin and the location of its bioactive domain are unknown. Molecular dynamic simulation (MD) showed that the loss of H4 exposes the hydrophobic nucleus of PRL and leads to the compression of the molecule into a three-helix bundle that buries the hydrophobic nucleus again. Compression occurs by the movement of loop 1 (L1) and its interaction with α-helix 1 (H1) generating a new L1 conformation with electrostatic and hydrophobic surfaces distinct from those of PRL, that may correspond to a bioactive domain. Consistent with this model, a recombinant protein containing the first 79 amino acids comprising H1 and L1 of human PRL inhibited the proliferation and migration of endothelial cells and upregulated the vasoinhibin target genes, IL1A and ICAM1. This bioactivity was comparable to that of a conventional vasoinhibin having the 123 residues encompassing H1, L1, Η2, L2, and Η3 of human PRL. These findings extend the vasoinhibin family to smaller proteins and provide important structural information, which will aid in antiangiogenic drug development.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Endotelio Vascular/citología , Proteínas de la Membrana/metabolismo , Neovascularización Fisiológica , Proteínas Tirosina Fosfatasas/metabolismo , Inhibidores de la Angiogénesis/química , Movimiento Celular , Proliferación Celular , Células Cultivadas , Endotelio Vascular/fisiología , Humanos , Proteínas de la Membrana/química , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Proteínas Tirosina Fosfatasas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA