Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
1.
Oncotarget ; 15: 248-254, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38588464

ABSTRACT

Acute myeloid leukemia (AML) is characterized by the rapid proliferation of mutagenic hematopoietic progenitors in the bone marrow. Conventional therapies include chemotherapy and bone marrow stem cell transplantation; however, they are often associated with poor prognosis. Notably, growth hormone-releasing hormone (GHRH) receptor antagonist MIA-602 has been shown to impede the growth of various human cancer cell lines, including AML. This investigation examined the impact of MIA-602 as monotherapy and in combination with Doxorubicin on three Doxorubicin-resistant AML cell lines, KG-1A, U-937, and K-562. The in vitro results revealed a significant reduction in cell viability for all treated wild-type cells. Doxorubicin-resistant clones were similarly susceptible to MIA-602 as the wild-type counterpart. Our in vivo experiment of xenografted nude mice with Doxorubicin-resistant K-562 revealed a reduction in tumor volume with MIA-602 treatment compared to control. Our study demonstrates that these three AML cell lines, and their Doxorubicin-resistant clones, are susceptible to GHRH antagonist MIA-602.


Subject(s)
Growth Hormone-Releasing Hormone , Leukemia, Myeloid, Acute , Sermorelin/analogs & derivatives , Mice , Animals , Humans , Mice, Nude , Cell Proliferation , Cell Line, Tumor , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Doxorubicin/pharmacology
2.
Biol Reprod ; 106(1): 145-154, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34792103

ABSTRACT

Endometrial stromal cells remodeling is critical during human pregnancy. Growth hormone-releasing hormone and its functional receptor have been shown to be expressed in gynecological cancer cells and eutopic endometrial stromal cells. Recent studies have demonstrated the potential clinical uses of antagonists of growth hormone-releasing hormone as effective antitumor agents because of its directly antagonistic effect on the locally produced growth hormone-releasing hormone in gynecological tumors. However, the impact of growth hormone-releasing hormone antagonists on normal endometrial stromal cell growth remained to be elucidated. The aim of this study was to investigate the effect of a growth hormone-releasing hormone antagonist (JMR-132) on cell proliferation and apoptosis of human decidual stromal cells and the underlying molecular mechanisms. Our results showed that growth hormone-releasing hormone and the splice variant 1 of growth hormone-releasing hormone receptor are expressed in human decidual stromal cells isolated from the decidual tissues of early pregnant women receiving surgical abortion. In addition, treatment of stroma cells with JMR-132 induced cell apoptosis with increasing cleaved caspase-3 and caspase-9 activities and decrease cell viability in a time- and dose-dependent manner. Using a dual inhibition approach (pharmacological inhibitors and siRNA-mediated knockdown), we showed that JMR-132-induced activation of apoptotic signals are mediated by the activation of ERK1/2 and JNK signaling pathways and the subsequent upregulation of GADD45alpha. Taken together, JMR-132 suppresses cell survival of decidual stromal cells by inducing apoptosis through the activation of ERK1/2- and JNK-mediated upregulation of GADD45alpha in human endometrial stromal cells. Our findings provide new insights into the potential impact of growth hormone-releasing hormone antagonist on the decidual programming in humans.


Subject(s)
Apoptosis/drug effects , Decidua/cytology , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Stromal Cells/drug effects , Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , Cells, Cultured , Decidua/drug effects , Embryo Implantation/drug effects , Female , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Pregnancy , Sermorelin/analogs & derivatives , Sermorelin/pharmacology , Stromal Cells/physiology , Up-Regulation/drug effects
3.
Mol Cell Endocrinol ; 535: 111379, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34252492

ABSTRACT

The endoplasmic reticulum (ER) stress is one of the mechanisms related to decreased insulin secretion and beta cell death, contributing to the progress of type 2 diabetes mellitus (T2D). Thus, investigating agents that can influence this process would help prevent the development of T2D. Recently, the growth-hormone-releasing hormone (GHRH) action has been demonstrated in INS-1E cells, in which it increases cell proliferation and insulin secretion. As the effects of GHRH and its agonists have not been fully elucidated in the beta cell, we proposed to investigate them by evaluating the role of the GHRH agonist, MR-409, in cells under ER stress. Our results show that the agonist was unable to ameliorate or prevent ER stress. However, cells exposed to the agonist showed less oxidative stress and greater survival even under ER stress. The mechanisms by which GHRH agonist, MR-409, leads to these outcomes require further investigation.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Indoles/adverse effects , Insulin-Secreting Cells/cytology , Sermorelin/analogs & derivatives , Animals , Cell Line , Cell Proliferation/drug effects , Cell Survival , Gene Expression Regulation/drug effects , Growth Hormone-Releasing Hormone/agonists , Growth Hormone-Releasing Hormone/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Sermorelin/pharmacology
4.
Mol Psychiatry ; 26(12): 7465-7474, 2021 12.
Article in English | MEDLINE | ID: mdl-34331008

ABSTRACT

Anxiety and depression have been suggested to increase the risk for post-traumatic stress disorders (PTSD). A link between all these mental illnesses, inflammation and oxidative stress is also well established. Recent behavior studies by our group clearly demonstrate a powerful anxiolytic and antidepressant-like effects of a novel growth hormone releasing hormone (GHRH) antagonist of MIAMI class, MIA-690, probably related to modulatory effects on the inflammatory and oxidative status. In the present work we investigated the potential beneficial effects of MIA-602, another recently developed GHRH antagonist, in mood disorders, as anxiety and depression, and the possible brain pathways involved in its protective activity, in adult mice. MIA-602 exhibited antinflammatory and antioxidant effects in ex vivo and in vivo experimental models, inducing anxiolytic and antidepressant-like behavior in mice subcutaneously treated for 4 weeks. The beneficial effect of MIA-602 on inflammatory and oxidative status and synaptogenesis resulting in anxiolytic and antidepressant-like effects could be related by increases of nuclear factor erythroid 2-related factor 2 (Nrf2) and of brain-derived neurotrophic factor (BDNF) signaling pathways in the hippocampus and prefrontal cortex. These results strongly suggest that GHRH analogs should be tried clinically for the treatment of mood disorders including PTSD.


Subject(s)
Stress Disorders, Post-Traumatic , Animals , Brain-Derived Neurotrophic Factor , Mice , Mood Disorders/drug therapy , Receptors, Neuropeptide , Receptors, Pituitary Hormone-Regulating Hormone , Sermorelin/analogs & derivatives , Sermorelin/pharmacology , Stress Disorders, Post-Traumatic/drug therapy
5.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article in English | MEDLINE | ID: mdl-34244423

ABSTRACT

Optic neuropathies are leading causes of irreversible visual impairment and blindness, currently affecting more than 100 million people worldwide. Glaucoma is a group of optic neuropathies attributed to progressive degeneration of retinal ganglion cells (RGCs). We have previously demonstrated an increase in survival of RGCs by the activation of macrophages, whereas the inhibition of macrophages was involved in the alleviation on endotoxin-induced inflammation by antagonist of growth hormone-releasing hormone (GHRH). Herein, we hypothesized that GHRH receptor (GHRH-R) signaling could be involved in the survival of RGCs mediated by inflammation. We found the expression of GHRH-R in RGCs of adult rat retina. After optic nerve crush, subcutaneous application of GHRH agonist MR-409 or antagonist MIA-602 promoted the survival of RGCs. Both the GHRH agonist and antagonist increased the phosphorylation of Akt in the retina, but only agonist MR-409 promoted microglia activation in the retina. The antagonist MIA-602 reduced significantly the expression of inflammation-related genes Il1b, Il6, and Tnf Moreover, agonist MR-409 further enhanced the promotion of RGC survival by lens injury or zymosan-induced macrophage activation, whereas antagonist MIA-602 attenuated the enhancement in RGC survival. Our findings reveal the protective effect of agonistic analogs of GHRH on RGCs in rats after optic nerve injury and its additive effect to macrophage activation, indicating a therapeutic potential of GHRH agonists for the protection of RGCs against optic neuropathies especially in glaucoma.


Subject(s)
Growth Hormone-Releasing Hormone/agonists , Macrophages/pathology , Neuroprotection , Optic Nerve Injuries/pathology , Retinal Ganglion Cells/pathology , Animals , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Growth Hormone/metabolism , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Inflammation/genetics , Inflammation/pathology , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Neuroprotection/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Inbred F344 , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , STAT3 Transcription Factor/metabolism , Sermorelin/analogs & derivatives , Sermorelin/pharmacology , Signal Transduction/drug effects , Vitreous Body/drug effects , Vitreous Body/metabolism , Zymosan/pharmacology
6.
Peptides ; 142: 170582, 2021 08.
Article in English | MEDLINE | ID: mdl-34051291

ABSTRACT

In addition to its metabolic and endocrine effects, growth hormone-releasing hormone (GHRH) was found to modulate feeding behavior in mammals. However, the role of recently synthetized GHRH antagonist MIA-690 and MR-409, a GHRH agonist, on feeding regulation remains to be evaluated. We investigated the effects of chronic subcutaneous administration of MIA-690 and MR-409 on feeding behavior and energy metabolism, in mice. Compared to vehicle, MIA-690 increased food intake and body weight, while MR-409 had no effect. Both analogs did not modify locomotor activity, as well as subcutaneous, visceral and brown adipose tissue (BAT) mass. A significant increase of hypothalamic agouti-related peptide (AgRP) gene expression and norepinephrine (NE) levels, along with a reduction of serotonin (5-HT) levels were found after MIA-690 treatment. MIA-690 was also found able to decrease gene expression of leptin in visceral adipose tissue. By contrast, MR-409 had no effect on the investigated markers. Concluding, chronic peripheral administration of MIA-690 could play an orexigenic role, paralleled by an increase in body weight. The stimulation of feeding could be mediated, albeit partially, by elevation of AgRP gene expression and NE levels and decreased 5-HT levels in the hypothalamus, along with reduced leptin gene expression, in the visceral adipose tissue.


Subject(s)
Body Weight , Eating , Energy Metabolism , Feeding Behavior/drug effects , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Hypothalamus/drug effects , Sermorelin/analogs & derivatives , Animals , Female , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Sermorelin/pharmacology
7.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article in English | MEDLINE | ID: mdl-33468654

ABSTRACT

Therapies for heart failure with preserved ejection fraction (HFpEF) are lacking. Growth hormone-releasing hormone agonists (GHRH-As) have salutary effects in ischemic and nonischemic heart failure animal models. Accordingly, we hypothesized that GHRH-A treatment ameliorates chronic kidney disease (CKD)-induced HFpEF in a large-animal model. Female Yorkshire pigs (n = 16) underwent 5/6 nephrectomy via renal artery embolization and 12 wk later were randomized to receive daily subcutaneous injections of GHRH-A (MR-409; n = 8; 30 µg/kg) or placebo (n = 8) for 4 to 6 wk. Renal and cardiac structure and function were serially assessed postembolization. Animals with 5/6 nephrectomy exhibited CKD (elevated blood urea nitrogen [BUN] and creatinine) and faithfully recapitulated the hemodynamic features of HFpEF. HFpEF was demonstrated at 12 wk by maintenance of ejection fraction associated with increased left ventricular mass, relative wall thickness, end-diastolic pressure (EDP), end-diastolic pressure/end-diastolic volume (EDP/EDV) ratio, and tau, the time constant of isovolumic diastolic relaxation. After 4 to 6 wk of treatment, the GHRH-A group exhibited normalization of EDP (P = 0.03), reduced EDP/EDV ratio (P = 0.018), and a reduction in myocardial pro-brain natriuretic peptide protein abundance. GHRH-A increased cardiomyocyte [Ca2+] transient amplitude (P = 0.009). Improvement of the diastolic function was also evidenced by increased abundance of titin isoforms and their ratio (P = 0.0022). GHRH-A exerted a beneficial effect on diastolic function in a CKD large-animal model as demonstrated by improving hemodynamic, structural, and molecular characteristics of HFpEF. These findings have important therapeutic implications for the HFpEF syndrome.


Subject(s)
Cardiotonic Agents/pharmacology , Growth Hormone-Releasing Hormone/agonists , Heart Failure/drug therapy , Renal Insufficiency, Chronic/drug therapy , Sermorelin/analogs & derivatives , Stroke Volume/physiology , Animals , Blood Urea Nitrogen , Calcium/metabolism , Connectin/genetics , Connectin/metabolism , Creatinine/blood , Disease Models, Animal , Female , Gene Expression Regulation , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Heart Failure/etiology , Heart Failure/genetics , Heart Failure/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Natriuretic Peptide, Brain/blood , Natriuretic Peptide, Brain/genetics , Nephrectomy/methods , Peptide Fragments/blood , Peptide Fragments/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Renal Insufficiency, Chronic/complications , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Sermorelin/pharmacology , Swine
8.
Horm Metab Res ; 52(5): 336-339, 2020 May.
Article in English | MEDLINE | ID: mdl-32403147

ABSTRACT

Growth hormone releasing hormone is a hypothalamic neuropeptide, which regulates the release of growth hormone from the anterior pituitary gland. Growth hormone releasing hormone antagonists are anticancer agents, associated with strong anti-inflammatory activities. In the present study, we investigated the effects of the GHRH antagonist MIA-602 in the integrity of the brain microvascular endothelium in vitro. Our observations suggest that MIA-602 protects against the H2O2-induced breakdown of the brain endothelium and enhances its integrity by inducing P53, deactivating cofilin, and suppressing the RhoA inflammatory pathway. Thus, GHRH antagonists may offer an exciting possibility for the treatment of pathologies related to the blood brain barrier dysfunction, including the Parkinson's and Alzheimer's diseases.


Subject(s)
Brain/blood supply , Endothelium/pathology , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Hydrogen Peroxide/toxicity , Microvessels/pathology , Neuroprotective Agents/pharmacology , Actins/metabolism , Endothelium/drug effects , HeLa Cells , Humans , Sermorelin/analogs & derivatives , Sermorelin/pharmacology , Tumor Suppressor Protein p53/metabolism
9.
Clin Exp Pharmacol Physiol ; 47(9): 1632-1634, 2020 09.
Article in English | MEDLINE | ID: mdl-32289177

ABSTRACT

Growth hormone-releasing hormone is a hypothalamic neuropeptide, which regulates the secretion of growth hormone by the anterior pituitary gland. Recent evidence suggest that it exerts growth factor activities in a diverse variety of in vivo and in vitro experimental malignancies, which are counteracted by growth hormone-releasing hormone antagonists. Those peptides support lung endothelial barrier integrity by suppressing major inflammatory pathways and by inducing the endothelial defender P53. The present effort provides information regarding the effects of growth hormone-releasing hormone in the regulation of P53 and the unfolded protein response. Furthermore, it suggests the possible application of growth hormone-releasing hormone antagonists towards the management of acute lung injury, including the lethal acute respiratory distress syndrome.


Subject(s)
Acute Lung Injury/metabolism , Growth Hormone-Releasing Hormone/metabolism , Lung/metabolism , Tumor Suppressor Protein p53/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/therapeutic use , Humans , Lung/drug effects , Lung/pathology , Sermorelin/analogs & derivatives , Sermorelin/therapeutic use , Signal Transduction , Unfolded Protein Response
10.
Proc Natl Acad Sci U S A ; 117(12): 6726-6732, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32156725

ABSTRACT

The extrahypothalamic growth hormone-releasing hormone (GHRH) and its cognate receptors (GHRH-Rs) and splice variants are expressed in a variety of cancers. It has been shown that the pituitary type of GHRH-R (pGHRH-R) mediates the inhibition of tumor growth induced by GHRH-R antagonists. However, GHRH-R antagonists can also suppress some cancers that do not express pGHRH-R, yet the underlying mechanisms have not been determined. Here, using human esophageal squamous cell carcinoma (ESCC) as a model, we were able to reveal that SV1, a known splice variant of GHRH-R, is responsible for the inhibition induced by GHRH-R antagonist MIA-602. We demonstrated that GHRH-R splice variant 1 (SV1) is a hypoxia-driven promoter of tumor progression. Hypoxia-elevated SV1 activates a key glycolytic enzyme, muscle-type phosphofructokinase (PFKM), through the nuclear factor kappa B (NF-κB) pathway, which enhances glycolytic metabolism and promotes progression of ESCC. The malignant actions induced by the SV1-NF-κB-PFKM pathway could be reversed by MIA-602. Altogether, our studies demonstrate a mechanism by which GHRH-R antagonists target SV1. Our findings suggest that SV1 is a hypoxia-induced oncogenic promoter which can be an alternative target of GHRH-R antagonists.


Subject(s)
Biomarkers, Tumor/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Regulation, Neoplastic , Receptors, LHRH/genetics , Sermorelin/analogs & derivatives , Alternative Splicing , Animals , Apoptosis , Cell Proliferation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Female , Glycolysis , Humans , Mice , Mice, Nude , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphofructokinase-1, Muscle Type/genetics , Phosphofructokinase-1, Muscle Type/metabolism , Receptors, LHRH/antagonists & inhibitors , Sermorelin/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Proc Natl Acad Sci U S A ; 117(11): 6067-6074, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32123064

ABSTRACT

Ocular inflammation is a major cause of visual impairment attributed to dysregulation of the immune system. Previously, we have shown that the receptor for growth-hormone-releasing hormone (GHRH-R) affects multiple inflammatory processes. To clarify the pathological roles of GHRH-R in acute ocular inflammation, we investigated the inflammatory cascades mediated by this receptor. In human ciliary epithelial cells, the NF-κB subunit p65 was phosphorylated in response to stimulation with lipopolysaccharide (LPS), resulting in transcriptional up-regulation of GHRH-R. Bioinformatics analysis and coimmunoprecipitation showed that GHRH-R had a direct interaction with JAK2. JAK2, but not JAK1, JAK3, and TYK2, was elevated in ciliary body and iris after treatment with LPS in a rat model of endotoxin-induced uveitis. This elevation augmented the phosphorylation of STAT3 and production of proinflammatory factors, including IL-6, IL-17A, COX2, and iNOS. In explants of iris and ciliary body, the GHRH-R antagonist, MIA-602, suppressed phosphorylation of STAT3 and attenuated expression of downstream proinflammatory factors after LPS treatment. A similar suppression of STAT3 phosphorylation was observed in human ciliary epithelial cells. In vivo studies showed that blocking of the GHRH-R/JAK2/STAT3 axis with the JAK inhibitor Ruxolitinib alleviated partially the LPS-induced acute ocular inflammation by reducing inflammatory cells and protein leakage in the aqueous humor and by repressing expression of STAT3 target genes in rat ciliary body and iris and in human ciliary epithelial cells. Our findings indicate a functional role of the GHRH-R/JAK2/STAT3-signaling axis in acute anterior uveitis and suggest a therapeutic strategy based on treatment with antagonists targeting this signaling pathway.


Subject(s)
Epithelial Cells/pathology , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Signal Transduction/immunology , Uveitis/pathology , Animals , Cell Line , Ciliary Body/cytology , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/immunology , Humans , Janus Kinase 2/metabolism , Lipopolysaccharides/immunology , Male , Nitriles , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines , Rats , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Neuropeptide/immunology , Receptors, Pituitary Hormone-Regulating Hormone/antagonists & inhibitors , Receptors, Pituitary Hormone-Regulating Hormone/immunology , STAT3 Transcription Factor/metabolism , Sermorelin/analogs & derivatives , Sermorelin/pharmacology , Sermorelin/therapeutic use , Signal Transduction/drug effects , Uveitis/drug therapy , Uveitis/immunology
12.
Sci Rep ; 10(1): 732, 2020 01 20.
Article in English | MEDLINE | ID: mdl-31959947

ABSTRACT

Growth hormone-releasing hormone (GHRH) antagonist MIA-690 and GHRH agonist MR-409, previously synthesized and developed by us have demonstrated potent antitumor effects. However, little is known about the effects of these analogs on brain functions. We investigated the potential antinflammatory and antioxidant effects of GHRH antagonist MIA-690 and GHRH agonist MR-409, on isolated mouse prefrontal cortex specimens treated with lipopolysaccharide (LPS). Additionally, we studied their effects on emotional behavior after chronic in vivo treatment. Ex vivo, MIA-690 and MR-409 inhibited LPS-induced inflammatory and pro-oxidative markers. In vivo, both MIA-690 and MR-409 induced anxiolytic and antidepressant-like effects, increased norepinephrine and serotonin levels and decreased nuclear factor-kB, tumor necrosis factor-α and interleukin-6 gene expression in prefrontal cortex. Increased nuclear factor erythroid 2-related factor 2 expression was also found in mice treated with MIA-690 and MR-409. MIA-690 showed higher efficacy in inhibiting all tested inflammatory and oxidative markers. In addition, MR-409 induced a down regulation of the gene and protein expression of pituitary-type GHRH-receptor in prefrontal cortex of mice after 4 weeks of treatment at 5 µg/day. In conclusion, our results demonstrate anxiolytic and antidepressant-like effects of GHRH analogs that could involve modulatory effects on monoaminergic signaling, inflammatory and oxidative status.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Behavior, Animal/drug effects , Emotions/drug effects , Growth Hormone-Releasing Hormone/agonists , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Sermorelin/analogs & derivatives , Animals , Anti-Anxiety Agents , Antidepressive Agents , Gene Expression/drug effects , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Norepinephrine/metabolism , Prefrontal Cortex/metabolism , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/genetics , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Sermorelin/pharmacology , Serotonin/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
13.
Proc Natl Acad Sci U S A ; 116(44): 22269-22274, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31611400

ABSTRACT

Adrenocortical carcinomas (ACCs) are rare and highly malignant cancers associated with poor survival of patients. Currently, mitotane, a nonspecific derivative of the pesticide DDT (1,1-(dichlorobiphenyl)-2,2-dichloroethane), is used as the standard treatment, but its mechanism of action in ACCs remains elusive. Here we demonstrate that the human ACC NCI-H295R cell line is remarkably sensitive to induction of ferroptosis, while mitotane does not induce this iron-dependent mode of regulated necrosis. Supplementation with insulin, transferrin, and selenium (ITS) is commonly used to keep NCI-H295R cells in cell culture. We show that this supplementation prevents spontaneous ferroptosis, especially when it contains polyunsaturated fatty acids (PUFAs), such as linoleic acid. Inhibitors of apoptosis (zVAD, emricasan) do not prevent the mitotane-induced cell death but morphologically prevent membrane blebbing. The expression of glutathione peroxidase 4 (GPX4) in H295R cells, however, is significantly higher when compared to HT1080 fibrosarcoma cells, suggesting a role for ferroptosis. Direct inhibition of GPX4 in H295R cells led to high necrotic populations compared to control, while cotreatment with ferrostatin-1 (Fer-1) completely reverted ferroptosis. Interestingly, the analysis of public databases revealed that several key players of the ferroptosis pathway are hypermethylated and/or mutated in human ACCs. Finally, we also detected that growth hormone-releasing hormone (GHRH) antagonists, such as MIA602, kill H295R cells in a nonapoptotic manner. In summary, we found elevated expression of GPX4 and higher sensitivity to ferroptosis in ACCs. We hypothesize that instead of treatment with mitotane, human adrenocortical carcinomas may be much more sensitive to induction of ferroptosis.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Adrenocortical Carcinoma/metabolism , Ferroptosis/drug effects , 3T3 Cells , Animals , Apoptosis/drug effects , HEK293 Cells , HT29 Cells , Humans , Insulin/metabolism , Iron/metabolism , Linoleic Acid/metabolism , Mice , Mitotane/toxicity , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Selenium/metabolism , Sermorelin/analogs & derivatives , Sermorelin/pharmacology , Transferrin/metabolism
14.
Lung ; 197(5): 541-549, 2019 10.
Article in English | MEDLINE | ID: mdl-31392398

ABSTRACT

PURPOSE: Growth hormone-releasing hormone (GHRH) is a 44-amino acid peptide that regulates growth hormone (GH) secretion. We hypothesized that a GHRH receptor (GHRH-R) antagonist, MIA-602, would inhibit bleomycin-induced lung inflammation and/or fibrosis in C57Bl/6J mice. METHODS: We tested whether MIA-602 (5 µg or vehicle given subcutaneously [SC] on days 1-21) would decrease lung inflammation (at day 14) and/or fibrosis (at day 28) in mice treated with intraperitoneal (IP) bleomycin (0.8 units on days 1, 3, 7, 10, 14, and 21). Bleomycin resulted in inflammation and fibrosis around airways and vessels evident histologically at days 14 and 28. RESULTS: Inflammation (histopathologic scores assessed blindly) was visibly less evident in mice treated with MIA-602 for 14 days. After 28 days, lung hydroxyproline (HP) content increased significantly in mice treated with vehicle; in contrast, lung HP did not increase significantly compared to naïve controls in mice treated with GHRH-R antagonist. GHRH-R antagonist increased basal and maximal oxygen consumption of cultured lung fibroblasts. Multiple genes related to chemotaxis, IL-1, chemokines, regulation of inflammation, and extracellular signal-regulated kinases (ERK) were upregulated in lungs of mice treated with bleomycin and MIA-602. MIA-602 also prominently suppressed multiple genes related to the cellular immune response including those for T-cell differentiation, receptor signaling, activation, and cytokine production. CONCLUSIONS: MIA-602 reduced lung inflammation and fibrosis due to bleomycin. Multiple genes related to immune response and T-cell functions were downregulated, supporting the view that MIA-602 can modulate the cellular immune response to bleomycin lung injury.


Subject(s)
Bleomycin , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/pharmacology , Lung/drug effects , Pneumonia/prevention & control , Pulmonary Fibrosis/prevention & control , Sermorelin/analogs & derivatives , Animals , Cells, Cultured , Cytoprotection , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Growth Hormone-Releasing Hormone/metabolism , Hydroxyproline/metabolism , Inflammation Mediators/metabolism , Lung/metabolism , Lung/pathology , Male , Mice, Inbred C57BL , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Sermorelin/pharmacology , Signal Transduction
15.
Invest Ophthalmol Vis Sci ; 59(12): 5060-5066, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30357400

ABSTRACT

Purpose: The aim of the study was to investigate the signaling of growth hormone-releasing hormone receptor (GHRH-R) in the pathogenesis of pterygium and determine the apoptotic effect of GHRH-R antagonist on pterygium epithelial cells (PECs). Methods: Fourteen samples of primary pterygium of grade T3 with size of corneal invasion ≥ 4 mm were obtained for investigation by histology, immunofluorescence, electron microscopy, explant culture, and flow cytometry. Results: We found that PECs were localized in the basal layer of the epithelium in advancing regions of the head of pterygium. These cells harbored clusters of rough endoplasmic reticulum, ribosomes, and mitochondria, which were consistent with their aggressive proliferation. Immunofluorescence studies and Western blots showed that GHRH-R and the downstream growth hormone receptor (GH-R) were intensively expressed in PECs. Their respective ligands, GHRH and GH, were also elevated in the pterygium tissues as compared to conjunctival cells. Explanted PECs were strongly immunoreactive to GHRH-R and exhibited differentiation and proliferation that led to lump formation. Treatment with GHRH-R antagonist MIA-602 induced apoptosis of PECs in a dose-dependent manner, which was accompanied by a downregulation of ERK1 and upregulation of Caspase 3 expression. Conclusions: Our results revealed that GHRH-R signaling is involved in survival and proliferation of PECs and suggest a potential therapeutic approach for GHRH-R antagonist in the treatment of pterygium.


Subject(s)
Apoptosis/drug effects , Pterygium/pathology , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Pituitary Hormone-Regulating Hormone/antagonists & inhibitors , Sermorelin/analogs & derivatives , Blotting, Western , Caspase 3/metabolism , Cell Count , Cell Proliferation , Cell Survival , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fluorescent Antibody Technique, Indirect , Growth Hormone/metabolism , Growth Hormone-Releasing Hormone/metabolism , Humans , Microscopy, Electron, Transmission , Mitogen-Activated Protein Kinase 3/metabolism , Pterygium/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Neuropeptide/metabolism , Receptors, Pituitary Hormone-Regulating Hormone/metabolism , Receptors, Somatotropin/metabolism , Sermorelin/pharmacology , Signal Transduction
16.
Proc Natl Acad Sci U S A ; 115(47): 12028-12033, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30373845

ABSTRACT

The effects of the growth hormone-releasing hormone (GHRH) agonist MR409 on various human cancer cells were investigated. In H446 small cell lung cancer (SCLC) and HCC827 and H460 (non-SCLC) cells, MR409 promoted cell viability, reduced cell apoptosis, and induced the production of cellular cAMP in vitro. Western blot analyses showed that treatment of cancer cells with MR409 up-regulated the expression of cyclins D1 and D2 and cyclin-dependent kinases 4 and 6, down-regulated p27kip1, and significantly increased the expression of the pituitary-type GHRH receptor (pGHRH-R) and its splice-variant (SV1). Hence, in vitro MR409 exerts agonistic action on lung cancer cells in contrast to GHRH antagonists. However, in vivo, MR409 inhibited growth of lung cancers xenografted into nude mice. MR409 given s.c. at 5 µg/day for 4 to 8 weeks significantly suppressed growth of HCC827, H460, and H446 tumors by 48.2%, 48.7%, and 65.6%, respectively. This inhibition of tumor growth by MR409 was accompanied by the down-regulation of the expression of pGHRH-R and SV1 in the pituitary gland and tumors. Tumor inhibitory effects of MR409 in vivo were also observed in other human cancers, including gastric, pancreatic, urothelial, prostatic, mammary, and colorectal. This inhibition of tumor growth parallel to the down-regulation of GHRH-Rs is similar and comparable to the suppression of sex hormone-dependent cancers after the down-regulation of receptors for luteinizing hormone-releasing hormone (LHRH) by LHRH agonists. Further oncological investigations with GHRH agonists are needed to elucidate the underlying mechanisms.


Subject(s)
Receptors, Neuropeptide/drug effects , Receptors, Pituitary Hormone-Regulating Hormone/drug effects , Sermorelin/analogs & derivatives , Alternative Splicing/drug effects , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Female , Growth Hormone-Releasing Hormone/agonists , Growth Hormone-Releasing Hormone/pharmacology , Humans , Mice , Mice, Nude , RNA Splicing/drug effects , Sermorelin/metabolism , Sermorelin/pharmacology , Small Cell Lung Carcinoma/metabolism , Xenograft Model Antitumor Assays/methods
17.
Prostate ; 78(12): 915-926, 2018 09.
Article in English | MEDLINE | ID: mdl-29748961

ABSTRACT

BACKGROUND: Therapeutic strategies should be designed to transform aggressive prostate cancer phenotypes to a chronic situation. To evaluate the effects of the new growth hormone-releasing hormone receptor (GHRH-R) antagonists: MIA-602, MIA-606, and MIA-690 on processes associated with cancer progression as cell proliferation, adhesion, migration, and angiogenesis. METHODS: We used three human prostate cell lines (RWPE-1, LNCaP, and PC3). We analyzed several molecules such as E-cadherin, ß-catenin, Bcl2, Bax, p53, MMP2, MMP9, PCNA, and VEGF and signaling mechanisms that are involved on effects exerted by GHRH-R antagonists. RESULTS: GHRH-R antagonists decreased cell viability and provoked a reduction in proliferation in LNCaP and PC3 cells. Moreover, GHRH-R antagonists caused a time-dependent increase of cell adhesion in all three cell lines and retarded the wound closure with the highest value with MIA-690 in PC3 cells. GHRH-R antagonists also provoked a large number of cells in SubG0 phase revealing an increase in apoptotic cells in PC3 cell line. CONCLUSIONS: Taken all together, GHRH-R antagonists of the MIAMI series appear to be inhibitors of tumor progression in prostate cancer and should be considered for use in future therapeutic strategies on this malignancy.


Subject(s)
Prostatic Neoplasms/drug therapy , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Pituitary Hormone-Regulating Hormone/antagonists & inhibitors , Sermorelin/analogs & derivatives , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Male , PC-3 Cells , Prostatic Neoplasms/pathology , Resting Phase, Cell Cycle , Sermorelin/pharmacology , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/metabolism , Wound Healing/drug effects , beta Catenin/analysis
18.
Br J Haematol ; 181(4): 476-485, 2018 05.
Article in English | MEDLINE | ID: mdl-29663325

ABSTRACT

Growth hormone-releasing hormone (GHRH) is secreted by the hypothalamus and acts on the pituitary gland to stimulate the release of growth hormone (GH). GHRH can also be produced by human cancers, in which it functions as an autocrine/paracrine growth factor. We have previously shown that synthetic antagonistic analogues of GHRH are able to successfully suppress the growth of 60 different human cancer cell lines representing over 20 cancers. Nevertheless, the expression of GHRH and its receptors in leukaemias has never been examined. Our study demonstrates the presence of GHRH receptor (GHRH-R) on 3 of 4 human acute myeloid leukaemia (AML) cell lines-K-562, THP-1, and KG-1a-and significant inhibition of proliferation of these three cell lines in vitro following incubation with the GHRH antagonist MIA-602. We further show that this inhibition of proliferation is associated with the upregulation of pro-apoptotic genes and inhibition of Akt signalling in leukaemic cells. Treatment with MIA-602 of mice bearing xenografts of these human AML cell lines drastically reduced tumour growth. The expression of GHRH-R was further confirmed in 9 of 9 samples from patients with AML. These findings offer a new therapeutic approach to this malignancy and suggest a possible role of GHRH-R signalling in the pathology of AML.


Subject(s)
Apoptosis/drug effects , Drug Delivery Systems/methods , Leukemia, Myeloid, Acute/drug therapy , Receptors, Neuropeptide/antagonists & inhibitors , Receptors, Pituitary Hormone-Regulating Hormone/antagonists & inhibitors , Sermorelin/analogs & derivatives , Signal Transduction/drug effects , Animals , Female , Humans , K562 Cells , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Nude , Proto-Oncogene Proteins c-akt/metabolism , Sermorelin/pharmacology , THP-1 Cells , Xenograft Model Antitumor Assays
19.
Int J Cancer ; 142(11): 2394-2404, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29435973

ABSTRACT

We investigated the effects of novel antagonists of growth hormone releasing hormone (GHRH)-MIA602 and MIA690-on three human small cell lung cancer (SCLC) lines (H446, DMS53 and H69) and two non-SCLC (NSCLC) lines (HCC827 and H460). In vitro exposure of cancer cells to these GHRH antagonists significantly inhibited cell viability, increased cell apoptosis, decrease cellular levels of cAMP and reduced cell migration. In vivo, the antagonists strongly inhibited tumor growth in xenografted nude mice models. Subcutaneous administration of MIA602 at the dose of 5 µg/day for 4-8 weeks reduced the growth of HCC827, H460 and H446 tumors by 69.9%, 68.3% and 53.4%, respectively, while MIA690 caused a reduction of 76.8%, 58.3% and 54.9%, respectively. Western blot and qRT-PCR analyses demonstrated a downregulation of expression of the pituitary-type GHRH-R and its splice-variant, cyclinD1/2, cyclin-dependent kinase4/6, p21-activated kinase-1, phosphorylation of activator of transcription 3 and cAMP response element binding protein; and an upregulation of expression of E-cadherin, ß-catenin and P27kip1 in cancer cells and in xenografted tumor tissues. The study demonstrates the involvement of GHRH antagonists in multiple signaling pathways in lung cancers. Our findings suggest the merit of further investigation with these GHRH antagonists on the management of both SCLC and NSCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Sermorelin/analogs & derivatives , Small Cell Lung Carcinoma/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Female , Gene Expression , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Humans , Mice , Mice, Nude , STAT3 Transcription Factor/metabolism , Sermorelin/pharmacology , Signal Transduction/drug effects , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Xenograft Model Antitumor Assays
20.
Proc Natl Acad Sci U S A ; 114(50): 13248-13253, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29180438

ABSTRACT

The potential therapeutic effects of agonistic analogs of growth hormone-releasing hormone (GHRH) and their mechanism of action were investigated in diabetic retinopathy (DR). Streptozotocin-induced diabetic rats (STZ-rats) were treated with 15 µg/kg GHRH agonist, MR-409, or GHRH antagonist, MIA-602. At the end of treatment, morphological and biochemical analyses assessed the effects of these compounds on retinal neurovascular injury induced by hyperglycemia. The expression levels of GHRH and its receptor (GHRH-R) measured by qPCR and Western blotting were significantly down-regulated in retinas of STZ-rats and in human diabetic retinas (postmortem) compared with their respective controls. Treatment of STZ-rats with the GHRH agonist, MR-409, prevented retinal morphological alteration induced by hyperglycemia, particularly preserving survival of retinal ganglion cells. The reverse, using the GHRH antagonist, MIA-602, resulted in worsening of retinal morphology and a significant alteration of the outer retinal layer. Explaining these results, we have found that MR-409 exerted antioxidant and anti-inflammatory effects in retinas of the treated rats, as shown by up-regulation of NRF-2-dependent gene expression and down-regulation of proinflammatory cytokines and adhesion molecules. MR-409 also significantly down-regulated the expression of vascular endothelial growth factor while increasing that of pigment epithelium-derived factor in diabetic retinas. These effects correlated with decreased vascular permeability. In summary, our findings suggest a neurovascular protective effect of GHRH analogs during the early stage of diabetic retinopathy through their antioxidant and anti-inflammatory properties.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Diabetic Retinopathy/drug therapy , Growth Hormone-Releasing Hormone/agonists , Sermorelin/analogs & derivatives , Animals , Anti-Inflammatory Agents/therapeutic use , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cytokines/genetics , Cytokines/metabolism , Diabetic Retinopathy/metabolism , GA-Binding Protein Transcription Factor/genetics , GA-Binding Protein Transcription Factor/metabolism , Growth Hormone-Releasing Hormone/antagonists & inhibitors , Humans , Male , Rats , Rats, Sprague-Dawley , Retina/drug effects , Retina/metabolism , Sermorelin/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL