Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 495
Filtrar
1.
Redox Rep ; 29(1): 2391139, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39138590

RESUMEN

Oxalate-induced damage to renal tubular epithelial cells (RTECs) is an essential factor in the incident kidney stone, but the specific mechanism is unclear. Recent research has pinpointed interacting areas within the endoplasmic reticulum and mitochondria, called mitochondria-associated membranes (MAMs). These studies have linked endoplasmic reticulum stress (ERS) and oxidative imbalance to kidney disease development. The sigma-1 receptor (S1R), a specific protein found in MAMs, is involved in various physiological processes, but its role in oxalate-induced kidney stone formation remains unclear. In this study, we established cellular and rat models of oxalate-induced kidney stone formation to elucidate the S1R's effects against ERS and apoptosis and its mechanism in oxalate-induced RTEC injury. We found that oxalate downregulated S1R expression in RTECs and escalated oxidative stress and ERS, culminating in increased apoptosis. The S1R agonist dimemorfan up-regulated S1R expression and mitigated ERS and oxidative stress, thereby reducing apoptosis. This protective effect was mediated through S1R inhibition of the CHOP pathway. Animal experiments demonstrated that S1R's activation attenuated oxalate-induced kidney injury and alleviated kidney stone formation. This is the first study to establish the connection between S1R and kidney stones, suggesting S1R's protective role in inhibiting ERS-mediated apoptosis to ameliorate kidney stone formation.


Asunto(s)
Apoptosis , Estrés del Retículo Endoplásmico , Retículo Endoplásmico , Células Epiteliales , Túbulos Renales , Mitocondrias , Nefrolitiasis , Receptores sigma , Receptor Sigma-1 , Animales , Masculino , Ratas , Apoptosis/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Túbulos Renales/metabolismo , Túbulos Renales/patología , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Nefrolitiasis/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Receptores sigma/metabolismo
2.
Funct Integr Genomics ; 24(4): 134, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39107544

RESUMEN

Distal hereditary motor neuropathy (dHMN) is a progressive neurological disease characterized by distal limb muscle weakness and amyotrophy. Sigma 1 receptor (σ1R), a gene product of SIGMAR1, mutations have been reported to induce dHMN, but its mechanism remains unknown. This study aims to explore the effect of C238T and 31_50del mutations in σ1R on neuronal SH-SY5Y cell functions. The SH-SY5Y cells that overexpressed σ1R, C238T mutant σ1R (σ1RC238T) or 31_50del mutant σ1R (σ1R31_50del) were constructed by pEGFPN1 vectors. We used Western blot (WB) and immunofluorescence (IF) staining to detect the expression of σ1R and green fluorescent proteins (GFP). Then, we evaluated the impact of σ1R mutation on apoptosis, autophagy, endoplasmic reticulum stress, and the involvement of the unfolded protein response (UPR) pathway in SH-SY5Y cells. We found that σ1RC238T and σ1R31_50del downregulated σ1R and promoted the apoptosis of SH-SY5Y cells. σ1RC238T and σ1R31_50del increased p-PERK, p-eIF2α, p-JNK, BIP, ATF4, CHOP, ATF6, XBP1, Caspase3, Caspase12 expressions and Ca2+ concentration, whereas decreased ATP content in SH-SY5Y cells. Besides, the expressions of LC3B, Lamp1, ATG7, Beclin-1 and phosphorylation of AMPK and ULK1 were increased, while the p62 level decreased after C238T or 31_50del mutation of σ1R. Additionally, AMPK knockdown abolished the apoptosis mediated by σ1RC238T or σ1R31_50del in SH-SY5Y cells. Our results indicated that C238T or 31_50del mutation in σ1R promoted motor neuron apoptosis through the AMPK/ULK1 pathway in dHMN. This study shed light on a better understanding of the neurons pathological mechanisms mediated by σ1R C238T and σ1R 31-50del in dHMN.


Asunto(s)
Apoptosis , Homólogo de la Proteína 1 Relacionada con la Autofagia , Autofagia , Estrés del Retículo Endoplásmico , Receptores sigma , Receptor Sigma-1 , Humanos , Receptores sigma/metabolismo , Receptores sigma/genética , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Homólogo de la Proteína 1 Relacionada con la Autofagia/genética , Línea Celular Tumoral , Transducción de Señal , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Respuesta de Proteína Desplegada , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación
3.
Bioorg Med Chem Lett ; 110: 129885, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-38996940

RESUMEN

Herein, we report the synthesis of new 4-amino-2-(piperidin-3-yl)isoindoline-1,3-diones and their biological evaluation in a series of in vitro experiments. The synthetic production of these materials was initiated upon the condensation of appropriate nitrophthalic acid derivatives with various 3-aminopiperidines; subsequent reduction provided the final products in moderate to good yields. Readily available chiral pool reagents facilitated entry into optically enriched samples, while the piperidine scaffold furnished a variety of amide and alkylated entries. In total, 16 candidates were produced, and their ensuing treatment in LPS-challenged RAW cells effected slight reductions in secreted TNF-α but provided more robust and dose-dependent declines in nitrite and IL-6 levels relative to basal amounts, all concurrent with maintenance of cellular viability across the concentration ranges screened. The secondary amine cohort including rac-6, (R)-7, and (S)-8 rendered the most pronounced dose-dependent reductions in nitrite and IL-6. When dosed at 30 µM, (R)-7 demonstrated the most compelling effects, with decreases of 32 % and 40 % for nitrite and IL-6, respectively. Notable reductions in the inflammatory markers were also observed for 19 which effected declines in TNF-α (14 %), nitrite (19 %), and IL-6 (11 %) when treated at 30 µM. Additionally, four representative compounds were further evaluated against numerous CNS receptors, channels, and transporters, with 6, 9, and 19 demonstrating varying degrees of nanomolar-to-low-micromolar binding to the σ-1 and σ-2 receptors and also to serotonin receptors 5HT2A, 5HT2B and 5HT3. In this regard, 6 displayed perhaps the most noteworthy affinities, with binding at σ-2 (Ki = 2.2uM), 5HT2B (Ki = 561 nM) and 5HT3 (Ki = 536 nM). Furthermore, no pronounced or dose-dependent Cereblon/DDB1 binding was observed for the screened representative compounds 6, 9, 18 and 19.


Asunto(s)
Inflamación , Lipopolisacáridos , Receptores de Serotonina , Receptores sigma , Animales , Lipopolisacáridos/farmacología , Lipopolisacáridos/antagonistas & inhibidores , Ratones , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Receptores de Serotonina/metabolismo , Receptores sigma/metabolismo , Células RAW 264.7 , Piperidinas/química , Piperidinas/farmacología , Piperidinas/síntesis química , Relación Dosis-Respuesta a Droga , Estructura Molecular , Relación Estructura-Actividad , Interleucina-6/metabolismo , Supervivencia Celular/efectos de los fármacos , Biomarcadores/metabolismo
4.
Nat Commun ; 15(1): 5619, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965213

RESUMEN

The sigma-1 receptor (σ1R) is a non-opioid membrane receptor, which responds to a diverse array of synthetic ligands to exert various pharmacological effects. Meanwhile, candidates for endogenous ligands of σ1R have also been identified. However, how endogenous ligands bind to σ1R remains unknown. Here, we present crystal structures of σ1R from Xenopus laevis (xlσ1R) bound to two endogenous neurosteroid ligands, progesterone (a putative antagonist) and dehydroepiandrosterone sulfate (DHEAS) (a putative agonist), at 2.15-3.09 Å resolutions. Both neurosteroids bind to a similar location in xlσ1R mainly through hydrophobic interactions, but surprisingly, with opposite binding orientations. DHEAS also forms hydrogen bonds with xlσ1R, whereas progesterone interacts indirectly with the receptor through water molecules near the binding site. Binding analyses are consistent with the xlσ1R-neurosteroid complex structures. Furthermore, molecular dynamics simulations and structural data reveal a potential water entry pathway. Our results provide insight into binding of two endogenous neurosteroid ligands to σ1R.


Asunto(s)
Sulfato de Deshidroepiandrosterona , Simulación de Dinámica Molecular , Progesterona , Receptores sigma , Receptor Sigma-1 , Xenopus laevis , Receptores sigma/metabolismo , Receptores sigma/química , Animales , Ligandos , Sitios de Unión , Progesterona/metabolismo , Progesterona/química , Sulfato de Deshidroepiandrosterona/metabolismo , Sulfato de Deshidroepiandrosterona/química , Unión Proteica , Cristalografía por Rayos X , Neuroesteroides/metabolismo , Neuroesteroides/química , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas
5.
Biochim Biophys Acta Mol Basis Dis ; 1870(7): 167284, 2024 10.
Artículo en Inglés | MEDLINE | ID: mdl-38851304

RESUMEN

AIM: Methamphetamine (METH) chronic exposure is an important risk factor for hypertension development. However, the mechanisms behind METH-induced hypertension remain unclear. Therefore, we aimed to reveal the potential mechanisms underlying METH-induced hypertension. METHODS AND RESULTS: We structured the mouse hypertension model by METH, and observed that METH-treated mice have presented vascular remodeling (large-and small-size arteries) with collagen deposit around the vessel and increasing blood pressure (BP) and Sigma1 receptor (Sigmar1) in vascular tissue. We hypothesized that Sigmar1 is crucial in METH-induced hypertension and vascular remodeling. Sigmar1 knockout (KO) mice and antagonist (BD1047) pretreated mice exposed to METH for six-week showed higher BP and more collagen deposited around vessels than wild-type (WT) mice exposed to METH for six-week, in contrast, mice pretreated with Sigmar1 agonist (PRE-084) had unchanged BP and perivascular collagen despite the six-week METH exposure. Furthermore, we found that METH exposure induced vascular smooth muscle cells (VSMCs) and mesenchymal stem cells to differentiate into the myofibroblast-like cell and secrete collagen into surrounding vessels. Mechanically, Sigmar1 can suppress the COL1A1 expression by blocking the classical fibrotic TGF-ß/Smad2/3 signaling pathway in METH-exposed VSMCs and mesenchymal stem cells. CONCLUSION: Our results suggest that Sigmar1 is involved in METH-induced hypertension and vascular fibrosis by blocking the activation of the TGF-ß/Smad2/3 signaling pathway. Accordingly, Sigmar1 may be a novel therapeutic target for METH-induced hypertension and vascular fibrosis.


Asunto(s)
Hipertensión , Metanfetamina , Músculo Liso Vascular , Receptores sigma , Receptor Sigma-1 , Animales , Masculino , Ratones , Presión Sanguínea/efectos de los fármacos , Colágeno/metabolismo , Modelos Animales de Enfermedad , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Hipertensión/patología , Hipertensión/genética , Células Madre Mesenquimatosas/metabolismo , Metanfetamina/efectos adversos , Metanfetamina/toxicidad , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Miocitos del Músculo Liso/efectos de los fármacos , Receptores sigma/metabolismo , Receptores sigma/genética , Transducción de Señal/efectos de los fármacos , Remodelación Vascular/efectos de los fármacos
6.
Arch Toxicol ; 98(10): 3323-3336, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38896176

RESUMEN

Ochratoxin A (OTA), a secondary fungal metabolite known for its nephrotoxic effects, is prevalent in various feeds and food items. Our recent study suggests that OTA-induced nephrotoxicity is linked to the Sigma-1 receptor (Sig-1R)-mediated mitochondrial pathway apoptosis in human proximal tubule epithelial-originated kidney-2 (HK-2) cells. However, the contribution of Sig-1R to OTA-induced nephrotoxicity involving other forms of regulated cell death, such as ferroptosis, remains unexplored. In this investigation, cell viability, malondialdehyde (MDA) levels, glutathione (GSH) levels, and protein expressions in HK-2 cells treated with OTA and/or Ferrostatin-1/blarcamesine hydrochloride/BD1063 dihydrochloride were assessed. The results indicate that a 24 h-treatment with 1 µM OTA significantly induces ferroptosis by inhibiting Sig-1R, subsequently promoting nuclear receptor coactivator 4 (NCOA4), long-chain fatty acid-CoA ligase 4 (ACSL4), arachidonate 5-lipoxygenase (ALOX5), autophagy protein 5 (ATG5), and ATG7, inhibiting ferritin heavy chain (FTH1), solute carrier family 7 member 11 (SLC7A11/xCT), glutathione peroxidase 4 (GPX4), peroxiredoxin 6 (PRDX6), and ferroptosis suppressor protein 1 (FSP1), reducing GSH levels, and increasing MDA levels (P < 0.05). In conclusion, OTA induces ferroptosis by inhibiting Sig-1R, subsequently promoting ferritinophagy, inhibiting GPX4/FSP1 antioxidant systems, reducing GSH levels, and ultimately increasing lipid peroxidation levels in vitro.


Asunto(s)
Ferroptosis , Ocratoxinas , Receptores sigma , Receptor Sigma-1 , Ocratoxinas/toxicidad , Ferroptosis/efectos de los fármacos , Receptores sigma/metabolismo , Humanos , Línea Celular , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Supervivencia Celular/efectos de los fármacos , Glutatión/metabolismo
7.
Molecules ; 29(11)2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38893570

RESUMEN

Sigma receptors (SRs), including SR1 and SR2 subtypes, have attracted increasing interest in recent years due to their involvement in a wide range of activities, including the modulation of opioid analgesia, neuroprotection, and potential anticancer activity. In this context, haloperidol (HAL), a commonly used antipsychotic drug, also possesses SR activity and cytotoxic effects. Herein, we describe the identification of novel SR ligands, obtained by a chemical hybridization approach. There wereendowed with pan-affinity for both SR subtypes and evaluated their potential anticancer activity against SH-SY5Y and HUH-7 cancer cell lines. Through a chemical hybridization approach, we identified novel compounds (4d, 4e, 4g, and 4j) with dual affinity for SR1 and SR2 receptors. These compounds were subjected to cytotoxicity testing using a resazurin assay. The results revealed potent cytotoxic effects against both cancer cell lines, with IC50 values comparable to HAL. Interestingly, the cytotoxic potency of the novel compounds resembled that of the SR1 antagonist HAL rather than the SR2 agonist siramesine (SRM), indicating the potential role of SR1 antagonism in their mechanism of action. The further exploration of their structure-activity relationships and their evaluation in additional cancer cell lines will elucidate their therapeutic potential and may pave the way for the development of novel anticancer agents that target SRs.


Asunto(s)
Antineoplásicos , Diseño de Fármacos , Haloperidol , Receptores sigma , Receptores sigma/metabolismo , Receptores sigma/antagonistas & inhibidores , Haloperidol/farmacología , Haloperidol/análogos & derivados , Humanos , Antineoplásicos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Relación Estructura-Actividad , Estructura Molecular , Supervivencia Celular/efectos de los fármacos , Ligandos , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales
8.
ACS Chem Neurosci ; 15(11): 2265-2282, 2024 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-38743607

RESUMEN

Prion diseases are invariably fatal neurodegenerative diseases of humans and other animals for which there are no effective treatment options. Previous work from our laboratory identified phenethylpiperidines as a novel class of anti-prion compounds. While working to identify the molecular target(s) of these molecules, we unexpectedly discovered ten novel antiprion compounds based on their known ability to bind to the sigma receptors, σ1R and σ2R, which are currently being tested as therapeutic or diagnostic targets for cancer and neuropsychiatric disorders. Surprisingly, however, knockout of the respective genes encoding σ1R and σ2R (Sigmar1 and Tmem97) in prion-infected N2a cells did not alter the antiprion activity of these compounds, demonstrating that these receptors are not the direct targets responsible for the antiprion effects of their ligands. Further investigation of the most potent molecules established that they are efficacious against multiple prion strains and protect against downstream prion-mediated synaptotoxicity. While the precise details of the mechanism of action of these molecules remain to be determined, the present work forms the basis for further investigation of these compounds in preclinical studies. Given the therapeutic utility of several of the tested compounds, including rimcazole and haloperidol for neuropsychiatric conditions, (+)-pentazocine for neuropathic pain, and the ongoing clinical trials of SA 4503 and ANAVEX2-73 for ischemic stroke and Alzheimer's disease, respectively, this work has immediate implications for the treatment of human prion disease.


Asunto(s)
Enfermedades por Prión , Receptores sigma , Receptores sigma/metabolismo , Receptores sigma/efectos de los fármacos , Animales , Ligandos , Enfermedades por Prión/tratamiento farmacológico , Enfermedades por Prión/metabolismo , Ratones , Humanos , Priones/efectos de los fármacos , Priones/metabolismo , Receptor Sigma-1 , Línea Celular Tumoral
9.
Cell Death Dis ; 15(5): 309, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38697978

RESUMEN

Sigma-2-ligands (S2L) are characterized by high binding affinities to their cognate sigma-2 receptor, overexpressed in rapidly proliferating tumor cells. As such, S2L were developed as imaging probes (ISO1) or as cancer therapeutics, alone (SV119 [C6], SW43 [C10]) and as delivery vehicles for cytotoxic drug cargoes (C6-Erastin, C10-SMAC). However, the exact mechanism of S2L-induced cytotoxicity remains to be fully elucidated. A series of high-affinity S2L were evaluated regarding their cytotoxicity profiles across cancer cell lines. While C6 and C10 displayed distinct cytotoxicities, C0 and ISO1 were essentially non-toxic. Confocal microscopy and lipidomics analysis in cellular and mouse models revealed that C10 induced increases in intralysosomal free cholesterol and in cholesterol esters, suggestive of unaltered intracellular cholesterol trafficking. Cytotoxicity was caused by cholesterol excess, a phenomenon that contrasts the effects of NPC1 inhibition. RNA-sequencing revealed gene clusters involved in cholesterol homeostasis and ER stress response exclusively by cytotoxic S2L. ER stress markers were confirmed by qPCR and their targeted modulation inhibited or enhanced cytotoxicity of C10 in a predicted manner. Moreover, C10 increased sterol regulatory element-binding protein 2 (SREBP2) and low-density lipoprotein receptor (LDLR), both found to be pro-survival factors activated by ER stress. Furthermore, inhibition of downstream processes of the adaptive response to S2L with simvastatin resulted in synergistic treatment outcomes in combination with C10. Of note, the S2L conjugates retained the ER stress response of the parental ligands, indicative of cholesterol homeostasis being involved in the overall cytotoxicity of the drug conjugates. Based on these findings, we conclude that S2L-mediated cell death is due to free cholesterol accumulation that leads to ER stress. Consequently, the cytotoxic profiles of S2L drug conjugates are proposed to be enhanced via concurrent ER stress inducers or simvastatin, strategies that could be instrumental on the path toward tumor eradication.


Asunto(s)
Colesterol , Estrés del Retículo Endoplásmico , Receptores sigma , Colesterol/metabolismo , Receptores sigma/metabolismo , Receptores sigma/genética , Humanos , Animales , Ratones , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ligandos , Línea Celular Tumoral , Muerte Celular/efectos de los fármacos , Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología
10.
Bioorg Med Chem ; 97: 117515, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-38043245

RESUMEN

Over-expression of sigma-2 receptor in cancer cells provides an opportunity to develop molecular probes for diagnosis, even for non-receptor specific malignancies like triple negative breast cancers. In this work, a novel sigma-2 receptor ligand [THQ-DTPA] has been synthesized and characterized using 6,7-dimethoxy-1,2,3,4-tetrahydroisoquinoline (THQ) and diethylenetriaminepentaacetic acid (DTPA). The ligand is further chelated with 99mTc for application as metal based radiotracer [99mTc-THQ-DTPA]. Radiolabelling with 99mTc was achieved in an excellent yield of 98.0 ± 0.5% using stannous chloride as a reducing agent. The radioligand was found to be stable in human serum up-to 24 h, bio-compatible with less than 4% hemolysis, and exhibited high binding with sigma receptors isolated from rat liver membrane (Kd of 16.32 ± 4.93 nM and Bmax of 0.5232 ± 0.06 pmol/mg). Bio-distribution studies in triple-negative breast tumor bearing nude mice showed high tumor uptake after 30 min of injection with tumor/muscle (T/M) ratio of 3.58 ± 0.09. At 240 min, the T/M ratio (2.84 ± 0.20) decreased by 35% when administered in sigma blocked tumor bearing mice (1.81 ± 0.16) suggesting the selectivity of the ligand. Tumor imaging in gamma camera indicated a contrast of 3.56 at 30 min p.i. The above findings indicate that the ligand 99mTc-THQ-DTPA binds to sigma-2 receptors with high affinity and has potential for triple-negative breast tumor imaging.


Asunto(s)
Receptores sigma , Neoplasias de la Mama Triple Negativas , Ratas , Ratones , Humanos , Animales , Ligandos , Neoplasias de la Mama Triple Negativas/diagnóstico por imagen , Ratones Desnudos , Ácido Pentético , Receptores sigma/metabolismo , Radiofármacos , Línea Celular Tumoral , Tomografía Computarizada de Emisión de Fotón Único
11.
Int J Mol Sci ; 24(19)2023 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-37834119

RESUMEN

Sigma receptors are non-opiate/non-phencyclidine receptors that bind progesterone and/or heme and also several unrelated xenobiotics/chemicals. They reside in the plasma membrane and in the membranes of the endoplasmic reticulum, mitochondria, and nucleus. Until recently, the biology/pharmacology of these proteins focused primarily on their role in neuronal functions in the brain/retina. However, there have been recent developments in the field with the discovery of unexpected roles for these proteins in iron/heme homeostasis. Sigma receptor 1 (S1R) regulates the oxidative stress-related transcription factor NRF2 and protects against ferroptosis, an iron-induced cell death process. Sigma receptor 2 (S2R), which is structurally unrelated to S1R, complexes with progesterone receptor membrane components PGRMC1 and PGRMC2. S2R, PGRMC1, and PGRMC2, either independently or as protein-protein complexes, elicit a multitude of effects with a profound influence on iron/heme homeostasis. This includes the regulation of the secretion of the iron-regulatory hormone hepcidin, the modulation of the activity of mitochondrial ferrochelatase, which catalyzes iron incorporation into protoporphyrin IX to form heme, chaperoning heme to specific hemoproteins thereby influencing their biological activity and stability, and protection against ferroptosis. Consequently, S1R, S2R, PGRMC1, and PGRMC2 potentiate disease progression in hemochromatosis and cancer. These new discoveries usher this intriguing group of non-traditional progesterone receptors into an unchartered territory in biology and medicine.


Asunto(s)
Ferroptosis , Receptores sigma , Receptores sigma/metabolismo , Hemo/metabolismo , Receptores de Progesterona/metabolismo , Hierro , Homeostasis
12.
Pharmacol Rep ; 75(6): 1588-1596, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37796435

RESUMEN

BACKGROUND: Multifunctional thiosemicarbazones (TSCs) able to bind sigma receptors and chelate metals are considered as a promising avenue for the treatment of pancreatic cancer due to the encouraging results obtained on in vitro and in vivo models. Here, we assessed the biochemical mechanism of these TSCs also on lung (A549) and breast (MCF7) cancer cells. METHODS: The density of sigma-2 receptors in normal (BEAS-2B and MCF10A) and in lung and breast (A549 and MCF7) cancer cells was evaluated by flow cytometry. In these cells, cytotoxicity (MTT assay) and activation of ER- and mitochondria-dependent cell death pathways (by spectrofluorimetric assays to measure Caspases 3/7/9; qRT-PCR detection of GRP78, ATF6, IRE1, PERK; MitoSOX, DCFDA-AM and JC-1 staining), induced by the TSCs FA4, MLP44, PS3 and ACThio1, were evaluated. RESULTS: FA4 and PS3 exerted more potent cytotoxicity than MLP44 and ACThio1 in all cancer cell lines, where the density of sigma-2 receptors was higher than in normal cells. Remarkably, FA4 promoted ER- and mitochondria-dependent cell death pathways in both cell models, whereas the other TSCs had variable, cell-dependent effects on the activation of the two proapoptotic pathways. CONCLUSIONS: Our data suggest that FA4 is a promising compound that deserves to be further studied for lung and breast cancer treatment. However, the other multifunctional TSCs also hold promise for the development of therapies towards a personalized medicine approach. Indeed, the presence of the sigma-2 receptor-targeting moiety would lead to a more specific tumor delivery embracing the characteristics of individual tumor types.


Asunto(s)
Antineoplásicos , Carcinoma , Neoplasias Pulmonares , Receptores sigma , Tiosemicarbazonas , Humanos , Receptores sigma/metabolismo , Apoptosis , Tiosemicarbazonas/farmacología , Antineoplásicos/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Pulmón/metabolismo , Línea Celular Tumoral
13.
Nucl Med Biol ; 122-123: 108369, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37516066

RESUMEN

INTRODUCTION: As sigma receptors are abundantly expressed on different types of cancer cells, several radiolabeled sigma receptor ligands have been developed for cancer imaging and therapy. Previously, we synthesized and evaluated radioiodinated aza-vesamicol derivatives, [125I]pIC3NV, [125I]mIC2N5V, and [125I]mIC3N5V. They accumulated in tumors, and [125I]mIC2N5V and [125I]mIC3N5V showed higher tumor to non-target tissue ratios than [125I]pIC3NV. Therefore, we synthesized and evaluated the corresponding 211At-labeled compounds, [211At]mAtC2N5V and [211At]mAtC3N5V, for targeted alpha therapy (TAT). METHODS: [211At]mAtC2N5V and [211At]mAtC3N5V were prepared by the standard method of electrophilic astatodestannylation of the corresponding trimethylstannyl precursors. Cellular uptake experiments, and biodistribution experiments and therapeutic experiments in tumor-bearing mice were performed. RESULTS: The radiochemical yields of [211At]mAtC2N5V and [211At]mAtC3N5V were 45.5 ± 14.4% and 56.9 ± 13.8%, respectively. After HPLC purification, their radiochemical purities were over 95%. [211At]mAtC2N5V and [211At]mAtC3N5V showed high uptake in DU-145 cells. They demonstrated high accumulation in tumors (6.9 ± 1.4%injected dose/g and 5.1 ± 1.4%injected dose/g at 1 h, respectively) and similar biodistribution tendencies compared with the corresponding 125I-labeled compounds. A single injection of [211At]mAtC2N5V (0.48 MBq) or [211At]mAtC3N5V (0.48 MBq) significantly inhibited tumor growth. CONCLUSION: These results indicated that [211At]mAtC2N5V and [211At]mAtC3N5V could be potential candidates for TAT.


Asunto(s)
Neoplasias , Receptores sigma , Ratones , Animales , Receptores sigma/metabolismo , Distribución Tisular , Ligandos , Radiofármacos/uso terapéutico , Línea Celular Tumoral
14.
Molecules ; 28(8)2023 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-37110664

RESUMEN

Neurodegeneration is a slow and progressive loss of neuronal cells or their function in specific regions of the brain or in the peripheral system. Among several causes responsible for the most common neurodegenerative diseases (NDDs), cholinergic/dopaminergic pathways, but also some endogenous receptors, are often involved. In this context, sigma 1 receptor (S1R) modulators can be used as neuroprotective and antiamnesic agents. Herein, we describe the identification of novel S1R ligands endowed with antioxidant properties, potentially useful as neuroprotective agents. We also computationally assessed how the most promising compounds might interact with the S1R protein's binding sites. The in silico predicted ADME properties suggested that they could be able to cross the brain-blood-barrier (BBB), and to reach the targets. Finally, the observation that at least two novel ifenprodil analogues (5d and 5i) induce an increase of the mRNA levels of the antioxidant NRF2 and SOD1 genes in SH-SY5Y cells suggests that they might be effective agents for protecting neurons against oxidative damage.


Asunto(s)
Neuroblastoma , Fármacos Neuroprotectores , Receptores sigma , Humanos , Antioxidantes/farmacología , Antioxidantes/química , Ligandos , Neuroprotección , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/química , Receptores sigma/metabolismo
15.
Int J Mol Sci ; 24(7)2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-37047353

RESUMEN

Sigma-2 receptor (S2R) is a S2R ligand-binding site historically associated with reportedly 21.5 kDa proteins that have been linked to several diseases, such as cancer, Alzheimer's disease, and schizophrenia. The S2R is highly expressed in various tumors, where it correlates with the proliferative status of the malignant cells. Recently, S2R was reported to be the transmembrane protein TMEM97. Prior to that, we had been investigating the translocator protein (TSPO) as a potential 21.5 kDa S2R candidate protein with reported heme and sterol associations. Here, we investigate the contributions of TMEM97 and TSPO to S2R activity in MCF7 breast adenocarcinoma and MIA PaCa-2 (MP) pancreatic carcinoma cells. Additionally, the role of the reported S2R-interacting partner PGRMC1 was also elucidated. Proximity ligation assays and co-immunoprecipitation show a functional association between S2R and TSPO. Moreover, a close physical colocalization of TMEM97 and TSPO was found in MP cells. In MCF7 cells, co-immunoprecipitation only occurred with TMEM97 but not with PGRMC1, which was further confirmed by confocal microscopy experiments. Treatment with the TMEM97 ligand 20-(S)-hydroxycholesterol reduced co-immunoprecipitation of both TMEM97 and PGRMC1 in immune pellets of immunoprecipitated TSPO in MP cells. To the best of our knowledge, this is the first suggestion of a (functional) interaction between TSPO and TMEM97 that can be affected by S2R ligands.


Asunto(s)
Receptores sigma , Humanos , Ligandos , Unión Proteica , Receptores sigma/metabolismo , Sitios de Unión , Receptores de GABA/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Progesterona/metabolismo
16.
J Med Chem ; 66(3): 1990-2019, 2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36692906

RESUMEN

Sigma 2 receptor (σ2R) is overexpressed in select cancers and is regarded as a biomarker for tumor proliferation. σ2R ligands are emerging as promising theranostics for cancer and neurodegenerative diseases. Herein, we describe the design and synthesis of a series of novel quinolyl pyrazinamides as selective and potent σ2R ligands that show sub-micromolar potency in pancreatic cancer cell lines. Compounds 14 (JR1-157) and 17 (JR2-298) bind σ2R with Ki of 47 and 10 nM, respectively. Importantly, compound 14 has an oral bioavailability of 60% and shows significant in vivo efficacy without obvious toxicity in a syngeneic model of pancreatic cancer. The cytotoxicity of the quinolyl pyrazinamides significantly enhanced in the presence of copper and diminished in the presence of the copper-chelator tetrathiomolybdate. In conclusion, compound 14 is water-soluble, metabolically stable, orally active, and increases the expression of the autophagy marker LC3B and warrants further development for the treatment of pancreatic cancer.


Asunto(s)
Neoplasias Pancreáticas , Receptores sigma , Humanos , Ligandos , Pirazinamida , Cobre , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Receptores sigma/metabolismo , Neoplasias Pancreáticas
17.
Bioorg Med Chem ; 73: 117032, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-36202063

RESUMEN

The overexpression of σ receptors (σRs) in various types of tumors has prompted a deep investigation of their role in cancer pathophysiology. Consequently, σR ligands have been widely studied in vitro and in vivo for their antiproliferative effects as a novel potential class of chemotherapeutic agents, both alone and in combination with other anticancer drugs. A growing body of evidence highlights that σR ligands can inhibit cancer cells' survival, migration, and proliferation, thanks to the modulation of a wide panel of tumorigenic pathways. In addition to their antitumor activity, σR ligands are gaining momentum as radiotracers for PET and SPECT imaging applications. The purpose of this review is to report on recent advances in the development of σR ligands. In particular, herein, we describe the structure-activity relationships of structurally diverse mixed σ1R/σ2R ligands that showed promising antitumor profiles towards a variety of cancer cell lines.


Asunto(s)
Antineoplásicos , Neoplasias , Receptores sigma , Antineoplásicos/farmacología , Humanos , Ligandos , Receptores sigma/metabolismo , Relación Estructura-Actividad
18.
Eur J Med Chem ; 243: 114696, 2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36088757

RESUMEN

Initially associated with cancer diagnosis and therapy, the sigma 2 receptor (σ2R) has recently been implicated in several disorders of the central nervous system (CNS). It remained a poorly characterized target until we identified it as the transmembrane protein 97 (TMEM97). As part of a program to identify novel compounds that bind with high affinity and selectivity to σ2R/TMEM97 relative to the sigma 1 receptor (σ1R) and other CNS proteins, we employed a scaffold simplification strategy to design novel sets of piperazine-substituted aminotetralins based on analogous norbenzomorphans that we previously developed. JVW-1601 was identified as a reference aminotetralin analog that had high affinity (Ki = 5.5 nM) and selectivity (36-fold) for σ2R/TMEM97 versus σ1R. An expanded investigation of structure-activity relationships (SAR) in several structural regions of this compound was conducted, and among the ligands thus prepared, many had Ki values < 20 nM for σ2R/TMEM97 and selectivities of >20-fold versus σ1R. Structural features that enhance σ2R/TMEM97 affinity and selectivity were identified, leading to an optimized compound having a high σ2R/TMEM97 affinity (Ki of 4.5 nM) and 366-fold selectivity relative to σ1R. Significantly, during the course of this work we discovered JVW-1625, which enabled the isolation and identification of σ2R as TMEM97 and resolved a question that had eluded researchers for decades. Computational docking studies for selected aminotetralins suggest they adopt similar poses upon binding to σ2R/TMEM97, engaging in highly conserved salt bridges with Asp29 and cation-π interactions with Tyr150. Collectively, these studies show that aminotetralins are useful tool compounds for studying the mechanism and function of σ2R/TMEM97.


Asunto(s)
Receptores sigma , Receptores sigma/metabolismo , Ligandos , Relación Estructura-Actividad
19.
Mol Med ; 28(1): 87, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35922746

RESUMEN

BACKGROUND: Angiogenesis plays a critical role on post-infarction heart failure (PIHF), the presence of which facilitates additional blood supply to maintain the survival of residual cardiomyocytes. The sigma-1 receptor (S1R) has been substantiated to stimulate angiogenesis, with the effect on a model of PIHF remaining unknown. AIMS: This study aims to investigate the effects of S1R on PIHF and the underlying mechanisms involved. METHODS: Rats were implemented left anterior descending artery ligation followed by rearing for 6 weeks to induce a phenotype of heart failure. Daily intraperitoneal injection of S1R agonist or antagonist for 5 weeks was applied from 2nd week after surgery. The effects exerted by S1R were detected by echocardiography, hemodynamic testing, western blot, Sirius red dyeing, ELISA, immunohistochemistry and fluorescence. We also cultured HUVECs to verify the mechanisms in vitro. RESULTS: Stimulation of S1R significantly ameliorated the cardiac function resulted from PIHF, in addition to the observation of reduced fibrosis in the peri-infarct region and the apoptosis of residual cardiomyocytes, which were associated with augmentation of microvascular density in peri-infarct region through activation of the JAK2/STAT3 pathway. We also indicated that suppression of JAK2/STAT3 pathway by specific inhibitor in vitro reversed the pro-angiogenic effects of S1R on HUVECs, which further confirmed that angiogenesis, responsible for PIHF amelioration, by S1R stimulation was in a JAK2/STAT3 pathway-dependent manner. CONCLUSION: S1R stimulation improved PIHF-induced cardiac dysfunction and ventricular remodeling through promoting angiogenesis by activating the JAK2/STAT3 pathway.


Asunto(s)
Insuficiencia Cardíaca , Receptores sigma , Animales , Infarto , Ratas , Receptores sigma/agonistas , Receptores sigma/metabolismo , Roedores/metabolismo , Factor de Transcripción STAT3/metabolismo , Receptor Sigma-1
20.
Biomolecules ; 12(6)2022 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-35740887

RESUMEN

Sigma-1 receptor (S1R) is an important endoplasmic reticulum chaperone with various functions in health and disease. The purpose of the current work was to elucidate the involvement of S1R in cancer energy metabolism under its basal, activated, and inactivated states. For this, two cancer cell lines that differentially express S1R were treated with S1R agonist, (+)-SKF10047, and antagonist, BD1047. The effects of the agonist and antagonist on cancer energy metabolism were studied using single-cell fluorescence microscopy analysis of real-time ion and metabolite fluxes. Our experiments revealed that S1R activation by agonist increases mitochondrial bioenergetics of cancer cells while decreasing their reliance on aerobic glycolysis. S1R antagonist did not have a major impact on mitochondrial bioenergetics of tested cell lines but increased aerobic glycolysis of S1R expressing cancer cell line. Our findings suggest that S1R plays an important role in cancer energy metabolism and that S1R ligands can serve as tools to modulate it.


Asunto(s)
Neoplasias , Receptores sigma , Retículo Endoplásmico/metabolismo , Metabolismo Energético , Ligandos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Receptores sigma/agonistas , Receptores sigma/metabolismo , Receptor Sigma-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA