Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 211
Filtrar
1.
Semin Cell Dev Biol ; 146: 70-79, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36604249

RESUMEN

Viruses rely on host cells for energy and synthesis machinery required for genome replication and particle assembly. Due to the dependence of viruses on host cells, viruses have evolved multiple mechanisms by which they can induce metabolic changes in the host cell to suit their specific requirements. The host immune response also involves metabolic changes to be able to react to viral insult. Polyamines are small ubiquitously expressed polycations, and their metabolism is critical for viral replication and an adequate host immune response. This is due to the variety of functions that polyamines have, ranging from condensing DNA to enhancing the translation of polyproline-containing proteins through the hypusination of eIF5A. Here, we review the diverse mechanisms by which viruses exploit polyamines, as well as the mechanisms by which immune cells utilize polyamines for their functions. Furthermore, we highlight potential avenues for further study of the host-virus interface.


Asunto(s)
Interacciones Microbiota-Huesped , Poliaminas , Virosis , Replicación Viral , Virus , Humanos , Inmunidad Adaptativa , Antineoplásicos/farmacología , Antivirales/farmacología , Eflornitina/farmacología , Interacciones Microbiota-Huesped/inmunología , Poliaminas/antagonistas & inhibidores , Poliaminas/metabolismo , Virosis/metabolismo , Virosis/virología , Virus/metabolismo , Procesamiento Proteico-Postraduccional , Lisina , Factor 5A Eucariótico de Iniciación de Traducción
2.
Eur J Pharmacol ; 910: 174456, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34464603

RESUMEN

Spermine, spermidine and putrescine polyamines are naturally occurring ubiquitous positively charged amines and are essential metabolites for biological functions in our life. These compounds play a crucial role in many cell processes, including cellular proliferation, growth, and differentiation. Intracellular levels of polyamines depend on their biosynthesis, transport and degradation. Polyamine levels are high in cancer cells, which leads to the promotion of tumor growth, invasion and metastasis. Targeting polyamine metabolism as an anticancer strategy is considerably rational. Due to compensatory mechanisms, a single strategy does not achieve satisfactory clinical effects when using a single agent. Combination regimens are more clinically promising for cancer chemoprevention because they work synergistically with causing little or no adverse effects due to each individual agent being used at lower doses. Moreover, bioactive substances have advantages over single chemical agents because they can affect multiple targets. In this review, we discuss anticancer strategies targeting polyamine metabolism and describe how combination treatments and effective natural active ingredients are promising therapies. The existing research suggests that polyamine metabolic enzymes are important therapeutic targets and that combination therapies can be more effective than monotherapies based on polyamine depletion.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Homeostasis/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Poliaminas/antagonistas & inhibidores , Poliaminas/metabolismo , Animales , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Humanos , Poliaminas/química
3.
J Immunother ; 44(8): 283-291, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34133404

RESUMEN

Polyamines are known to play a significant role in cancer progression and treatment using difluoromethylornithine (DFMO), an inhibitor of polyamine biosynthesis, has shown some clinical promise. It is interesting to note that, while DFMO is directly cytostatic in vitro, recent work has suggested that it achieves its antitumor efficacy in vivo by enhancing adaptive antitumor immune responses. On the basis of these data, we hypothesized that DFMO might act as an immune sensitizer to increase tumor responsiveness to checkpoint blockade. To test this hypothesis, we treated tumors with DFMO, in either the presence or absence of additional PD-1 blockade, and subsequently analyzed their immunological and therapeutic responses. Our data demonstrates that treatment with DFMO significantly enhances both the viability and activation status of intratumoral CD8+ T cells, most likely through an indirect mechanism. When combined with PD-1 blockade, this increased viability resulted in unique proinflammatory cytokine profiles and transcriptomes within the tumor microenvironment and improved therapeutic outcomes. Taken together, these data suggest that DFMO might represent a potential immunomodulatory agent that can enhance current PD-1-based checkpoint therapies.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Eflornitina/uso terapéutico , Factores Inmunológicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Poliaminas/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Citocinas/inmunología , Sinergismo Farmacológico , Quimioterapia Combinada , Eflornitina/farmacología , Femenino , Humanos , Factores Inmunológicos/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Ratones Endogámicos C57BL , Neoplasias/inmunología , Poliaminas/metabolismo
4.
Amino Acids ; 52(8): 1169-1180, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32816168

RESUMEN

Polyamines (PAs), such as spermidine (SPD) and spermine (SPM), are essential to promote cell growth, survival, proliferation, and longevity. In the adult central nervous system (CNS), SPD and SPM are accumulated predominantly in healthy adult glial cells where PA synthesis is not present. To date, the accumulation and biosynthesis of PAs in developing astrocytes are not well understood. The purpose of the present study was to determine the contribution of uptake and/or synthesis of PAs using proliferation of neonatal astrocytes as an endpoint. We inhibited synthesis of PAs using α-difluoromethylornithine (DFMO; an inhibitor of the PA biosynthetic enzyme ornithine decarboxylase (ODC)) and inhibited uptake of PAs using trimer44NMe (PTI; a novel polyamine transport inhibitor). DFMO, but not PTI alone, blocked proliferation, suggesting that PA biosynthesis was present. Furthermore, exogenous administration of SPD rescued cell proliferation when PA synthesis was blocked by DFMO. When both synthesis and uptake of PAs were inhibited (DFMO + PTI), exogenous SPD no longer supported proliferation. These data indicate that neonatal astrocytes synthesize sufficient quantities of PAs de novo to support cell proliferation, but are also able to import exogenous PAs. This suggests that the PA uptake mechanism is present in both neonates as well as in adults and can support cell proliferation in neonatal astrocytes when ODC is blocked.


Asunto(s)
Astrocitos/metabolismo , Poliaminas/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Eflornitina , Poliaminas/antagonistas & inhibidores , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley , Espermidina/metabolismo , Espermina/metabolismo
5.
Mol Cancer Ther ; 19(10): 2012-2022, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32747421

RESUMEN

Despite unprecedented advances in the treatment of cancer through the use of immune checkpoint blockade (ICB), responses are not universal and alternative strategies are needed to enhance responses to ICB. We have shown previously that a novel polyamine blocking therapy (PBT), consisting of cotreatment with α-difluoromethylornithine (DFMO) to block polyamine biosynthesis and a Trimer polyamine transport inhibitor, decreases myeloid-derived suppressor cells (MDSC) and M2-like tumor-associated macrophages (TAM). Both MDSCs and TAMs promote tumor progression, inhibit antitumor immunity, and limit the efficacy of ICB. In this study, we investigated the use of PBT to heighten therapeutic responses to PD-1 blockade in mice bearing 4T1 mammary carcinoma and B16F10 melanoma tumors. Whereas PBT inhibited primary tumor growth in both tumor models, 4T1 lung metastases were also dramatically decreased in mice treated with PBT. Reductions in MDSC and TAM subpopulations in 4T1 tumors from PBT-treated mice were accompanied by reduced cytoprotective autophagy only in tumor-infiltrating MDSC and macrophage subpopulations but not in the lung or spleen. PBT treatment blunted M2-like alternative activation of bone marrow-derived macrophages and reduced STAT3 activation in MDSC cultures while increasing the differentiation of CD80+, CD11c+ macrophages. PBT significantly enhanced the antitumor efficacy of PD-1 blockade in both 4T1 and B16F10 tumors resistant to anti-PD-1 monotherapy, increasing tumor-specific cytotoxic T cells and survival of tumor-bearing animals beyond that with PBT or PD-1 blockade alone. Our results suggest that cotreatment with DFMO and the Trimer polyamine transport inhibitor may improve the therapeutic efficacy of immunotherapies in patients with cancer with resistant tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Células Mieloides/metabolismo , Poliaminas/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Femenino , Humanos , Ratones
6.
J Plant Physiol ; 246-247: 153092, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32065919

RESUMEN

The mechanisms regulating, and modulating potato wound-healing processes are of great importance in reducing tuber infections, reducing shrinkage and maintaining quality and nutritional value for growers and consumers. Wound-induced changes in tuber polyamine metabolism have been linked to the modulation of wound healing (WH) and in possibly providing the crucial amount of H2O2 required for suberization processes. In this investigation we determined the effect of inhibition of specific steps within the pathway of polyamine metabolism on polyamine content and the initial accumulation of suberin polyphenolics (SPP) during WH. The accumulation of SPP represents a critical part of the beginning or inchoate phase of tuber WH during closing-layer formation because it serves as a barrier to bacterial infection and is a requisite for the accumulation of suberin polyaliphatics which provide the barrier to fungal infection. Results showed that the inhibitor treatments that caused changes in polyamine content generally did not influence wound-induced accumulation of SPP. Such lack of correlation was found for inhibitors involved in metabolism and oxidation of putrescine (arginine decarboxylase, ornithine decarboxylase, and diamine oxidase). However, accumulation of SPP was dramatically reduced by treatment with guazatine, a potent inhibitor of polyamine oxidase (PAO), and methylglyoxal-bis(guanylhydrazone), a putative inhibitor of S-adenosylmethione decarboxylase which may also cross-react to inhibit PAO. The mode of action of these inhibitors is presumed to be blockage of essential H2O2 production within the WH cell wall. These results are of great importance in understanding the mechanisms modulating WH and ultimately controlling related infections and associated postharvest losses.


Asunto(s)
Diaminas/antagonistas & inhibidores , Lípidos/biosíntesis , Proteínas de Plantas/metabolismo , Tubérculos de la Planta/metabolismo , Poliaminas/antagonistas & inhibidores , Solanum tuberosum/metabolismo , Carboxiliasas/metabolismo , Diaminas/metabolismo , Guanidinas/metabolismo , Mitoguazona/metabolismo , Oxidación-Reducción , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Poliaminas/metabolismo , Putrescina/metabolismo , Solanum tuberosum/enzimología , Poliamino Oxidasa
7.
Microbiol Mol Biol Rev ; 81(4)2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28904024

RESUMEN

Polyamines are small, abundant, aliphatic molecules present in all mammalian cells. Within the context of the cell, they play a myriad of roles, from modulating nucleic acid conformation to promoting cellular proliferation and signaling. In addition, polyamines have emerged as important molecules in virus-host interactions. Many viruses have been shown to require polyamines for one or more aspects of their replication cycle, including DNA and RNA polymerization, nucleic acid packaging, and protein synthesis. Understanding the role of polyamines has become easier with the application of small-molecule inhibitors of polyamine synthesis and the use of interferon-induced regulators of polyamines. Here we review the diverse mechanisms in which viruses require polyamines and investigate blocking polyamine synthesis as a potential broad-spectrum antiviral approach.


Asunto(s)
Poliaminas/metabolismo , Virosis/metabolismo , Replicación Viral , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , ADN Viral/metabolismo , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Humanos , Ratones , Poliaminas/antagonistas & inhibidores , ARN Viral/metabolismo , Proteínas Virales/metabolismo , Virosis/tratamiento farmacológico , Replicación Viral/efectos de los fármacos
8.
Curr Pharm Des ; 23(23): 3359-3366, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28671057

RESUMEN

Polyamines are essential for many biological processes in all organisms. Here we show a current landscape of studies and strategies implemented for the study of polyamine metabolism, as well as molecular aspects that implicate the role of key enzymes, transport proteins, inhibitors, and the study of novel molecules as potential therapeutic targets. This review focused on the synthesis, interconversion and function of these molecules in Trichomonas vaginalis, a common sexually transmitted parasite of humans.


Asunto(s)
Antiparasitarios/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Poliaminas/metabolismo , Trichomonas vaginalis/efectos de los fármacos , Trichomonas vaginalis/metabolismo , Secuencia de Aminoácidos , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Femenino , Humanos , Poliaminas/antagonistas & inhibidores , Espermidina/metabolismo , Espermina/metabolismo , Vaginitis por Trichomonas/tratamiento farmacológico , Vaginitis por Trichomonas/metabolismo , Trichomonas vaginalis/genética
9.
Curr Pharm Des ; 23(23): 3325-3341, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28571553

RESUMEN

There is an urgent need for the identification and validation of new therapeutic targets in protozoan parasites because currently available drugs are limited in number and usefulness, and no vaccines are available. The discovery that alpha-difluoromethylornithine, an inhibitor of polyamine biosynthesis, is an efficacious treatment for African Sleeping Sickness caused by the protozoan parasite Trypanosoma brucei, has validated the polyamine pathway as a target in protozoan parasites. Polyamines are ubiquitous organic cations that play critical roles in key cellular processes such as growth, differentiation, and macromolecular biosynthesis. In recent years, remarkable progress has been made in the characterization of the polyamine pathway in a variety of protozoan parasites and this review will highlight surprising and unique features that could lead to new therapeutic strategies.


Asunto(s)
Antiprotozoarios/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Parásitos/efectos de los fármacos , Poliaminas/antagonistas & inhibidores , Proteínas Protozoarias/antagonistas & inhibidores , Animales , Humanos , Parásitos/metabolismo , Poliaminas/metabolismo , Proteínas Protozoarias/metabolismo , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/metabolismo , Tripanosomiasis Africana/tratamiento farmacológico , Tripanosomiasis Africana/metabolismo
10.
J Neurovirol ; 23(4): 568-576, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28462488

RESUMEN

Peripheral neuropathy (PN) is a major comorbidity of HIV infection that is caused in part by chronic immune activation. HIV-PN is associated with infiltration of monocytes/macrophages to the dorsal root ganglia (DRG) causing neuronal loss and formation of Nageotte nodules. Here, we used an oral form of methylglyoxal-bis-guanylhydrazone (MGBG), a polyamine biosynthesis inhibitor, to specifically reduce activation of myeloid cells. MGBG is selectively taken up by monocyte/macrophages in vitro and inhibits HIV p24 expression and DNA viral integration in macrophages. Here, MGBG was administered to nine SIV-infected, CD8-depleted rhesus macaques at 21 days post-infection (dpi). An additional nine SIV-infected, CD8-depleted rhesus macaques were used as untreated controls. Cell traffic to tissues was measured by in vivo BrdU pulse labeling. MGBG treatment significantly diminished DRG histopathology and reduced the number of CD68+ and CD163+ macrophages in DRG tissue. The number of recently trafficked BrdU+ cells in the DRG was significantly reduced with MGBG treatment. Despite diminished DRG pathology, intraepidermal nerve fiber density (IENFD) did not recover after treatment with MGBG. These data suggest that MGBG alleviated DRG pathology and inflammation.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Ganglios Espinales/efectos de los fármacos , Mitoguazona/farmacología , Monocitos/efectos de los fármacos , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Administración Oral , Animales , Linfocitos T CD8-positivos/virología , Movimiento Celular/efectos de los fármacos , ADN Viral/genética , Ganglios Espinales/inmunología , Ganglios Espinales/patología , Ganglios Espinales/virología , Proteína p24 del Núcleo del VIH/genética , Depleción Linfocítica , Macaca mulatta , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Macrófagos/virología , Masculino , Monocitos/inmunología , Monocitos/patología , Monocitos/virología , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/inmunología , Fibras Nerviosas/patología , Fibras Nerviosas/virología , Enfermedades del Sistema Nervioso Periférico/inmunología , Enfermedades del Sistema Nervioso Periférico/patología , Enfermedades del Sistema Nervioso Periférico/virología , Poliaminas/antagonistas & inhibidores , Poliaminas/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/crecimiento & desarrollo
11.
PLoS One ; 12(4): e0175917, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28423064

RESUMEN

Synthesizing polycationic polymers directly from existing drugs overcomes the drug-loading limitations often associated with pharmacologically inert nanocarriers. We recently described nanocarriers formed from a first-generation polyamine analogue, bis(ethyl)norspermine (BENSpm), that could simultaneously target polyamine metabolism while delivering therapeutic nucleic acids. In the current study, we describe the synthesis and evaluation of self-immolative nanocarriers derived from the second-generation polyamine analogue PG-11047. Polyamines are absolutely essential for proliferation and their metabolism is frequently dysregulated in cancer. Through its effects on polyamine metabolism, PG-11047 effectively inhibits tumor growth in cancer cell lines of multiple origins as well as in human tumor mouse xenografts. Promising clinical trials have been completed verifying the safety and tolerance of this rotationally restricted polyamine analogue. We therefore used PG-11047 as the basis for Nano11047, a biodegradable, prodrug nanocarrier capable of targeting polyamine metabolism. Following exposure of lung cancer cell lines to Nano11047, uptake and intracellular degradation into the parent compound PG-11047 was observed. The release of PG-11047 highly induced the polyamine catabolic enzyme activities of spermidine/spermine N1-acetyltransferase (SSAT) and spermine oxidase (SMOX). By contrast, the activity of ornithine decarboxylase (ODC), a rate-limiting enzyme in polyamine biosynthesis and a putative oncogene, was decreased. Consequently, intracellular levels of the natural polyamines were depleted concurrent with tumor cell growth inhibition. This availability of Nano11047 as a novel drug form and potential nucleic acid delivery vector will potentially benefit and encourage future clinical studies.


Asunto(s)
Antineoplásicos/farmacología , Portadores de Fármacos , Regulación Neoplásica de la Expresión Génica , Ornitina Descarboxilasa/genética , Profármacos/farmacología , Espermina/análogos & derivados , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Antineoplásicos/síntesis química , Biotransformación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/metabolismo , Nanoestructuras/química , Ornitina Descarboxilasa/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Poliaminas/antagonistas & inhibidores , Poliaminas/metabolismo , Profármacos/síntesis química , Profármacos/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Espermina/síntesis química , Espermina/metabolismo , Espermina/farmacología , Carga Tumoral/efectos de los fármacos , Poliamino Oxidasa
12.
BMC Gastroenterol ; 16(1): 87, 2016 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-27480131

RESUMEN

BACKGROUND: Molecular studies suggest inhibition of colorectal mucosal polyamines (PAs) may be a promising approach to prevent colorectal cancer (CRC). Inhibition of ornithine decarboxylase (ODC) using low-dose eflornithine (DFMO, CPP-1X), combined with maximal PA export using low-dose sulindac, results in greatly reduced levels of normal mucosal PAs. In a clinical trial, this combination (compared with placebo) reduced the 3-year incidence of subsequent high-risk adenomas by >90 %. Familial Adenomatous Polyposis (FAP) is characterized by marked up-regulation of ODC in normal intestinal epithelial and adenoma tissue, and therefore PA reduction might be a potential strategy to control progression of FAP-related intestinal polyposis. CPP FAP-310, a randomized, double-blind, Phase III trial was designed to examine the safety and efficacy of sulindac and DFMO (alone or in combination) for preventing a clinically relevant FAP-related progression event in individuals with FAP. METHODS: Eligible adults with FAP will be randomized to: CPP-1X 750 mg and sulindac 150 mg, CPP-1X placebo and sulindac 150 mg, or CPP-1X 750 mg and sulindac placebo once daily for 24 months. Patients will be stratified based on time-to-event prognosis into one of the three treatment arms: best (ie, longest time to first FAP-related event [rectal/pouch polyposis]), intermediate (duodenal polyposis) and worst (pre-colectomy). Stage-specific, "delayed time to" FAP-related events are the primary endpoints. Change in polyp burden (upper and/or lower intestine) is a key secondary endpoint. DISCUSSION: The trial is ongoing. As of February 1, 2016, 214 individuals have been screened; 138 eligible subjects have been randomized to three treatment groups at 15 North American sites and 6 European sites. By disease strata, 26, 80 and 32 patients are included for assessment of polyp burden in the rectum/pouch, duodenal polyposis and pre-colectomy groups, respectively. Median age is 40 years; 59 % are men. The most common reasons for screening failure include minimal polyp burden (n = 22), withdrawal of consent (n = 9) and extensive polyposis requiring immediate surgical intervention (n = 9). Enrollment is ongoing. TRIAL REGISTRATION: This trial is registered at ClinicalTrials.gov ( NCT01483144 ; November 21, 2011) and the EU Clinical Trials Register( EudraCT 2012-000427-41 ; May 15, 2014).


Asunto(s)
Poliposis Adenomatosa del Colon/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Eflornitina/uso terapéutico , Sulindac/uso terapéutico , Poliposis Adenomatosa del Colon/metabolismo , Adulto , Antineoplásicos/efectos adversos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Progresión de la Enfermedad , Método Doble Ciego , Neoplasias Duodenales/tratamiento farmacológico , Neoplasias Duodenales/metabolismo , Eflornitina/efectos adversos , Femenino , Humanos , Mucosa Intestinal/metabolismo , Masculino , Poliaminas/antagonistas & inhibidores , Poliaminas/metabolismo , Sulindac/efectos adversos
13.
Clin Cancer Res ; 22(17): 4391-404, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-27012811

RESUMEN

PURPOSE: Deregulated MYC drives oncogenesis in many tissues yet direct pharmacologic inhibition has proven difficult. MYC coordinately regulates polyamine homeostasis as these essential cations support MYC functions, and drugs that antagonize polyamine sufficiency have synthetic-lethal interactions with MYC Neuroblastoma is a lethal tumor in which the MYC homologue MYCN, and ODC1, the rate-limiting enzyme in polyamine synthesis, are frequently deregulated so we tested optimized polyamine depletion regimens for activity against neuroblastoma. EXPERIMENTAL DESIGN: We used complementary transgenic and xenograft-bearing neuroblastoma models to assess polyamine antagonists. We investigated difluoromethylornithine (DFMO; an inhibitor of Odc, the rate-limiting enzyme in polyamine synthesis), SAM486 (an inhibitor of Amd1, the second rate-limiting enzyme), and celecoxib (an inducer of Sat1 and polyamine catabolism) in both the preemptive setting and in the treatment of established tumors. In vitro assays were performed to identify mechanisms of activity. RESULTS: An optimized polyamine antagonist regimen using DFMO and SAM486 to inhibit both rate-limiting enzymes in polyamine synthesis potently blocked neuroblastoma initiation in transgenic mice, underscoring the requirement for polyamines in MYC-driven oncogenesis. Furthermore, the combination of DFMO with celecoxib was found to be highly active, alone, and combined with numerous chemotherapy regimens, in regressing established tumors in both models, including tumors harboring highest risk genetic lesions such as MYCN amplification, ALK mutation, and TP53 mutation with multidrug resistance. CONCLUSIONS: Given the broad preclinical activity demonstrated by polyamine antagonist regimens across diverse in vivo models, clinical investigation of such approaches in neuroblastoma and potentially other MYC-driven tumors is warranted. Clin Cancer Res; 22(17); 4391-404. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Transformación Celular Neoplásica/efectos de los fármacos , Neuroblastoma/etiología , Neuroblastoma/patología , Poliaminas/antagonistas & inhibidores , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Celecoxib/farmacología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Eflornitina/farmacología , Genes myc , Homeostasis/efectos de los fármacos , Humanos , Ratones , Ratones Transgénicos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/mortalidad , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Poliaminas/metabolismo , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Artículo en Inglés | MEDLINE | ID: mdl-26780219

RESUMEN

Polyamines are a family of low molecular weight organic cations produced in part by the coordinated actions of arginase II (Arg II) and ornithine decarboxylase (Odc). Although gill polyamine homeostasis is affected by acute transfer to fresh water, little is known of its function in fish osmoregulation. The current study investigated the role of polyamines in the compensatory response of hypoosmotic challenge in the euryhaline fish, Fundulus grandis. Adult F. grandis were acclimated to 5 ppt water, transferred abruptly to 5, 2, 1, 0.5 and 0.1 ppt water, and assessed for osmoregulatory function, gill morphology, and polyamine homeostasis. The plasma osmolality, Na(+) concentration, and Cl(-) concentration were only significantly reduced during exposure to salinities at or below 0.5 ppt, although these effects were transient except in the 0.1 ppt treatment. The phenotype of mitochondrion-rich cells (MRCs) shifted from a seawater-type to a freshwater-type only at salinities that also produced a plasma osmotic disturbance. Hypoosmotic exposure increased the concentrations of putrescine, spermidine, and spermine in the gill over the entire 7 day period. Exposure to 0.1 ppt water also transiently increased gill caspase-3 activity and gill mRNA levels of the immediate-early response genes, c-fos and c-myc, thus tightly associating polyamines with gill remodeling during freshwater acclimation. Furthermore, arginase II and ornithine decarboxylase mRNA levels were most highly expressed in MRCs, and these levels were further increased only in the 0.1 ppt treatment. Reduction of gill polyamine levels following administration of the Odc inhibitor, alpha-dl-difluoromethylornithine (DFMO), inhibited gill caspase-3 activity, but surprisingly reduced the magnitude of the plasma osmotic imbalance elicited by exposure to 0.1 ppt water. We used isolated opercular epithelia mounted on Ussing chambers to assess the influence of polyamines on the attenuating response of hypotonic shock on active Cl(-) secretion. Spermidine partially reduced the decrease of short-circuit current (Isc) and membrane conductance (Gt) produced by hypotonic exposure. These data suggest polyamines blunt the hypotonic inhibition of NaCl secretion and may lead to early apoptosis of seawater ionocytes and their replacement by FW-type ionocytes.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Apoptosis , Fundulidae/fisiología , Branquias/metabolismo , Osmorregulación , Poliaminas/metabolismo , Estrés Fisiológico , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Arginasa/genética , Arginasa/metabolismo , Caspasa 3/química , Caspasa 3/genética , Caspasa 3/metabolismo , Proteínas de Peces/antagonistas & inhibidores , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Fundulidae/sangre , Regulación de la Expresión Génica/efectos de los fármacos , Branquias/efectos de los fármacos , Branquias/ultraestructura , Golfo de México , Isoenzimas/genética , Isoenzimas/metabolismo , Louisiana , Potenciales de la Membrana/efectos de los fármacos , Ornitina Descarboxilasa/química , Ornitina Descarboxilasa/genética , Ornitina Descarboxilasa/metabolismo , Inhibidores de la Ornitina Descarboxilasa/farmacología , Osmorregulación/efectos de los fármacos , Poliaminas/antagonistas & inhibidores , Poliaminas/sangre , Distribución Aleatoria , Salinidad , Cloruro de Sodio/sangre , Cloruro de Sodio/metabolismo , Estrés Fisiológico/efectos de los fármacos
15.
Biochem Pharmacol ; 96(3): 179-89, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26070250

RESUMEN

Tumor cell growth requires large iron quantities and the deprivation of this metal induced by synthetic metal chelators is therefore an attractive method for limiting the cancer cell proliferation. The antiproliferative effect of the Quilamine HQ1-44, a new iron chelator vectorized toward tumor cells by a polyamine chain, is related to its high selectivity for the Polyamine Transport System (PTS), allowing its preferential uptake by tumoral cells. The difference in PTS activation between healthy cells and tumor cells enables tumor cells to be targeted, whereas the strong dependence of these cells on iron ensures a secondary targeting. Here, we demonstrated in vitro that HQ1-44 inhibits DNA synthesis and cell proliferation of HCT116 cells by modulating the intracellular metabolism of both iron and polyamines. Moreover, in vivo, in xenografted athymic nude mice, we found that HQ1-44 was as effective as cis-platin in reducing HCT116 tumor growth, without its side effects. Furthermore, as suggested by in vitro data, the depletion in exogenous or endogenous polyamines, known to activate the PTS, dramatically enhanced the antitumor efficiency of HQ1-44. These data support the need for further studies to assess the value of HQ1-44 as an adjuvant treatment in cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , ADN de Neoplasias/antagonistas & inhibidores , Eflornitina/farmacología , Quelantes del Hierro/farmacología , Poliaminas/antagonistas & inhibidores , Animales , Transporte Biológico/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , ADN de Neoplasias/biosíntesis , Femenino , Células HCT116 , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Trasplante de Neoplasias , Poliaminas/metabolismo , Trasplante Heterólogo , Carga Tumoral/efectos de los fármacos
16.
J Biol Chem ; 290(10): 6106-19, 2015 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-25593318

RESUMEN

Polyamines are essential for cell proliferation, and their levels are elevated in many human tumors. The oncogene n-myc is known to potentiate polyamine metabolism. Neuroblastoma, the most frequent extracranial solid tumor in children, harbors the amplification of n-myc oncogene in 25% of the cases, and it is associated with treatment failure and poor prognosis. We evaluated several metabolic features of the human neuroblastoma cell lines Kelly, IMR-32, and SK-N-SH. We further investigated the effects of glycolysis impairment in polyamine metabolism in these cell lines. A previously unknown linkage between glycolysis impairment and polyamine reduction is unveiled. We show that glycolysis inhibition is able to trigger signaling events leading to the reduction of N-Myc protein levels and a subsequent decrease of both ornithine decarboxylase expression and polyamine levels, accompanied by cell cycle blockade preceding cell death. New anti-tumor strategies could take advantage of the direct relationship between glucose deprivation and polyamine metabolism impairment, leading to cell death, and its apparent dependence on n-myc. Combined therapies targeting glucose metabolism and polyamine synthesis could be effective in the treatment of n-myc-expressing tumors.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Neuroblastoma/genética , Poliaminas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Línea Celular Tumoral , Desoxiglucosa/administración & dosificación , Regulación Neoplásica de la Expresión Génica , Glucólisis/efectos de los fármacos , Humanos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología , Poliaminas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética
17.
PLoS One ; 9(6): e99184, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24921942

RESUMEN

In Alzheimer's disease (AD), the ß-amyloid peptide (Aß) has been causally linked to synaptic dysfunction and cognitive impairment. Several studies have shown that N-Methyl-D-Aspartate receptors (NMDAR) activation is involved in the detrimental actions of Aß. Polyamines, like spermidine and spermine, are positive modulators of NMDAR function and it has been shown that their levels are regulated by Aß. In this study we show here that interruption of NMDAR modulation by polyamines through blockade of its binding site at NMDAR by arcaine (0.02 nmol/site), or inhibition of polyamine synthesis by DFMO (2.7 nmol/site), reverses Aß25-35-induced memory impairment in mice in a novel object recognition task. Incubation of hippocampal cell cultures with Aß25-35 (10 µM) significantly increased the nuclear accumulation of Jacob, which is a hallmark of NMDAR activation. The Aß-induced nuclear translocation of Jacob was blocked upon application of traxoprodil (4 nM), arcaine (4 µM) or DFMO (5 µM), suggesting that activation of the polyamine binding site at NMDAR located probably at extrasynaptic sites might underlie the cognitive deficits of Aß25-35-treated mice. Extrasynaptic NMDAR activation in primary neurons results in a stripping of synaptic contacts and simplification of neuronal cytoarchitecture. Aß25-35 application in hippocampal primary cell cultures reduced dendritic spine density and induced alterations on spine morphology. Application of traxoprodil (4 nM), arcaine (4 µM) or DFMO (5 µM) reversed these effects of Aß25-35. Taken together these data provide evidence that polyamine modulation of extrasynaptic NMDAR signaling might be involved in Aß pathology.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Trastornos de la Memoria/inducido químicamente , Trastornos de la Memoria/patología , Fragmentos de Péptidos/toxicidad , Poliaminas/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Animales , Biguanidas/farmacología , Sitios de Unión , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/patología , Conducta Exploratoria/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/patología , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Ratones , Proteínas del Tejido Nervioso/metabolismo , Piperidinas/farmacología , Poliaminas/metabolismo , Espermidina/administración & dosificación , Espermidina/farmacología , Sinapsis/efectos de los fármacos
18.
Cancer Immunol Res ; 2(3): 274-85, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24778323

RESUMEN

Correcting T-cell immunosuppression may unleash powerful antitumor responses; however, knowledge about the mechanisms and modifiers that may be targeted to improve therapy remains incomplete. Here, we report that polyamine elevation in cancer, a common metabolic aberration in aggressive lesions, contributes significantly to tumor immunosuppression and that a polyamine depletion strategy can exert antitumor effects that may also promote immunity. A polyamine-blocking therapy (PBT) that combines the well-characterized ornithine decarboxylase (ODC) inhibitor difluoromethylornithine (DFMO) with AMXT 1501, a novel inhibitor of the polyamine transport system, blocked tumor growth in immunocompetent mice but not in athymic nude mice lacking T cells. PBT had little effect on the proliferation of epithelial tumor cells, but it increased the number of apoptotic cells. Analysis of CD45(+) tumor immune infiltrates revealed that PBT decreased levels of Gr-1(+)CD11b(+) myeloid suppressor cells and increased CD3(+) T cells. Strikingly, in a model of neoadjuvant therapy, mice administered with PBT one week before surgical resection of engrafted mammary tumors exhibited resistance to subsequent tumor rechallenge. Collectively, our results indicate that therapies targeting polyamine metabolism do not act exclusively as antiproliferative agents, but also act strongly to prevent immune escape by the tumor. PBT may offer a general approach to heighten immune responses in cancer.


Asunto(s)
Antineoplásicos/farmacología , Eflornitina/farmacología , Poliaminas/antagonistas & inhibidores , Microambiente Tumoral/efectos de los fármacos , Animales , Línea Celular Tumoral , Terapia de Inmunosupresión , Ratones , Ratones Endogámicos , Ratones Desnudos , Neoplasias Experimentales , Microambiente Tumoral/genética
19.
Free Radic Biol Med ; 71: 36-48, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24589373

RESUMEN

Nitric oxide (NO) and polyamines (PAs) are two kinds of important signal in mediating plant tolerance to abiotic stress. In this study, we observed that both NO and PAs decreased alkaline stress in tomato plants, which may be a result of their role in regulating nutrient balance and reactive oxygen species (ROS), thereby protecting the photosynthetic system from damage. Further investigation indicated that NO and PAs induced accumulation of each other. Furthermore, the function of PAs could be removed by a NO scavenger, cPTIO. On the other hand, application of MGBG, a PA synthesis inhibitor, did little to abolish the function of NO. To further elucidate the mechanism by which NO and PAs alleviate alkaline stress, the expression of several genes associated with abiotic stress was analyzed by qRT-PCR. NO and PAs significantly upregulated ion transporters such as the plasma membrane Na(+)/H(+) antiporter (SlSOS1), vacuolar Na(+)/H(+) exchanger (SlNHX1 and SlNHX2), and Na(+) transporter and signal components including ROS, MAPK, and Ca(2+) signal pathways, as well as several transcription factors. All of these play important roles in plant adaptation to stress conditions.


Asunto(s)
Antioxidantes/metabolismo , Óxido Nítrico/metabolismo , Raíces de Plantas/metabolismo , Poliaminas/metabolismo , Hidróxido de Sodio/farmacología , Solanum lycopersicum/metabolismo , Adaptación Fisiológica , Benzoatos/farmacología , Transporte Biológico , Calcio/metabolismo , Regulación de la Expresión Génica de las Plantas , Hidroponía , Imidazoles/farmacología , Solanum lycopersicum/efectos de los fármacos , Solanum lycopersicum/genética , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mitoguazona/farmacología , Óxido Nítrico/antagonistas & inhibidores , Raíces de Plantas/efectos de los fármacos , Raíces de Plantas/genética , Poliaminas/antagonistas & inhibidores , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Plantones/efectos de los fármacos , Plantones/genética , Plantones/metabolismo , Transducción de Señal , Intercambiadores de Sodio-Hidrógeno/genética , Intercambiadores de Sodio-Hidrógeno/metabolismo , Estrés Fisiológico
20.
J Med Chem ; 57(2): 348-63, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24405276

RESUMEN

The development of polyamine transport inhibitors (PTIs), in combination with the polyamine biosynthesis inhibitor difluoromethylornithine (DFMO), provides a method to target cancers with high polyamine requirements. The DFMO+PTI combination therapy results in sustained intracellular polyamine depletion and cell death. A series of substituted benzene derivatives were evaluated for their ability to inhibit the import of spermidine in DFMO-treated Chinese hamster ovary (CHO) and L3.6pl human pancreatic cancer cells. Several design features were discovered which strongly influenced PTI potency, sensitivity to amine oxidases, and cytotoxicity. These included changes in (a) the number of polyamine chains appended to the ring system, (b) the polyamine sequence, (c) the attachment linkage of the polyamine to the aryl core, and (d) the presence of a terminal N-methyl group. Of the series tested, the optimal design was N(1),N(1'),N(1″)-(benzene-1,3,5-triyltris(methylene))tris(N(4)-(4-(methylamino)butyl)butane-1,4-diamine, 6b, which contained three N-methylhomospermidine motifs. This PTI exhibited decreased sensitivity to amine oxidases and low toxicity as well as high potency (EC50 = 1.4 µM) in inhibiting the uptake of spermidine (1 µM) in DFMO-treated L3.6pl human pancreatic cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Derivados del Benceno/farmacología , Eflornitina/farmacología , Poliaminas/metabolismo , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Derivados del Benceno/síntesis química , Derivados del Benceno/química , Transporte Biológico/efectos de los fármacos , Células CHO , Línea Celular Tumoral , Cricetinae , Cricetulus , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Humanos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Neoplasias Pancreáticas , Poliaminas/antagonistas & inhibidores , Relación Estructura-Actividad , Poliamino Oxidasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...