Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Biochem Biophys Rep ; 38: 101681, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38455592

RESUMO

Myeloid differentiation factor 88 (MyD88), which is a key regulator of nuclear factor kappa B (NF-κB), plays an important role in tumorigenesis in lymphoid malignancies such as Waldenstrom's macroglobulinemia (WM). However, its biological function in multiple myeloma (MM), which is a malignant plasma cell disorder like WM, remains unexplored. In this article, we first demonstrated that higher expression MyD88 was significantly correlated with poor survival in patients with MM using multiple publicly available datasets. Interestingly, bioinformatic analysis also revealed that MyD88 gene alteration, which is recognized in nearly 80% of patients with WM, was extremely rare in MM. In addition, ST2825 (a specific inhibitor of MyD88) suppressed cell growth followed by apoptosis. Furthermore, ST2825 induced intracellular reactive oxygen species (ROS) in MM cells, and N-acetyl-l-cysteine, which is known as a ROS scavenger, significantly decreased the number of apoptotic MM cells evoked by ST2825 treatment. Taken together, our results indicated that ST2825 leads to ROS-dependent apoptosis in MM cells and could be an attractive therapeutic candidate for patients with MM. By highlighting the pathological mechanism of MyD88 in MM, this study also provides novel treatment strategies to conquer MM.

2.
Jpn J Clin Oncol ; 54(1): 47-53, 2024 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-37791389

RESUMO

BACKGROUND: Accumulating evidence has demonstrated platinum-based chemotherapy followed by maintenance therapy with a poly Adenosine diphosphate (ADP)-ribose polymerase inhibitor (olaparib) show benefits in unresectable pancreatic cancer with a germline (g)BRCA1/2 mutation. Evaluation of the germline BRCA1 and BRCA2 mutation is essential for making decisions on a treatment strategy for patients with unresectable pancreatic cancer. However, the detection rates of germline BRCA1 and BRCA2 mutations and efficacy of maintenance with olaparib remain undetermined, prospectively, in Japan. METHODS & RESULTS: In this prospective analysis, the rate of germline BRCA1 and BRCA2 mutations and efficacy of chemotherapy were analyzed in 136 patients with pancreatic cancer who underwent BRACAnalysis® (85 patients) or FoundationOne® CDx (51 patients) between January 2020 and July 2022. A total of six patients (4.4%) had a germline BRCA1 and BRCA2 mutation. Five patients were treated with modified FOLFIRINOX and one with fluorouracil and oxaliplatin. All patients continued platinum-based chemotherapy for ˃4 months and were subsequently treated with olaparib as a maintenance therapy. The response rate to platinum-based chemotherapy in the germline BRCA1 and BRCA2 mutation-positive group was significantly better than that of the germline BRCA1 and BRCA2 mutation-negative group (66% vs 23%, P = 0.04). All patients harbouring a germline BRCA1 and BRCA2 mutation were able to switch to olaparib. The median progression-free survival using olaparib was 5.7 months (range 3.0-9.2). CONCLUSIONS: The rate of germline BRCA1 and BRCA2 mutations found in patients with unresectable pancreatic cancer was comparable to those of previous studies.An analysis of germline BRCA1 and BRCA2 mutations has benefits for all patients with unresectable pancreatic cancer with regard to decisions on therapeutic strategies in a clinical practice setting.


Assuntos
Antineoplásicos , Neoplasias Ovarianas , Neoplasias Pancreáticas , Feminino , Humanos , Proteína BRCA1/genética , Antineoplásicos/uso terapêutico , Estudos Prospectivos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Proteína BRCA2/genética , Neoplasias Ovarianas/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Genes BRCA1 , Genes BRCA2 , Mutação , Ftalazinas/uso terapêutico , Ftalazinas/efeitos adversos , Mutação em Linhagem Germinativa
3.
Clin J Gastroenterol ; 17(1): 164-169, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37882973

RESUMO

A 50-year-old man with a history of total gastrectomy, distal pancreatectomy, splenectomy, and Roux-en-Y reconstruction was admitted to our hospital with a gallbladder tumor that had infiltrated the liver and abdominal wall. Because malignant cells were not collected during the percutaneous biopsy, we planned to perform an endoscopic ultrasound-guided fine-needle biopsy with a 22-G Franseen needle using a forward-viewing echoendoscope. Using intermittent manual compression, the forward-viewing echoendoscope reached the duodenum under fluoroscopic guidance. Endoscopic ultrasound-guided fine-needle biopsy was performed using a 22-G needle and 20-mL syringe and yielded a sufficient specimen with a single puncture. Malignant cells were promptly identified during on-site evaluation. The composition of the specimen (> 20% cancer cells and tissue area exceeding 25 mm2) enabled comprehensive genomic profiling. Subsequently, high-tumor mutational burden was diagnosed based on comprehensive genomic profiling, and pembrolizumab was initiated as a second-line therapy. Even in cases involving Roux-en-Y reconstruction, endoscopic ultrasound-guided fine-needle biopsy using a forward-viewing echoendoscope can result in collection of a high-quality specimen.


Assuntos
Carcinoma in Situ , Neoplasias da Vesícula Biliar , Masculino , Humanos , Pessoa de Meia-Idade , Neoplasias da Vesícula Biliar/diagnóstico por imagem , Neoplasias da Vesícula Biliar/genética , Neoplasias da Vesícula Biliar/cirurgia , Endossonografia , Duodeno , Gastrectomia , Genômica , Aspiração por Agulha Fina Guiada por Ultrassom Endoscópico
4.
Intern Med ; 63(15): 2215-2219, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38104989

RESUMO

A 26-year-old man presented with a tumor in the left soleus muscle. The tumor was diagnosed as a locally advanced leiomyosarcoma. The patient was treated with irradiation followed by wide resection. One year after surgery, the patient presented with multiple lung metastases. Despite aggressive sequential chemotherapy, systemic metastatic tumors continued to develop. To explore therapeutic options for the patient, we performed DNA-based CGP with FoundationOne® CDx (F1). F1 identified an out-of-strand rearrangement of the NOS1AP::NTRK1 gene, which has not been previously reported. In contrast, RNA sequencing revealed an in-frame LMNA::NTRK1 gene, which is an oncogenic fusion gene.


Assuntos
Lamina Tipo A , Leiomiossarcoma , Humanos , Masculino , Adulto , Leiomiossarcoma/genética , Leiomiossarcoma/diagnóstico , Leiomiossarcoma/patologia , Lamina Tipo A/genética , Receptor trkA/genética , Análise de Sequência de RNA , Proteínas de Fusão Oncogênica/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/diagnóstico , Neoplasias Musculares/genética , Neoplasias Musculares/secundário , Neoplasias Musculares/patologia
5.
Front Immunol ; 14: 1259071, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37753079

RESUMO

Ipilimumab plus nivolumab therapy is approved for patients with unresectable advanced esophageal squamous cell carcinoma (ESCC). Although a combination of immune checkpoint inhibitors (ICIs), compared to conventional chemotherapy, can improve overall survival in patients with advanced ESCC, this increases the incidence of immune-related adverse events (irAEs). Here, we describe an ESCC case that developed pemphigus vulgaris (PV), an extremely rare cutaneous irAE, during ipilimumab plus nivolumab treatment. The patient achieved a partial response to treatment. The PV was successfully managed after the cessation of ipilimumab and the use of a topical steroid. We should thus re-treat ESCC with nivolumab monotherapy. In the era of ICIs as standard cancer therapeutics, diagnostic criteria for blistering diseases need to be established to properly manage patients with cutaneous irAEs.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Pênfigo , Humanos , Carcinoma de Células Escamosas do Esôfago/diagnóstico , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Pênfigo/diagnóstico , Pênfigo/tratamento farmacológico , Ipilimumab/efeitos adversos , Nivolumabe/efeitos adversos , Neoplasias Esofágicas/tratamento farmacológico
6.
Reprod Biol ; 23(1): 100726, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36603297

RESUMO

Expression of desmin, an intermediate filament, in the myometrium and cervix were investigated in peripartum rats (full term day 22 of pregnancy (DP22)). Des mRNA was expressed in lesser amounts in the cervix at peripartum (DP17 and 21, and day of birth 1 (DB1)), compared to those in the cervixes of ovariectomized rats. Immunohistochemical analysis revealed that desmin protein was diffusely present in the myometrium, and locally in the epithelium of the cervix. Western blot analysis showed that desmin protein levels in the myometrium increased 4- to 6-fold at DP17, 21 and DB1, and decreased rapidly at DB2 to the basal level observed in ovariectomized or non-pregnant rats. In contrast, cervical desmin protein levels increased approximately 10-fold at DP21 compared to those in ovariectomized rats, but decreased rapidly at DB1, indicating its decrease at parturition and an inconsistency between mRNA and protein expression. The administration of 17ß-estradiol to ovariectomized rats increased desmin protein levels in the myometrium and cervix after 24 h. S-nitrosylated desmin protein was detected in the myometrium and cervix at DP21. The mRNA expression of inducible nitric oxide synthase was consistent with the expression of desmin protein. Thus, desmin, which is regulated by estradiol, is differentially expressed in the myometrium and cervix at peripartum possibly for successful pregnancy and parturition. In the cervix, desmin protein expression seems to be regulated by estradiol at the translational level. S-nitrosylation of desmin may have a potential role in the peripartum uterus.


Assuntos
Colo do Útero , Miométrio , Gravidez , Feminino , Ratos , Animais , Miométrio/metabolismo , Desmina/metabolismo , Útero/metabolismo , Parto , Estradiol/metabolismo , RNA Mensageiro/genética
7.
Ann Palliat Med ; 12(1): 205-211, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36226651

RESUMO

BACKGROUND: Uterine leiomyosarcoma (U-LMS) is an aggressive malignancy linked to a high risk of metastasis to distant major organs. Although cardiac metastasis of U-LMS is extremely rare, it is often associated with life-threatening conditions, leading to dismal prognosis. Currently, there is no established therapy for patients with unresectable/relapsed cardiac metastasis of U-LMS. In this article, we report a case of locally relapsed cardiac metastasis of U-LMS, which occurred 3 years after surgical resection, successfully treated with eribulin. CASE DESCRIPTION: A 69-year-old female with cardiac symptoms (e.g., dyspnea, tachycardia, and easy fatigability) was sequentially treated with doxorubicin plus ifosfamide, gemcitabine plus docetaxel, and pazopanib. However, cardiac metastasis continued to grow despite treatment. Hence, eribulin was administered as fourth-line therapy. Exceptionally, eribulin markedly improved cardiac symptoms, and the patient achieved a durable response for 17 months without the development of severe adverse events. Moreover, the volume of the metastatic cardiac tumor was decreased by 70%. Despite the poor prognosis of this condition, the patient survived for 8 years from the diagnosis of cardiac metastasis due to surgery and the continuous administration of chemotherapies. CONCLUSIONS: Eribulin may be an effective and accessible treatment option for cardiac metastasis of U-LMS in patients with life-threatening conditions for whom surgery is not indicated. Additionally, the expression levels of phosphorylated AKT (p-AKT) may be a predictive biomarker for the sensitivity of patients with U-LMS to treatment with eribulin.

8.
Nitric Oxide ; 127: 10-17, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35835264

RESUMO

Neuronal differentiation of adipose tissue-derived stem cells (ASCs) is potently promoted by valproic acid (VPA) through a gaseous signaling molecule, nitric oxide (NO). Here, we investigated the involvement of hydrogen sulfide (H2S), another gaseous signaling molecule, in neuronal differentiation of ASCs. VPA-promoted neuronal differentiation of ASCs was accompanied by increased intracellular H2S and sulfane sulfur with increased mRNA expression of enzymes synthesizing sulfane sulfur including cystathionine ß-synthase (CBS), of which inhibition reduced the differentiation efficiency. H2S donors, GYY4137 (GYY) or NaHS, potently promoted neuronal differentiation of ASCs when cAMP-elevating agents, dibutyryl cyclic adenosine monophosphate and isobutyl methyl-xanthine, were added as neuronal induction medium (NIM). Neuronal differentiation of ASCs promoted by NaHS or GYY was accompanied by Ca2+ entry and increased mRNA expression of voltage-gated Ca2+ channels. NaHS or GYY also increased mRNA expression of enzymes of the NO-citrulline cycle including inducible NO synthase (iNOS). It was concluded from these results that H2S potently promoted differentiation of ASCs into neuronal cells expressing functional voltage-gated Ca2+ channels with the aid of cAMP-elevating agents, involving NO-mediated signaling cascade. These effects of H2S were also considered as a partial mechanism for the VPA-promoted neuronal differentiation of ASCs.


Assuntos
Sulfeto de Hidrogênio , Tecido Adiposo/metabolismo , Sulfeto de Hidrogênio/metabolismo , Sulfeto de Hidrogênio/farmacologia , Óxido Nítrico/metabolismo , RNA Mensageiro , Células-Tronco/metabolismo , Enxofre
9.
Biochem Biophys Res Commun ; 589: 55-62, 2022 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-34891042

RESUMO

The differentiation of adipose tissue-derived stem cells (ASCs) to neuronal cells is greatly promoted by valproic acid (VPA), and is synergistically enhanced by the following treatment with neuronal induction medium (NIM) containing cAMP-elevating agents. In the present study, we investigated the synergism between VPA and NIM in neuronal differentiation of ASCs, assessed by the expression of neurofilament medium polypeptide (NeFM), with respect to Ca2+ entry. VPA (2 mM) treatment for 3 days followed by NIM for 2 h synergistically increased the incidence of neuronal cells differentiated from ASCs to an extent more than VPA alone treatment for 6 days, shortening the time required for the differentiation. VPA increased intracellular Ca2+ and the mRNAs of voltage-gated Ca2+ channels, Cacna1b (Cav2.2) and Cacna1h (Cav3.2), in ASCs. Inward currents through Ca2+ channels were evoked electrophysiologically at high voltage potential in ASCs treated with VPA. NIM reduced the mRNAs of NeFM and Cacna1b in VPA-promoted neuronal differentiation of ASCs. It was concluded that functional N-type voltage-gated Ca2+ channels (Cav2.2) are selectively expressed in VPA-promoted neuronal differentiation of ASCs. NIM seems to enhance the mRNA translation of molecules required for the differentiation. Neuronal cells obtained from ASCs by this protocol will be used as a cell source for regenerative therapy of neurological disorders associated with altered Cav2.2 activity.


Assuntos
Tecido Adiposo/citologia , Canais de Cálcio Tipo N/metabolismo , Diferenciação Celular , Neurônios/citologia , Neurônios/metabolismo , Células-Tronco/citologia , Ácido Valproico/farmacologia , Animais , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Meios de Cultura , Masculino , Neurônios/efeitos dos fármacos , Ratos Wistar , Células-Tronco/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
10.
Nitric Oxide ; 106: 35-44, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33129925

RESUMO

Valproic acid (VPA) remarkably promotes the differentiation of adipose tissue-derived stem cells (ASCs) to mature neuronal cells through nitric oxide (NO) signaling due to up-regulated inducible NO synthase (iNOS) as early as within 3 days. Here, we investigated mechanisms of VPA-promoted neuronal differentiation of ASCs concerning the NO-citrulline cycle, the metabolic cycle producing NO. Cultured rat ASCs were differentiated to mature neuronal cells rich in dendrites and expressing a neuronal marker by treatments with VPA at 2 mM for 3 days and subsequently with the neuronal induction medium for 2 h. Inhibitor (α-methyl-d, l-aspartic acid, MDLA) of arginosuccinate synthase (ASS), a key enzyme of the NO-citrulline cycle, abolishes intracellular NO increase and VPA-promoted neuronal differentiation in ASCs. l-Arginine, the substrate of iNOS, restores the promotion effect of VPA, being against MDLA. Immunocytochemistry showed that ASS and iNOS were increased in ASCs expressing neurofilament medium polypeptide (NeFM), a neuronal marker, by VPA and NIM synergistically. Real-time RT-PCR analysis showed that mRNAs of Ass and arginosuccinate lyase (Asl) in the NO-citrulline cycle were increased by VPA. Chromatin immunoprecipitation assay indicated that Ass and Asl were up-regulated by VPA through the acetylation of their associated histone. From these results, it was considered that VPA up-regulated the whole NO-citrulline cycle, which enabled continuous NO production by iNOS in large amounts for potent iNOS-NO signaling to promote neuronal differentiation of ASCs. This may also indicate a mechanism enabling short-lived NO to function conveniently as a potent signaling molecule that can disappear quickly after its role.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Citrulina/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/metabolismo , Ácido Valproico/farmacologia , Tecido Adiposo/citologia , Animais , Arginina/farmacologia , Argininossuccinato Sintase/antagonistas & inibidores , Argininossuccinato Sintase/metabolismo , Inibidores Enzimáticos/farmacologia , Masculino , N-Metilaspartato/análogos & derivados , N-Metilaspartato/farmacologia , Neurônios/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Ratos Wistar , Regulação para Cima/efeitos dos fármacos
11.
J Reprod Immunol ; 141: 103167, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32629316

RESUMO

Interleukin (IL)-18 is an inflammasome-mediated cytokine produced by germ cells, Leydig cells, and resident macrophages that is indispensable in the maintenance of homeostasis in the testis. We previously demonstrated that endogenous IL-18 induces testicular germ cell apoptosis during acute inflammation when plasma IL-18 levels are very high. However, the impact of acute inflammation and IL-18 on Leydig cells remained unclear. TM3 cells, a mouse Leydig cell line, and RAW264.7 cells, a mouse macrophage cell line, were stimulated with lipopolysaccharide (LPS) or recombinant IL-18 (rIL-18). We assessed the expression of inflammatory cytokines, caspase cleavage, and markers of apoptotic pathways. In Leydig cells, caspase 3 cleavage was increased and death-receptor-mediated apoptotic pathways were activated after LPS stimulation. However, LPS stimulation did not increase IL-18 expression in the Leydig cell line. When high-dose rIL-18 was administered to the Leydig cell line to mimic levels seem after inflammation, rIL-18 upregulated Tnf-α mRNA, Fadd mRNA, and Fas protein, promoted cleavage of caspase-8 and caspase-3, and induced apoptosis. Low-dose rIL-18 did not stimulate apoptosis. To determine if the high level of IL-18 seen in the testes after inflammation was derived from immune cells, we examined IL-18 protein expression in a macrophage cell line, RAW264.7. In contrast to the TM3 cells, IL-18 was significantly increased in RAW264.7 cells after LPS stimulation. These results suggest that high-dose IL-18 derived from macrophages is harmful to Leydig cells. Reducing the overexpression of IL-18 could be a new therapeutic approach to prevent Leydig cell apoptosis as a result of acute inflammation.


Assuntos
Apoptose/imunologia , Estado Terminal , Interleucina-18/metabolismo , Células Intersticiais do Testículo/patologia , Orquite/imunologia , Animais , Modelos Animais de Doenças , Proteína de Domínio de Morte Associada a Fas/metabolismo , Humanos , Inflamassomos/metabolismo , Células Intersticiais do Testículo/imunologia , Células Intersticiais do Testículo/metabolismo , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Orquite/patologia , Células RAW 264.7 , Transdução de Sinais/imunologia
12.
Pancreas ; 49(1): 135-142, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31856089

RESUMO

OBJECTIVES: Pancreatic cancer (PC) is highly aggressive with multiple oncogenic mutations. The efficacy of current chemotherapy is poor, and new therapeutic targets are needed. The forkhead box (FOX) proteins are multidirectional transcriptional factors strongly implicated in malignancies. Their expression is consistently suppressed by several oncogenic pathways such as PI3K/AKT signaling activated in PC. A recent study showed that class IIa histone deacetylases (HDAC) can act as a transcriptional suppressor. In this study, we hypothesized that HDAC class IIa inhibition would upregulate FOXO3a expression, thereby inducing its transcription-dependent antitumor effects. METHODS: We confirmed the change of FOXO3a expression and the effect of the cell growth inhibition by HDAC class IIa inhibition in AsPC-1 cells. Because FOXO3a is subject to ubiquitylation-mediated proteasome degradation, we examined the synergistic activation of FOXO3a by HDAC class IIa selective inhibitor TMP269 combined with proteasome inhibitor carfilzomib. RESULTS: We observed that TMP269 induced FOXO3a expression in a dose-dependent manner and inhibited cell growth in AsPC-1 cells. G1/S arrest was observed. FOXO3a expression was further increased and cell growth inhibition was dramatically enhanced by TMP269 combined with carfilzomib. CONCLUSIONS: Dual inhibition of class IIa HDACs and proteasome could be a promising new strategy for modifying FOXO3a activity against PC.


Assuntos
Proteína Forkhead Box O3/genética , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Neoplasias Pancreáticas/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sinergismo Farmacológico , Proteína Forkhead Box O3/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Perfilação da Expressão Gênica/métodos , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Oligopeptídeos/farmacologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteassoma/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
13.
Nitric Oxide ; 93: 1-5, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31484045

RESUMO

Valproic acid (VPA) remarkably promotes the differentiation of adipose tissue-derived stem cells (ASCs) to mature neuronal cells, enabling neuronal induction within only three days. Here, we investigated the involvement of NO-signaling in the VPA-promoted neuronal differentiation of ASCs as a possible mechanism. Cultured rat ASCs were differentiated to matured neuronal cells rich in dendrites and expressing ßIII-tubulin protein, a neuronal marker, by treatments with VPA at 2 mM for 3 days and subsequently with the neuronal induction medium (NIM) containing cAMP-elevating agents for 2 h. Increased intracellular NO was detected in neuronal cells differentiated from ASCs treated with VPA by a fluorescence NO-specific probe, diaminofluorescein-FM diacetate. However, a NO donor (NOC18) increased the incidence of neuronal cells only to a lesser extent than VPA, indicating the insufficiency of exogenous NO. RT-PCR analysis of ASCs treated with VPA showed increased mRNA expression of inducible nitric oxide synthase (iNOS) with the acetylation of its associated histone H3K9. iNOS inhibitors (1400 W and dexamethasone) or a soluble guanylate cyclase (sGC) inhibitor (ODQ) decreased the incidence of neuronal cells differentiated from ASCs treated with VPA. These inhibitors also decreased the mRNA expression of mature neuronal markers, neurofilament medium polypeptide (NeFM) and microtubule-associated protein 2 (MAP2), as well as ßIII-tubulin (TUBB3), to various extents. It was considered from these results that VPA promoted mature neuronal differentiation of ASCs through the iNOS-NO-sGC signaling pathway. This provided insights into the regulated neuronal differentiation of ASCs in clinical applications.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/metabolismo , Ácido Valproico/farmacologia , Tecido Adiposo/citologia , Animais , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/metabolismo , Ratos Wistar , Guanilil Ciclase Solúvel/metabolismo
14.
Int J Clin Oncol ; 23(6): 1023-1028, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30121869

RESUMO

BACKGROUND: Concomitant chemoradiotherapy (CCRT) produces severe mucositis and swallowing dysfunction, often resulting in malnutrition. Intensive nutrition support (INS) in addition to percutaneous endoscopic gastrostomy (PEG) is reported to decrease adverse effects during CCRT. PATIENTS AND METHODS: Fifty-eight patients with oropharyngeal cancer treated by CDDP-based CCRT were retrospectively analyzed. Twenty-nine patients treated with INS in addition to PEG were classified as INS group, and other 29 patients treated with PEG but without INS were classified as control group. RESULTS: INS in addition to PEG significantly increased calorie intake in the second half of CCRT and reduced adverse events including mucositis (p = 0.0019), leukopenia (p = 0.04), and renal function (p = 0.006). Moreover, 21 out of 29 patients had successfully administration of 200 mg/m2 or more of CDDP, while only 10 out of 29 patients had enough amount of CDDP in control group. CONCLUSIONS: These results suggest that INS in addition to prophylactic PEG not only decreases adverse effects but also may potentially improve oncological outcome of the patients with oropharyngeal cancer treated by CCRT.


Assuntos
Quimiorradioterapia , Nutrição Enteral , Gastrostomia , Desnutrição/prevenção & controle , Neoplasias Orofaríngeas/terapia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Apoio Nutricional , Prognóstico , Estudos Retrospectivos
15.
Kobe J Med Sci ; 63(4): E113-E122, 2018 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-29955023

RESUMO

OBJECTIVE: TALK score has been developed as an index for predicting the possibility of larynx preservation and prognosis of the patients with advanced laryngeal and hypopharyngeal cancers treated by concurrent chemoradiotherapy (CCRT). In this study, we validated the original TALK score in our clinical setting and modified the score for Japanese population. METHODS: The subjects were 21 patients with laryngeal cancer and 50 patients with hypopharyngeal cancer who underwent CCRT at Kobe University Hospital between April 2007 and August 2012. Data regarding T stage, serum albumin level just before treatment, maximum alcohol use, Karnofsky Performance Status (KPS), 3-year overall survival rate and 3-year larynx-preservation rate were collected from medical records. In the revised TALK score (JTALK), the following scoring criteria were modified to adjust Japanese population: T stage of the primary tumor (≥T3), serum albumin level (<3.5 g/dl), maximum alcohol use (≥one 350-ml can of beer/day or an equivalent amount), and KPS (<80%). RESULTS: The original TALK score was not significantly associated with the larynx preservation rates or survival rates for laryngeal cancer or hypopharyngeal cancer. Instead, JTALK score was significantly associated with the 3-year larynx preservation rates and 3-year survival rates of the patients with hypopharyngeal and laryngeal cancer. CONCLUSION: These results indicate that JTALK score could be a useful index for predicting the possibility of larynx preservation and prognosis of Japanese patients with advanced laryngeal and hypopharyngeal cancer treated by CCRT.


Assuntos
Neoplasias Hipofaríngeas/terapia , Neoplasias Laríngeas/terapia , Tratamentos com Preservação do Órgão , Índice de Gravidade de Doença , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
16.
J Toxicol Sci ; 42(6): 723-729, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29142171

RESUMO

Nanomaterials have been extensively used in our daily life, and may also induce health effects and toxicity. Nanomaterials can translocate from the outside to internal organs, including the brain. For example, both nano-ZnO and nano-TiO2 translocate into the brain via the olfactory pathway in rodents, possibly leading to toxic effects on the brain. Although the effects of nano-ZnO and nano-TiO2 on neuronal viability or neuronal excitability have been studied, no work has focused on how these nanomaterials affect neuronal differentiation and development. In this study, we investigated the effects of nano-ZnO and nano-TiO2 on neurite outgrowth of PC12 cells, a useful model system for neuronal differentiation. Surprisingly, the number, length, and branching of differentiated PC12 neurites were significantly suppressed by the 7-day exposure to nano-ZnO (in the range of 1.0 × 10-4 to 1.0 × 10-1 µg/mL), at which the cell viability was not affected. The number and length were also significantly inhibited by the 7-day exposure to nano-TiO2 (1.0 × 10-3 to 1.0 µg/mL), which did not have cytotoxic effects. These results demonstrate that the neurite outgrowth in differentiated PC12 cells was suppressed by sub-cytotoxic concentrations of nano-ZnO or nano-TiO2.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Nanoestruturas/toxicidade , Crescimento Neuronal/efeitos dos fármacos , Titânio/toxicidade , Óxido de Zinco/toxicidade , Erros Inatos do Metabolismo dos Aminoácidos , Animais , Encéfalo/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Hipoplasia do Esmalte Dentário , Depressão Química , Diabetes Mellitus , Relação Dose-Resposta a Droga , Nanismo , Deficiência Intelectual , Microcefalia , Neurônios/citologia , Condutos Olfatórios/metabolismo , Células PC12 , Ratos , Titânio/metabolismo , Óxido de Zinco/metabolismo
17.
J Med Invest ; 64(3.4): 255-261, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28954992

RESUMO

Hyperglycemia induces nonconcordant regulation of renal mitochondrial respiratory complexes, increases oxidative stress, and causes diabetic nephropathy. Hypertension is a complication associated with diabetes and involves glomerular hyperfiltration, the effects of which on mitochondrial respiratory complexes are not well understood. To investigate the effect of glomerular hyperfiltration on renal mitochondrial respiratory complexes, we used the 5/6 nephrectomized BKS. Cg-Dock7m+/+Leprdb/J, Dock7m+/+Leprdb mice (db/m-5/6Nx mice) as a model for glomerular hyperfiltration. The BKS. Cg-Dock7m+/+Leprdb/J, +Leprdb/+Leprdb mice (db/db mice), a model for type 2 diabetes, was used as the positive control. We investigated the activities and protein levels of the mitochondrial complex, and the mitochondrial DNA and adenosine triphosphate content in the kidneys of these models. Blood chemistry and renal histopathological examination were performed for characterization of the disease. Both models showed expansion of the mesangial matrix of the glomeruli, which is indicative of glomerular hyperfiltration. The activities of complexes I and IV and the protein levels of complexes I and III were nonconcordant in db/m-5/6Nx mice. In conclusion, we demonstrated that nonconcordant regulation of mitochondrial complexes in db/m-5/6Nx mice involved with glomerular hyperfiltration. The progression and/or severity of nephropathy might be affected through a synergistic effect of mitochondrial dysfunction in hyperglycemia and glomerular hyperfiltration. J. Med. Invest. 64: 255-261, August, 2017.


Assuntos
Nefropatias Diabéticas/etiologia , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Rim/metabolismo , Mitocôndrias/metabolismo , Animais , Nefropatias Diabéticas/metabolismo , Hiperglicemia/fisiopatologia , Masculino , Camundongos , Nefrectomia
18.
An Bras Dermatol ; 92(2): 184-190, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28538876

RESUMO

BACKGROUND:: A single, effective therapeutic regimen for keloids has not been established yet, and the development of novel therapeutic approaches is expected. Butyrate, a short-chain fatty acid, and docosahexaenoic acid (DHA), a ω-3 polyunsaturated fatty acid, play multiple anti-inflammatory and anticancer roles via their respective mechanisms of action. OBJECTIVE:: In this study, we evaluated the antifibrogenic effects of their single and combined use on keloid fibroblasts. METHODS:: Keloid fibroblasts were treated with butyrate (0-16 mM) and/or DHA (0-100 µM) for 48 or 96 h. RESULTS:: Butyrate inhibited cell proliferation, and α-smooth muscle actin (α-SMA) and type III collagen expressions, with inhibition of the transforming growth factor (TGF)-ß1 and TGF-ß type I receptor expressions and increased prostaglandin E2 with upregulation of cyclooxygenase-1 expression with induction of histone acetylation. DHA inhibited α-SMA, type III collagen, and TGF-ß type I receptor expressions. Then, the butyrate/DHA combination augmented the antifibrogenic effects, resulting in additional inhibition of α-SMA, type I and III collagen expressions, with strong disruption of stress fiber and apoptosis induction. Moreover, the butyrate/DHA combination inhibited the cyclooxygenase-2 expression, suggesting stronger anti-inflammatory effect than each monotherapy. STUDY LIMITATIONS:: Activation in keloid tissue is affected not only by fibroblasts but also by epithelial cells and immune cells. Evaluation of the effects by butyrate and DHA in these cells or in an in vivo study is required. CONCLUSION:: This study demonstrated that butyrate and docosahexaenoic acid have antifibrogenic effects on keloid fibroblasts and that these may exert therapeutic effects for keloid.


Assuntos
Butiratos/uso terapêutico , Ácidos Docosa-Hexaenoicos/uso terapêutico , Fibroblastos , Queloide/tratamento farmacológico , Proliferação de Células , Células Cultivadas , Colágeno Tipo I , Colágeno Tipo III , Terapia Combinada , Humanos , Proteínas Serina-Treonina Quinases , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta
19.
An. bras. dermatol ; An. bras. dermatol;92(2): 184-190, Mar.-Apr. 2017. tab, graf
Artigo em Inglês | LILACS | ID: biblio-838060

RESUMO

Abstract: Background: A single, effective therapeutic regimen for keloids has not been established yet, and the development of novel therapeutic approaches is expected. Butyrate, a short-chain fatty acid, and docosahexaenoic acid (DHA), a ω-3 polyunsaturated fatty acid, play multiple anti-inflammatory and anticancer roles via their respective mechanisms of action. Objective: In this study, we evaluated the antifibrogenic effects of their single and combined use on keloid fibroblasts. Methods: Keloid fibroblasts were treated with butyrate (0-16 mM) and/or DHA (0-100 µM) for 48 or 96 h. Results: Butyrate inhibited cell proliferation, and α-smooth muscle actin (α-SMA) and type III collagen expressions, with inhibition of the transforming growth factor (TGF)-β1 and TGF-β type I receptor expressions and increased prostaglandin E2 with upregulation of cyclooxygenase-1 expression with induction of histone acetylation. DHA inhibited α-SMA, type III collagen, and TGF-β type I receptor expressions. Then, the butyrate/DHA combination augmented the antifibrogenic effects, resulting in additional inhibition of α-SMA, type I and III collagen expressions, with strong disruption of stress fiber and apoptosis induction. Moreover, the butyrate/DHA combination inhibited the cyclooxygenase-2 expression, suggesting stronger anti-inflammatory effect than each monotherapy. Study limitations: Activation in keloid tissue is affected not only by fibroblasts but also by epithelial cells and immune cells. Evaluation of the effects by butyrate and DHA in these cells or in an in vivo study is required. Conclusion: This study demonstrated that butyrate and docosahexaenoic acid have antifibrogenic effects on keloid fibroblasts and that these may exert therapeutic effects for keloid.


Assuntos
Humanos , Butiratos/uso terapêutico , Ácidos Docosa-Hexaenoicos/uso terapêutico , Fibroblastos , Queloide/tratamento farmacológico , Células Cultivadas , Proteínas Serina-Treonina Quinases , Receptores de Fatores de Crescimento Transformadores beta , Terapia Combinada , Colágeno Tipo I , Colágeno Tipo III , Proliferação de Células
20.
J Ultrasound Med ; 36(5): 879-885, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28195362

RESUMO

OBJECTIVES: Sodium butyrate, an inhibitor of histone deacetylase, has several therapeutic actions, including anti-inflammation. These actions depend on the concentration of sodium butyrate. In addition, lower concentrations have shown no effect on inflammation. Sonoporation by ultrasound can modify the permeability of the cell plasma membrane. Thus, the effects of sodium butyrate may be enhanced by the ultrasonic acoustics. Therefore, the facilitative effects of low-intensity ultrasound on histone acetylation and interleukin 6 (IL-6) regulation by sodium butyrate were investigated in this study. METHODS: Human dermal fibroblasts were treated with 1-mM sodium butyrate for 3 hours with 20 minutes of 0.1-W/cm2 pulsed or continuous ultrasound irradiation at the beginning of the sodium butyrate treatments. RESULTS: The combination of treatments with sodium butyrate and ultrasound significantly increased histone acetylation in fibroblasts (P < .05), whereas sodium butyrate could not increase histone acetylation. In addition, this combined treatment significantly suppressed the IL-6 messenger RNA expression level with lipopolysaccharide stimulation for 1 hour (P < .05). Meanwhile, the treatment with sodium butyrate alone could not suppress IL-6 messenger RNA expression in fibroblasts. These effects were achieved with both 20% pulsed and continuous ultrasound but not observed with ultrasound treatment alone. CONCLUSIONS: These results suggest that low-intensity ultrasound treatment promotes the physiologic actions of sodium butyrate as a histone deacetylase inhibitor.


Assuntos
Ácido Butírico/farmacologia , Fibroblastos/metabolismo , Histonas/metabolismo , Interleucina-6/metabolismo , RNA Mensageiro/metabolismo , Ondas Ultrassônicas , Acetilação , Western Blotting , Técnicas de Cultura de Células , Inibidores de Histona Desacetilases/farmacologia , Humanos , Reação em Cadeia da Polimerase em Tempo Real
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA