Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Metabolism ; 151: 155746, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38016540

RESUMEN

BACKGROUND: Multinucleation is a hallmark of osteoclast formation and has a unique ability to resorb bone matrix. During osteoclast differentiation, the cytoskeleton reorganization results in the generation of actin belts and eventual bone resorption. Tetraspanins are involved in adhesion, migration and fusion in various cells. However, its function in osteoclast is still unclear. In this study, we identified Tm4sf19, a member of the tetraspanin family, as a regulator of osteoclast function. MATERIALS AND METHODS: We investigate the effect of Tm4sf19 deficiency on osteoclast differentiation using bone marrow-derived macrophages obtained from wild type (WT), Tm4sf19 knockout (KO) and Tm4sf19 LELΔ mice lacking the large extracellular loop (LEL). We analyzed bone mass of young and aged WT, KO and LELΔ mice by µCT analysis. The effects of Tm4sf19 LEL-Fc fusion protein were accessed in osteoclast differentiation and osteoporosis animal model. RESULTS: We found that deficiency of Tm4sf19 inhibited osteoclast function and LEL of Tm4sf19 was responsible for its function in osteoclasts in vitro. KO and LELΔ mice exhibited higher trabecular bone mass compared to WT mice. We found that Tm4sf19 interacts with integrin αvß3 through LEL, and that this binding is important for cytoskeletal rearrangements in osteoclast by regulating signaling downstream of integrin αvß3. Treatment with LEL-Fc fusion protein inhibited osteoclast function in vitro and administration of LEL-Fc prevented bone loss in an osteoporosis mouse model in vivo. CONCLUSION: We suggest that Tm4sf19 regulates osteoclast function and that LEL-Fc may be a promising drug to target bone destructive diseases caused by osteoclast hyper-differentiation.


Asunto(s)
Enfermedades Óseas , Resorción Ósea , Osteoporosis , Tetraspaninas , Animales , Ratones , Resorción Ósea/genética , Resorción Ósea/metabolismo , Diferenciación Celular , Integrina alfaVbeta3/metabolismo , Osteoclastos , Osteoporosis/genética , Osteoporosis/metabolismo , Tetraspaninas/genética , Tetraspaninas/metabolismo
2.
Inflamm Regen ; 43(1): 35, 2023 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-37438837

RESUMEN

BACKGROUND: This study aimed to investigate how aging alters the homeostasis of the colonic intestinal epithelium and regeneration after tissue injury using organoid models and to identify its underlying molecular mechanism. METHODS: To investigate aging-related changes in the colonic intestinal epithelium, we conducted organoid cultures from old (older than 80 weeks) and young (6-10 weeks) mice and compared the number and size of organoids at day 5 of passage 0 and the growth rate of organoids between the two groups. RESULTS: The number and size of organoids from old mice was significantly lower than that from young mice (p < 0.0001) at day 5 of passage 0. The growth rate of old-mouse organoids from day 4 to 5 of passage 0 was significantly slower than that of young-mouse organoids (2.21 times vs. 1.16 times, p < 0.001). RNA sequencing showed that TGF-ß- and cell cycle-associated genes were associated with the aging effect. With regard to mRNA and protein levels, Smad3 and p-Smad3 in the old-mouse organoids were markedly increased compared with those in the young-mouse organoids. Decreased expression of ID1, increased expression of p16INK4a, and increased cell cycle arrest were observed in the old mouse-organoids. Treatment with SB431542, a type I TGF-ß receptor inhibitor, significantly increased the formation and growth of old-mouse organoids, and TGF-ß1 treatment markedly suppressed the formation of young-mouse organoids. In the acute dextran sulfate sodium-colitis model and its organoid experiments, the colonic epithelial regeneration after tissue injury in old mice was significantly decreased compared with young mice. CONCLUSIONS: Aging reduced the formation ability and growth rate of colonic epithelial organoids by increasing cell cycle arrest through TGF-ß-Smad3-p16INK4a signaling.

3.
Exp Mol Med ; 55(6): 1218-1231, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37258579

RESUMEN

The signaling pathways governing acetaminophen (APAP)-induced liver injury have been extensively studied. However, little is known about the ubiquitin-modifying enzymes needed for the regulation of APAP-induced liver injury. Here, we examined whether the Pellino3 protein, which has E3 ligase activity, is needed for APAP-induced liver injury and subsequently explored its molecular mechanism. Whole-body Peli3-/- knockout (KO) and adenovirus-mediated Peli3 knockdown (KD) mice showed reduced levels of centrilobular cell death, infiltration of immune cells, and biomarkers of liver injury, such as alanine aminotransferase (ALT) and aspartate aminotransferase (AST), upon APAP treatment compared to wild-type (WT) mice. Peli3 deficiency in primary hepatocytes decreased mitochondrial and lysosomal damage and reduced the mitochondrial reactive oxygen species (ROS) levels. In addition, the levels of phosphorylation at serine 9 in the cytoplasm and mitochondrial translocation of GSK3ß were decreased in primary hepatocytes obtained from Peli3-/- KO mice, and these reductions were accompanied by decreases in JNK phosphorylation and mitochondrial translocation. Pellino3 bound more strongly to GSK3ß compared with JNK1 and JNK2 and induced the lysine 63 (K63)-mediated polyubiquitination of GSK3ß. In rescue experiments, the ectopic expression of wild-type Pellino3 in Peli3-/- KO hepatocytes restored the mitochondrial translocation of GSK3ß, but this restoration was not obtained with expression of a catalytically inactive mutant of Pellino3. These findings are the first to suggest a mechanistic link between Pellino3 and APAP-induced liver injury through the modulation of GSK3ß polyubiquitination.


Asunto(s)
Acetaminofén , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas , Animales , Ratones , Acetaminofén/efectos adversos , Fosforilación , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/metabolismo , Hígado/metabolismo , Hepatocitos/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ratones Endogámicos C57BL
4.
Cancers (Basel) ; 14(23)2022 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-36497457

RESUMEN

(1) Background: Desmoid tumors have a relatively high local failure rate after primary treatment using surgery and/or radiotherapy. Moreover, desmoid tumors recur at the primary site for many patients. An effective therapeutic strategy for the desmoid tumor is needed to maintain quality of life and prolong survival. (2) Method: First of all, we collected desmoid tumor tissues and investigated the status of protein expression for beta-catenin and alpha-SMA through immunohistochemistry. Then, we performed targeted sequencing and whole RNA sequencing. To compare the data with other cancer types, we used NGS data from sarcoma patients at Yonsei Cancer Center (YCC-sarcoma cohort, n = 48) and The Cancer Genome Atlas (TCGA, n = 9235). Secondly, we established the novel patient-derived preclinical models (n = 2) for the validation of treatment strategy. The same gene alteration of primary tissue was demonstrated. (3) Results: We discovered specific gene sets related to the TGF-ß signaling pathway. Moreover, we selected the combination treatment comprising TGF-ß inhibitor, vactosertib, and imatinib. In screening for the anti-proliferation effect, the combination treatment of TGF-ß inhibitor was more effective for tumor suppression than monotherapy. (4) Conclusion: We found preclinical indications that TGF-ß inhibitors could prove useful as a potential treatment for patients with desmoid tumors. Moreover, we could find some examples in clinical trials.

5.
Nat Commun ; 13(1): 6274, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-36307405

RESUMEN

Although accumulating evidence indicates that alternative splicing is aberrantly altered in many cancers, the functional mechanism remains to be elucidated. Here, we show that epithelial and mesenchymal isoform switches of leucine-rich repeat Fli-I-interacting protein 2 (LRRFIP2) regulated by epithelial splicing regulatory protein 1 (ESRP1) correlate with metastatic potential of gastric cancer cells. We found that expression of the splicing variants of LRRFIP2 was closely correlated with that of ESRP1. Surprisingly, ectopic expression of the mesenchymal isoform of LRRFIP2 (variant 3) dramatically increased liver metastasis of gastric cancer cells, whereas deletion of exon 7 of LRRFIP2 by the CRISPR/Cas9 system caused an isoform switch, leading to marked suppression of liver metastasis. Mechanistically, the epithelial LRRFIP2 isoform (variant 2) inhibited the oncogenic function of coactivator-associated arginine methyltransferase 1 (CARM1) through interaction. Taken together, our data reveals a mechanism of LRRFIP2 isoform switches in gastric cancer with important implication for cancer metastasis.


Asunto(s)
Neoplasias Hepáticas , Neoplasias Gástricas , Humanos , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Empalme Alternativo , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Neoplasias Hepáticas/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Neoplasias Gástricas/genética , Factores de Transcripción/metabolismo , Metástasis de la Neoplasia
6.
Nat Commun ; 13(1): 3960, 2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35803931

RESUMEN

Mesenchymal stromal cells (MSCs) differentiation into different lineages is precisely controlled by signaling pathways. Given that protein kinases play a crucial role in signal transduction, here we show that Microtubule Associated Serine/Threonine Kinase Family Member 4 (Mast4) serves as an important mediator of TGF-ß and Wnt signal transduction in regulating chondro-osteogenic differentiation of MSCs. Suppression of Mast4 by TGF-ß1 led to increased Sox9 stability by blocking Mast4-induced Sox9 serine 494 phosphorylation and subsequent proteasomal degradation, ultimately enhancing chondrogenesis of MSCs. On the other hand, Mast4 protein, which stability was enhanced by Wnt-mediated inhibition of GSK-3ß and subsequent Smurf1 recruitment, promoted ß-catenin nuclear localization and Runx2 activity, increasing osteogenesis of MSCs. Consistently, Mast4-/- mice demonstrated excessive cartilage synthesis, while exhibiting osteoporotic phenotype. Interestingly, Mast4 depletion in MSCs facilitated cartilage formation and regeneration in vivo. Altogether, our findings uncover essential roles of Mast4 in determining the fate of MSC development into cartilage or bone.


Asunto(s)
Huesos , Cartílago , Células Madre Mesenquimatosas , Proteínas Asociadas a Microtúbulos , Proteínas Serina-Treonina Quinasas , Animales , Femenino , Ratones , Huesos/citología , Huesos/metabolismo , Cartílago/citología , Cartílago/metabolismo , Diferenciación Celular/genética , Condrogénesis/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Osteogénesis/genética , Proteínas Serina-Treonina Quinasas/genética , Factor de Crecimiento Transformador beta/metabolismo , Vía de Señalización Wnt
7.
BMC Emerg Med ; 22(1): 123, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35799105

RESUMEN

PURPOSE: A multidisciplinary approach is essential for trauma patients' treatment, particularly for cases with open lower extremity fractures, which are considered major traumas requiring a comprehensive approach. Recently, the social demand for severe-trauma centers has increased. This study analyzed the clinical impact of establishing a trauma center for the treatment of open lower extremity fractures. METHODS: A retrospective chart review was conducted for trauma patients admitted to our hospital. Patients were classified into two groups: before (January 2014-December 2015, 178 patients) and after establishment of a Level-1 trauma center (January 2017-December 2018, 125 patients). We included patients with open fracture below the knee level and Gustilo type II/III, but excluded those with life-threatening trauma that affected the treatment choice. RESULTS: Total 273 patient were included in this study, initial infection was significantly more common and external fixator application significantly less in post-center establishment group. The time to emergency operation decreased significantly from 13.89 ± 17.48 to 11.65 ± 19.33 h post-center setup. By multivariate analysis, the decreased primary amputation and increased limb salvage was attributed to establishment of the trauma center. CONCLUSION: With the establishment of the Level-1 trauma center, limbs of patients with open lower extremity fractures could be salvaged, and the need for primary amputation was decreased. Early control of initial open wound infection and minimizing external fixator use allowed early soft tissue reconstruction. The existence of the center ensured a shorter interval to emergency operation and facilitated interdepartmental cooperation, which promoted active limb salvage and contributed to patients' quality of life.


Asunto(s)
Fracturas Abiertas , Centros Traumatológicos , Fracturas Abiertas/cirugía , Humanos , Extremidad Inferior/lesiones , Extremidad Inferior/cirugía , Calidad de Vida , Estudios Retrospectivos , Resultado del Tratamiento
8.
Adv Skin Wound Care ; 35(4): 234-237, 2022 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-35311768

RESUMEN

ABSTRACT: Necrotizing fasciitis is a type of soft tissue infection that destroys subcutaneous tissue. It is particularly dangerous for patients with chronic diseases and those who are bedridden while recuperating. Although necrotizing fasciitis is often caused by trauma or postoperative infection, in rare cases, it can be attributed to pressure injury (PI). The disease progression is very aggressive and can be lethal for patients who are bedridden or immunocompromised.This case report describes a 47-year-old man with a history of diabetes and hypertension who became bedridden after a sudden status decline caused by nephrotic syndrome. He gradually developed an infection and rare deterioration of a PI on his upper back. After radiologic evaluation with magnetic resonance imaging and computed tomography, surgical intervention was performed and necrotizing fasciitis was confirmed. In this case of necrotizing fasciitis derived from a PI on the upper back, the infected area spread to the periphery at a rapid rate. The infection spread over his back and across the T1-T9 levels.In this report, the authors describe the integrated system of the thoracolumbar fascia and the very aggressive spread of necrotizing fasciitis. Because of the anatomic structure of the back and the characteristics of this infection, only aggressive surgical debridement could prevent the spread of infection and reduce the systemic effects of the infection. Physicians should be aware of the possibility of PIs in bedridden patients and, in cases of exacerbation of the wound, consider rapid surgical intervention after prompt examination and diagnosis to reduce mortality.


Asunto(s)
Fascitis Necrotizante , Úlcera por Presión , Humanos , Masculino , Persona de Mediana Edad , Fascitis Necrotizante/diagnóstico , Fascitis Necrotizante/etiología , Fascitis Necrotizante/terapia , Imagen por Resonancia Magnética
9.
Cell Death Dis ; 13(2): 169, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35194034

RESUMEN

Despite favorable responses to initial chemotherapy, drug resistance is a major cause limiting chemotherapeutic efficacy in many advanced cancers. However, mechanisms that drive drug-specific resistance in chemotherapy for patients with advanced cancers are still unclear. Here, we report a unique role of death-associated protein kinase-related apoptosis-inducing kinase 1 (DRAK1) associated with paclitaxel resistance in cervical cancer cells. Interestingly, DRAK1 protein level was markedly decreased in paclitaxel-resistant cervical cancer cells without affecting its mRNA expression, which resulted in an increase in tumor necrosis factor receptor-associated factor 6 (TRAF6) expression, as well as an activation of TRAF6-mediated nuclear factor-kappa B (NF-κB) signaling cascade, thereby promoting tumor progression. DRAK1 depletion markedly increased the chemotherapeutic IC50 values of paclitaxel in cervical cancer cells. Ectopic expression of DRAK1 inhibited growth of paclitaxel-resistant cervical cancer cells in vitro and in vivo. Furthermore, DRAK1 was markedly underexpressed in chemoresistant cervical cancer patient tissues compared with chemosensitive samples. We found that DRAK1 protein was destabilized through K48-linked polyubiquitination promoted by the Cullin scaffold protein 3 (CUL3) / speckle-type POZ (poxvirus and zinc finger protein) protein (SPOP) E3 ubiquitin ligase in paclitaxel-resistant cells. Collectively, these findings suggest that DRAK1 may serve as a potential predictive biomarker for overcoming paclitaxel resistance in cervical cancer.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Proteínas Cullin , Proteínas Nucleares , Proteínas Serina-Treonina Quinasas , Proteínas Represoras , Ubiquitina-Proteína Ligasas , Neoplasias del Cuello Uterino , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Femenino , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Paclitaxel/uso terapéutico , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/genética
10.
Cancers (Basel) ; 13(18)2021 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-34572878

RESUMEN

Few studies have examined the role of BAG2 in malignancies. We investigated the prognostic value of BAG2-expression in cancer-associated fibroblasts (CAFs) and tumor cells in predicting metastasis-free survival in patients with breast cancer. Tissue-microarray was constructed using human breast cancer tissues obtained by surgical resection between 1992 and 2015. BAG2 expression was evaluated by immunohistochemistry in CAFs or the tumor cells. BAG2 expression in the CAFs and cytoplasm of tumor cells was classified as positive and negative, and low and high, respectively. BAG2-CAF was evaluated in 310 patients and was positive in 67 (21.6%) patients. Kaplan-Meier plots showed that distant metastasis-free survival (DMFS) was lesser in patients with BAG2(+) CAF than in patients with BAG2(-) CAF (p = 0.039). Additionally, we classified the 310 patients into two groups: 109 in either BAG2-high or BAG2(+) CAF and 201 in BAG2-low and BAG2(-) CAF. DMFS was significantly reduced in patients with either BAG2-high or BAG2(+) CAF than in the patients of the other group (p = 0.005). Multivariable analysis demonstrated that DMFS was prolonged in patients with BAG2(-) CAF or BAG2-low. Evaluation of BAG2 expression on both CAFs and tumor cells could help in determining the risk of metastasis in breast cancer.

11.
Biomolecules ; 11(8)2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34439774

RESUMEN

The interaction of immune checkpoint molecules in the tumor microenvironment reduces the anti-tumor immune response by suppressing the recognition of T cells to tumor cells. Immune checkpoint inhibitor (ICI) therapy is emerging as a promising therapeutic option for cancer treatment. However, modulating the immune system with ICIs still faces obstacles with severe immunogenic side effects and a lack of response against many cancer types. Plant-derived natural compounds offer regulation on various signaling cascades and have been applied for the treatment of multiple diseases, including cancer. Accumulated evidence provides the possibility of efficacy of phytochemicals in combinational with other therapeutic agents of ICIs, effectively modulating immune checkpoint-related signaling molecules. Recently, several phytochemicals have been reported to show the modulatory effects of immune checkpoints in various cancers in in vivo or in vitro models. This review summarizes druggable immune checkpoints and their regulatory factors. In addition, phytochemicals that are capable of suppressing PD-1/PD-L1 binding, the best-studied target of ICI therapy, were comprehensively summarized and classified according to chemical structure subgroups. It may help extend further research on phytochemicals as candidates of combinational adjuvants. Future clinical trials may validate the synergetic effects of preclinically investigated phytochemicals with ICI therapy.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Fitoquímicos/química , Receptor de Muerte Celular Programada 1/metabolismo , Animales , Antígenos CD/metabolismo , Antineoplásicos/farmacología , Antígenos B7/metabolismo , Antígeno B7-H1/metabolismo , Antígeno CTLA-4/metabolismo , Camptotecina/química , Diterpenos/química , Compuestos Epoxi/química , Flavonoides/química , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Humanos , Inmunoterapia , Isotiocianatos/química , Ratones , Fenantrenos/química , Fitoquímicos/farmacología , Extractos Vegetales/farmacología , Receptores Inmunológicos/metabolismo , Saponinas/química , Sulfóxidos/química , Terpenos/química , Microambiente Tumoral/efectos de los fármacos , Proteína del Gen 3 de Activación de Linfocitos
12.
Cell Death Dis ; 12(2): 159, 2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33558527

RESUMEN

Although tetraarsenic hexoxide is known to exert an anti-tumor effect by inducing apoptosis in various cancer cells, its effect on other forms of regulated cell death remains unclear. Here, we show that tetraarsenic hexoxide induces the pyroptotic cell death through activation of mitochondrial reactive oxygen species (ROS)-mediated caspase-3/gasdermin E (GSDME) pathway, thereby suppressing tumor growth and metastasis of triple-negative breast cancer (TNBC) cells. Interestingly, tetraarsenic hexoxide-treated TNBC cells exhibited specific pyroptotic characteristics, including cell swelling, balloon-like bubbling, and LDH releases through pore formation in the plasma membrane, eventually suppressing tumor formation and lung metastasis of TNBC cells. Mechanistically, tetraarsenic hexoxide markedly enhanced the production of mitochondrial ROS by inhibiting phosphorylation of mitochondrial STAT3, subsequently inducing caspase-3-dependent cleavage of GSDME, which consequently promoted pyroptotic cell death in TNBC cells. Collectively, our findings highlight tetraarsenic hexoxide-induced pyroptosis as a new therapeutic strategy that may inhibit cancer progression of TNBC cells.


Asunto(s)
Antineoplásicos/farmacología , Trióxido de Arsénico/farmacología , Caspasa 3/metabolismo , Mitocondrias/efectos de los fármacos , Piroptosis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Receptores de Estrógenos/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Caspasa 3/genética , Línea Celular Tumoral , Activación Enzimática , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Ratones Endogámicos BALB C , Mitocondrias/metabolismo , Mitocondrias/patología , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
13.
Antioxidants (Basel) ; 9(11)2020 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-33207824

RESUMEN

Cisplatin resistance remains a significant obstacle for improving the clinical outcome of ovarian cancer patients. Recent studies have demonstrated that cisplatin is an important inducer of intracellullar reactive oxygen species (ROS), triggering cancer cell death. Sirtuin 2 (SIRT2), a member of class III NAD+ dependent histone deacetylases (HDACs), has been reported to be involved in regulating cancer hallmarks including drug response. In this study, we aimed to identify the role of SIRT2 in oxidative stress and cisplatin response in cancer. Two ovarian cancer cell lines featuring different sensitivities to cisplatin were used in this study. We found different expression patterns of SIRT2 in cisplatin-sensitive (A2780/S) and cisplatin-resistant (A2780/CP) cancer cells with cisplatin treatment, where SIRT2 expression was augmented only in A2780/S cells. Furthermore, cisplatin-induced ROS generation was responsible for the upregulation of SIRT2 in A2780/S cells, whereas overexpression of SIRT2 significantly enhanced the sensitivity of cisplatin-resistant counterpart cells to cisplatin. Our study proposes that targeting SIRT2 may provide new strategies to potentiate platinum-based chemotherapy in ovarian cancer patients.

14.
Nat Commun ; 11(1): 4681, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32943626

RESUMEN

Although advanced lipidomics technology facilitates quantitation of intracellular lipid components, little is known about the regulation of lipid metabolism in cancer cells. Here, we show that disruption of the Gdpd3 gene encoding a lysophospholipase D enzyme significantly decreased self-renewal capacity in murine chronic myelogenous leukaemia (CML) stem cells in vivo. Sophisticated lipidomics analyses revealed that Gdpd3 deficiency reduced levels of certain lysophosphatidic acids (LPAs) and lipid mediators in CML cells. Loss of Gdpd3 also activated AKT/mTORC1 signalling and cell cycle progression while suppressing Foxo3a/ß-catenin interaction within CML stem cell nuclei. Strikingly, CML stem cells carrying a hypomorphic mutation of Lgr4/Gpr48, which encodes a leucine-rich repeat (LRR)-containing G-protein coupled receptor (GPCR) acting downstream of Gdpd3, displayed inadequate disease-initiating capacity in vivo. Our data showing that lysophospholipid metabolism is required for CML stem cell maintenance in vivo establish a new, biologically significant mechanism of cancer recurrence that is independent of oncogene addiction.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Células Madre/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Proteína Forkhead Box O3/metabolismo , Lisofosfolípidos/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Recurrencia Local de Neoplasia/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Receptores Acoplados a Proteínas G/genética , Transducción de Señal , beta Catenina/metabolismo
15.
Cancer Res ; 80(12): 2537-2549, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32265222

RESUMEN

The adaptor protein TNF receptor-associated factor 6 (TRAF6) is a key mediator in inflammation. However, the molecular mechanisms controlling its activity and stability in cancer progression remain unclear. Here we show that death-associated protein kinase-related apoptosis-inducing kinase 1 (DRAK1) inhibits the proinflammatory signaling pathway by targeting TRAF6 for degradation, thereby suppressing inflammatory signaling-mediated tumor growth and metastasis in advanced cervical cancer cells. DRAK1 bound directly to the TRAF domain of TRAF6, preventing its autoubiquitination by interfering with homo-oligomerization, eventually leading to autophagy-mediated degradation of TRAF6. Depletion of DRAK1 in cervical cancer cells resulted in markedly increased levels of TRAF6 protein, promoting activation of the IL1ß signaling-associated pathway and proinflammatory cytokine production. DRAK1 was specifically underexpressed in metastatic cervical cancers and inversely correlated with TRAF6 expression in mouse xenograft model tumor tissues and human cervical tumor tissues. Collectively, our findings highlight DRAK1 as a novel antagonist of inflammation targeting TRAF6 for degradation that limits inflammatory signaling-mediated progression of advanced cervical cancer. SIGNIFICANCE: Serine/threonine kinase DRAK1 serves a unique role as a novel negative regulator of the inflammatory signaling mediator TRAF6 in cervical cancer progression.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Neoplasias del Cuello Uterino/patología , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/inmunología , Ratones , Estadificación de Neoplasias , Unión Proteica/inmunología , Dominios Proteicos , Multimerización de Proteína/inmunología , Estabilidad Proteica , Proteolisis , Transducción de Señal/inmunología , Análisis de Matrices Tisulares , Ubiquitinación/inmunología , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Breast Cancer ; 23(6): 635-646, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33408889

RESUMEN

PURPOSE: Acellular dermal matrix (ADM) supports tissue expanders or implants in implant-based breast reconstruction. The characteristics of ADM tissue are defined by the manufacturing procedure, such as decellularization, preservation, and sterilization, and are directly related to clinical outcomes. This study aimed to compare the properties of a new pre-hydrated-ADM (H-ADM-low) obtained using a decellularization reagent reduction process with a low concentration of detergent with those of radiation-sterilized H-ADM and freeze-dried ADM (FD-ADM). METHODS: ADMs were evaluated in terms of structure, mechanical quality, and cytotoxicity using histochemical staining, tensile strength testing, and in vitro cell viability analysis. RESULTS: The tissue structure of H-ADM-low (CGDERM ONE-STEP) was similar to that of native skin despite complete decellularization. By contrast, in FD-ADM, the tissue structure was damaged by the freeze-drying process, and radiation-sterilized H-ADM showed a compact fibrillar arrangement. Furthermore, matrix components such as collagen and elastin were preserved in H-ADM-low, whereas a loss of elastin fibers with fragmented distribution was observed in radiation-sterilized H-ADMs. H-ADM-low's tensile strength (58.84 MPa) was significantly greater than that of FD-ADM (38.60 MPa) and comparable with that of radiation-sterilized H-ADMs. The residual detergent content in H-ADM-low (47.45 mg/L) was 2.67-fold lower than that of H-ADM decellularized with a conventional detergent concentration (126.99 mg/mL), and this finding was consistent with the cell viability results (90.7% and 70.7%, respectively), indicating that H-ADM-low has very low cytotoxicity. CONCLUSIONS: H-ADM-low produced through aseptic processes retains the original tissue structure, demonstrates excellent mechanical properties, and does not affect cell viability. Therefore, this newer H-ADM is suitable for use in implant-based breast reconstruction.

17.
Nat Commun ; 10(1): 5805, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31862882

RESUMEN

The development of triple-negative breast cancer (TNBC) negatively impacts both quality of life and survival in a high percentage of patients. Here, we show that RING finger protein 208 (RNF208) decreases the stability of soluble Vimentin protein through a polyubiquitin-mediated proteasomal degradation pathway, thereby suppressing metastasis of TNBC cells. RNF208 was significantly lower in TNBC than the luminal type, and low expression of RNF208 was strongly associated with poor clinical outcomes. Furthermore, RNF208 was induced by 17ß-estradiol (E2) treatment in an estrogen receptor alpha (ΕRα)-dependent manner. Overexpression of RNF208 suppresses tumor formation and lung metastasis of TNBC cells. Mechanistically, RNF208 specifically polyubiquitinated the Lys97 residue within the head domain of Vimentin through interaction with the Ser39 residue of phosphorylated Vimentin, which exists as a soluble form, eventually facilitating proteasomal degradation of Vimentin. Collectively, our findings define RNF208 as a negative regulator of soluble Vimentin and a prognostic biomarker for TNBC cells.


Asunto(s)
Estradiol/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias de la Mama Triple Negativas/patología , Ubiquitina-Proteína Ligasas/metabolismo , Vimentina/metabolismo , Animales , Mama/patología , Línea Celular Tumoral , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Pulmón/patología , Ratones , Pronóstico , Estabilidad Proteica , Proteolisis , Análisis de Supervivencia , Análisis de Matrices Tisulares , Neoplasias de la Mama Triple Negativas/mortalidad , Ubiquitinación , Ensayos Antitumor por Modelo de Xenoinjerto
18.
PLoS One ; 14(8): e0221721, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31449546

RESUMEN

BACKGROUND: A20 protein has ubiquitin-editing activities and acts as a key regulator of inflammation and immunity. Previously, our group showed that A20 promotes tumor metastasis through multi-monoubiquitylation of SNAIL1 in basal-like breast cancer. Here, we investigated survival outcomes in patients with breast cancer according to A20 expression. PATIENTS AND METHODS: We retrospectively collected tumor samples from patients with breast cancer. Immunohistochemistry (IHC) with an A20-specific antibody was performed, and survival outcomes were analyzed. RESULTS: A20 expression was evaluated in 442 patients. High A20 expression was associated with advanced anatomical stage and young age. High A20 expression showed significantly inferior recurrence-free-survival and overall-survival (P<0.001 and P<0.001, respectively). Multivariate analysis showed that A20 was an independent prognostic marker for RFS (HRs: 2.324, 95% CIs: 1.446-3.736) and OS (HRs: 2.629, 95% CIs: 1.585-4.361). In human epidermal growth factor receptor 2 (HER2)-positive and triple negative breast cancer (TNBC) subtypes, high A20 levels were associated with poor OS. CONCLUSION: We found that A20 expression is a poor prognostic marker in breast cancer. The prognostic impact of A20 was pronounced in aggressive tumors, such as HER2-positive and TNBC subtypes. Our findings suggested that A20 may be a valuable target in patients with aggressive breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Intervalos de Confianza , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Invasividad Neoplásica , Recurrencia Local de Neoplasia/patología , Pronóstico , Modelos de Riesgos Proporcionales , Análisis de Supervivencia , Adulto Joven
20.
J Cancer Prev ; 23(1): 1-9, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29629343

RESUMEN

BACKGROUND: Smad3 linker phosphorylation plays essential roles in tumor progression and metastasis. We have previously reported that the mutation of Smad3 linker phosphorylation sites (Smad3-Erk/Pro-directed kinase site mutant constructs [EPSM]) markedly reduced the tumor progression while increasing the lung metastasis in breast cancer. METHODS: We performed high-throughput RNA-Sequencing of the human prostate cancer cell lines infected with adenoviral Smad3-EPSM to identify the genes regulated by Smad3-EPSM. RESULTS: In this study, we identified genes which are differentially regulated in the presence of Smad3-EPSM. We first confirmed that Smad3-EPSM strongly enhanced a capability of cell motility and invasiveness as well as the expression of epithelial-mesenchymal transition marker genes, CDH2, SNAI1, and ZEB1 in response to TGF-ß1 in human pancreatic and prostate cancer cell lines. We identified GADD45B, CTGF, and JUNB genes in the expression profiles associated with cell motility and invasiveness induced by the Smad3-EPSM. CONCLUSIONS: These results suggested that inhibition of Smad3 linker phosphorylation may enhance cell motility and invasiveness by inducing expression of GADD45B, CTGF, and JUNB genes in various cancers.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...