Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
Am J Cancer Res ; 14(2): 727-743, 2024.
Article in English | MEDLINE | ID: mdl-38455399

ABSTRACT

Hypoxia-inducible factors (HIFs) regulate cellular oxygen balance and play a central role in cancer metastasis and angiogenesis. Despite extensive research on HIFs, successful therapeutic strategies remain limited due to the intricate nature of their regulation. In this study, we identified SPATA20, a relatively understudied protein with a thioredoxin-like domain, as an upstream regulator of HIF-1α. Depleting SPATA20 induced HIF-1α expression, suggesting a tumor-suppressive role for SPATA20 in cancer cells. SPATA20 depletion increased HIF-1α protein levels and transcriptional activity without affecting its degradation. It appears that SPATA20 inhibits the de novo synthesis of HIF-1α, possibly by repressing the cap-dependent translation process involving AKT phosphorylation. Additionally, depletion of SPATA20 promoted cancer cell migration and invasion, which can be reversed by pharmacological inhibition of HIF-1α. Clinical data analysis revealed an inverse correlation between SPATA20 expression and colorectal cancer progression, providing evidence of its role as a potential biomarker. Utilizing SPATA20 as an indicator for HIF-1α-targeting therapy may be an attractive strategy for treating patients with hypoxia-driven cancers. In conclusion, this study demonstrates that SPATA20 deficiency promotes cancer progression by activating the HIF-1α signaling pathway.

2.
Ann Plast Surg ; 92(2): 240-244, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38198629

ABSTRACT

PURPOSE: This study aimed to compare the antibacterial and wound healing efficacies of chitosan hydrogel with povidone-iodine (PI) hydrogel. METHODS: The in vitro antibacterial activities of chitosan and PI hydrogels against Staphylococcus aureus and Escherichia coli were evaluated. Nine 6- to 8-week-old male Sprague-Dawley rats were divided into plain, PI, and chitosan hydrogel groups. Each rat received two 10-mm full-thickness dorsal wounds using an excisional splinting model. Each wound was treated with 0.2 mL of gel thrice over the course of 3 postoperative weeks. Weekly observations were conducted, and at the end of the third postoperative week, the rats were killed for histopathological and quantitative polymerase chain reaction evaluations. Data analysis included both 2- and 1-way analyses of variance. RESULTS: Chitosan hydrogel exhibited comparable in vitro antibacterial activity and a significantly enhanced in vivo wound closure rate compared with PI hydrogel. Three weeks after the surgery, the chitosan hydrogel group demonstrated marked differences in wound repair (P < 0.01). Histologically, increased collagen deposition was observed with chitosan hydrogel treatment. Immunohistochemistry for CD68 revealed a lower number of macrophages in the wounds treated with chitosan hydrogel. Quantitative polymerase chain reaction analysis indicated a superior collagen 1 to 3 ratio and reduced expression of proinflammatory cytokine mRNAs (interleukin 1b, interleukin 6, tumor necrosis factor α, and interferon γ) in the chitosan hydrogel group. CONCLUSION: Chitosan hydrogel demonstrates the potential to serve as an effective alternative to PI hydrogel, providing enhanced wound healing capabilities while maintaining comparable antimicrobial properties.


Subject(s)
Chitosan , Hydrogels , Male , Animals , Rats , Humans , Rats, Sprague-Dawley , Hydrogels/pharmacology , Chitosan/pharmacology , Povidone-Iodine/pharmacology , Anti-Bacterial Agents/pharmacology , Wound Healing , Collagen
3.
J Control Release ; 363: 201-220, 2023 11.
Article in English | MEDLINE | ID: mdl-37739015

ABSTRACT

Extracellular vesicles (EVs) play a pivotal role in intercellular communication and have been implicated in cancer progression. Hypoxia, a pervasive hallmark of cancer, is known to regulate EV biogenesis and function. Hypoxic EVs contain a specific set of proteins, nucleic acids, lipids, and metabolites, capable of reprogramming the biology and fate of recipient cells. Enhancing the intrinsic therapeutic efficacy of EVs can be achieved by strategically modifying their structure and contents. Moreover, the use of EVs as drug delivery vehicles holds great promise for cancer treatment. However, various hurdles must be overcome to enable their clinical application as cancer therapeutics. In this review, we aim to discuss the current knowledge on the hypoxic regulation of EVs. Additionally, we will describe the underlying mechanisms by which EVs contribute to cancer progression in hypoxia and outline the progress and limitations of hypoxia-related EV therapeutics for cancer.


Subject(s)
Extracellular Vesicles , Neoplasms , Humans , Extracellular Vesicles/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Cell Communication , Drug Delivery Systems , Hypoxia/metabolism
4.
Tissue Eng Regen Med ; 20(6): 905-919, 2023 10.
Article in English | MEDLINE | ID: mdl-37531072

ABSTRACT

BACKGROUND: Adipose-derived stem cells (ADSCs) exert immunomodulatory effects in the treatment of transplant rejection. This study aimed to evaluate the effects of ADSCs on the skin graft survival in a human-to-rat xenograft transplantation model and to compare single and multiple injections of ADSCs. METHODS: Full-thickness human skin xenografts were transplanted into the backs of Sprague-Dawley rats. The rats were injected subcutaneously on postoperative days 0, 3, and 5. The injections were as follows: triple injections of phosphate-buffered saline (PBS group), a single injection of ADSCs and double injections of PBS (ADSC × 1 group), and triple injections of ADSCs (ADSC × 3 group). The immunomodulatory effects of ADSCs on human skin xenografts were assessed. RESULTS: Triple injections of ADSCs considerably delayed cell-mediated xenograft rejection compared with the PBS and ADSC × 1 groups. The vascularization and collagen type 1-3 ratios in the ADSC × 3 group were significantly higher than those in the other groups. In addition, intragraft infiltration of CD3-, CD4-, CD8-, and CD68-positive cells was reduced in the ADSC × 3 group. Furthermore, in the ADSC × 3 group, the expression levels of proinflammatory cytokine interferon-gamma (IFN-γ) were decreased and immunosuppressive prostaglandin E synthase (PGES) was increased in the xenograft and lymph node samples. CONCLUSION: This study presented that triple injections of ADSCs appeared to be superior to a single injection in suppressing cell-mediated xenograft rejection. The immunomodulatory effects of ADSCs are associated with the downregulation of IFN-γ and upregulation of PGES in skin xenografts and lymph nodes.


Subject(s)
Adipose Tissue , Graft Survival , Humans , Rats , Animals , Rats, Sprague-Dawley , Transplantation, Heterologous , Heterografts , Stem Cells
5.
Nat Commun ; 14(1): 2363, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37185280

ABSTRACT

Chronic colonic inflammation is a feature of cancer and is strongly associated with tumorigenesis, but its underlying molecular mechanisms remain poorly understood. Inflammatory conditions increased ITF2 and p65 expression both ex vivo and in vivo, and ITF2 and p65 showed positive correlations. p65 overexpression stabilized ITF2 protein levels by interfering with the binding of Parkin to ITF2. More specifically, the C-terminus of p65 binds to the N-terminus of ITF2 and inhibits ubiquitination, thereby promoting ITF2 stabilization. Parkin acts as a E3 ubiquitin ligase for ITF2 ubiquitination. Intestinal epithelial-specific deletion of ITF2 facilitated nuclear translocation of p65 and thus increased colitis-associated cancer tumorigenesis, which was mediated by Azoxymethane/Dextran sulfate sodium or dextran sulfate sodium. Upregulated ITF2 expression was lost in carcinoma tissues of colitis-associated cancer patients, whereas p65 expression much more increased in both dysplastic and carcinoma regions. Therefore, these findings indicate a critical role for ITF2 in the repression of colitis-associated cancer progression and ITF2 would be an attractive target against inflammatory diseases including colitis-associated cancer.


Subject(s)
Carcinoma , Colitis-Associated Neoplasms , Colitis , Animals , Humans , Carcinogenesis/genetics , Colitis/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Inflammation/complications , NF-kappa B/metabolism , Ubiquitin-Protein Ligases/genetics , Transcription Factor RelA
6.
Front Endocrinol (Lausanne) ; 14: 1138676, 2023.
Article in English | MEDLINE | ID: mdl-37234799

ABSTRACT

Diabetic retinopathy (DR) is a disease that causes visual deficiency owing to vascular leakage or abnormal angiogenesis. Pericyte apoptosis is considered one of the main causes of vascular leakage in diabetic retina, but there are few known therapeutic agents that prevent it. Ulmus davidiana is a safe natural product that has been used in traditional medicine and is attracting attention as a potential treatment for various diseases, but its effect on pericyte loss or vascular leakage in DR is not known at all. In the present study, we investigated on the effects of 60% edible ethanolic extract of U. davidiana (U60E) and catechin 7-O-ß-D-apiofuranoside (C7A), a compound of U. davidiana, on pericyte survival and endothelial permeability. U60E and C7A prevented pericyte apoptosis by inhibiting the activation of p38 and JNK induced by increased glucose and tumor necrosis factor alpha (TNF-α) levels in diabetic retina. Moreover, U60E and C7A reduced endothelial permeability by preventing pericyte apoptosis in co-cultures of pericytes and endothelial cells. These results suggest that U60E and C7A could be a potential therapeutic agent for reducing vascular leakage by preventing pericyte apoptosis in DR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Ulmus , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/prevention & control , Diabetic Retinopathy/pathology , Pericytes , Endothelial Cells/pathology , Apoptosis , Diabetes Mellitus/pathology
7.
Int J Mol Sci ; 24(5)2023 Feb 23.
Article in English | MEDLINE | ID: mdl-36901857

ABSTRACT

Skin cancer is one of the most prevalent cancers in the Caucasian population. In the United States, it is estimated that at least one in five people will develop skin cancer in their lifetime, leading to significant morbidity and a healthcare burden. Skin cancer mainly arises from cells in the epidermal layer of the skin, where oxygen is scarce. There are three main types of skin cancer: malignant melanoma, basal cell carcinoma, and squamous cell carcinoma. Accumulating evidence has revealed a critical role for hypoxia in the development and progression of these dermatologic malignancies. In this review, we discuss the role of hypoxia in treating and reconstructing skin cancers. We will summarize the molecular basis of hypoxia signaling pathways in relation to the major genetic variations of skin cancer.


Subject(s)
Carcinoma, Basal Cell , Carcinoma, Squamous Cell , Melanoma , Skin Neoplasms , Humans , United States , Skin Neoplasms/pathology , Melanoma/metabolism , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/pathology , Hypoxia
8.
Transplant Proc ; 54(8): 2388-2395, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36184343

ABSTRACT

Adipose-derived stem cells (ASCs) have demonstrated immunomodulatory and anti-inflammatory effects in preclinical studies. The purpose of this study was to evaluate the effects of ASCs on the survival of xenogeneic full-thickness skin grafts and compare intravenous and subcutaneous injections of ASCs. We divided 30 male C57BL/6 mice into control, intravenous (IV), and subcutaneous (SC) injection groups. In one group of 10 mice, mouse ASCs were intravenously injected after human full-thickness skin grafting (IV group). In another group of 10 mice, ASCs were directly injected into the subcutaneous plane under the xenogeneic grafts (SC group). An additional group of 10 mice received no treatment and served as controls. Bioluminescent imaging showed that ASCs were concentrated at the grafts during the study period in both IV and SC groups. We performed graft survival assessment, histologic examination, and immunohistochemistry analysis. ASCs significantly prolonged xenograft survival at postoperative week 2 in the SC group compared with the control group (P < .05). Histologic evaluation revealed fewer inflammatory reactions in the SC group than in the control group at 1 week posttransplantation. In addition, we observed relative reduction in CD4- and CD8-positive cells in the SC group compared with the control group. Intravenous injection of ASCs led to increased graft survival and decreased inflammatory reactions, but these differences were not statistically significant. The results of this study indicate that subcutaneous injection of ASCs promoted the survival of xenogeneic full-thickness skin grafts in mice. The underlying mechanisms of the immunosuppressive effects of ASCs should be further investigated.


Subject(s)
Adipocytes , Adipose Tissue , Humans , Male , Mice , Animals , Transplantation, Heterologous , Mice, Inbred C57BL , Stem Cells , Disease Models, Animal , Stem Cell Transplantation
9.
Cancers (Basel) ; 14(14)2022 Jul 07.
Article in English | MEDLINE | ID: mdl-35884382

ABSTRACT

Solid cancers are composed of malignant cells and their surrounding matrix components. Hypoxia plays a critical role in shaping the tumor microenvironment that contributes to cancer progression and treatment failure. Cancer-associated fibroblasts (CAFs) are one of the most prominent components of the tumor microenvironment. CAFs are highly sensitive to hypoxia and participates in the crosstalk with cancer cells. Hypoxic CAFs modulate several mechanisms that induce cancer malignancy, such as extracellular matrix (ECM) remodeling, immune evasion, metabolic reprogramming, angiogenesis, metastasis, and drug resistance. Key signaling molecules regulating CAFs in hypoxia include transforming growth factor (TGF-ß) and hypoxia-inducible factors (HIFs). In this article, we summarize the mechanisms underlying the hypoxic regulation of CAFs and how hypoxic CAFs affect cancer development and progression. We also discuss the potential therapeutic strategies focused on targeting CAFs in the hypoxic tumor microenvironment.

10.
Exp Mol Med ; 54(6): 720-729, 2022 06.
Article in English | MEDLINE | ID: mdl-35764882

ABSTRACT

Surgery is unanimously regarded as the primary strategy to cure solid tumors in the early stages but is not always used in advanced cases. However, tumor surgery must be carefully considered because the risk of metastasis could be increased by the surgical procedure. Tumor surgery may result in a deep wound, which induces many biological responses favoring tumor metastasis. In particular, NETosis, which is the process of forming neutrophil extracellular traps (NETs), has received attention as a risk factor for surgery-induced metastasis. To reduce cancer mortality, researchers have made efforts to prevent secondary metastasis after resection of the primary tumor. From this point of view, a better understanding of surgery-induced metastasis might provide new strategies for more effective and safer surgical approaches. In this paper, recent insights into the surgical effects on metastasis will be reviewed. Moreover, in-depth opinions about the effects of NETs on metastasis will be discussed.


Subject(s)
Extracellular Traps , Neoplasms , Humans , Neoplasms/pathology , Neoplasms/surgery , Neutrophils/pathology , Risk Factors , Tumor Microenvironment
11.
BMB Rep ; 55(6): 287-292, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35410638

ABSTRACT

The acute response to hypoxia is mainly driven by hypoxiainducible factors, but their effects gradually subside with time. Hypoxia-specific histone modifications may be important for the stable maintenance of long-term adaptation to hypoxia. However, little is known about the molecular mechanisms underlying the dynamic alterations of histones under hypoxic conditions. We found that the phosphorylation of histone H3 at Ser-10 (H3S10) was noticeably attenuated after hypoxic challenge, which was mediated by the inhibition of aurora kinase B (AURKB). To understand the role of AURKB in epigenetic regulation, DNA microarray and transcription factor binding site analyses combined with proteomics analysis were performed. Under normoxia, phosphorylated AURKB, in concert with the repressor element-1 silencing transcription factor (REST), phosphorylates H3S10, which allows the AURKB-REST complex to access the MDM2 proto-oncogene. REST then acts as a transcriptional repressor of MDM2 and downregulates its expression. Under hypoxia, AURKB is dephosphorylated and the AURKB-REST complex fails to access MDM2, leading to the upregulation of its expression. In this study, we present a case of hypoxia-specific epigenetic regulation of the oxygen-sensitive AURKB signaling pathway. To better understand the cellular adaptation to hypoxia, it is worthwhile to further investigate the epigenetic regulation of genes under hypoxic conditions. [BMB Reports 2022; 55(6): 287-292].


Subject(s)
Oxygen , Transcription Factors , Aurora Kinase B/metabolism , Epigenesis, Genetic , Histones/metabolism , Humans , Hypoxia/genetics , Oxygen/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Transcription Factors/metabolism
12.
J Allergy Clin Immunol ; 149(1): 340-357, 2022 01.
Article in English | MEDLINE | ID: mdl-33957165

ABSTRACT

BACKGROUND: Diesel exhaust particles (DEPs) are associated with the prevalence and exacerbation of allergic respiratory diseases, including allergic rhinitis and allergic asthma. However, DEP-induced mechanistic pathways promoting upper airway disease and their clinical implications remain unclear. OBJECTIVE: We sought to investigate the mechanisms by which DEP exposure contributes to nasal polyposis using human-derived epithelial cells and a murine nasal polyp (NP) model. METHODS: Gene set enrichment and weighted gene coexpression network analyses were performed. Cytotoxicity, epithelial-to-mesenchymal transition (EMT) markers, and nasal polyposis were assessed. Effects of DEP exposure on EMT were determined using epithelial cells from normal people or patients with chronic rhinosinusitis with or without NPs. BALB/c mice were exposed to DEP through either a nose-only exposure system or nasal instillation, with or without house dust mite, followed by zinc finger E-box-binding homeobox (ZEB)2 small hairpin RNA delivery. RESULTS: Bioinformatics analyses revealed that DEP exposure triggered EMT features in airway epithelial cells. Similarly, DEP-exposed human nasal epithelial cells exhibited EMT characteristics, which were dependent on ZEB2 expression. Human nasal epithelial cells derived from patients with chronic rhinosinusitis presented more prominent EMT features after DEP treatment, when compared with those from control subjects and patients with NPs. Coexposure to DEP and house dust mite synergistically increased the number of NPs, epithelial disruptions, and ZEB2 expression. Most importantly, ZEB2 inhibition prevented DEP-induced EMT, thereby alleviating NP formation in mice. CONCLUSIONS: Our data show that DEP facilitated NP formation, possibly via the promotion of ZEB2-induced EMT. ZEB2 may be a therapeutic target for DEP-induced epithelial damage and related airway diseases, including NPs.


Subject(s)
Air Pollutants/toxicity , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Nasal Polyps , Vehicle Emissions/toxicity , Zinc Finger E-box Binding Homeobox 2/genetics , Adult , Aged , Allergens/administration & dosage , Animals , Cell Movement/drug effects , Cells, Cultured , Chronic Disease , Epithelial Cells/physiology , Female , Humans , Male , Mice, Inbred BALB C , Middle Aged , Nasal Polyps/genetics , Pyroglyphidae/immunology , RNA, Small Interfering/administration & dosage , Rhinitis/genetics , Sinusitis/genetics , Young Adult
13.
Nat Commun ; 12(1): 4288, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34257283

ABSTRACT

The commonly mutated human KRAS oncogene encodes two distinct KRAS4A and KRAS4B proteins generated by differential splicing. We demonstrate here that coordinated regulation of both isoforms through control of splicing is essential for development of Kras mutant tumors. The minor KRAS4A isoform is enriched in cancer stem-like cells, where it responds to hypoxia, while the major KRAS4B is induced by ER stress. KRAS4A splicing is controlled by the DCAF15/RBM39 pathway, and deletion of KRAS4A or pharmacological inhibition of RBM39 using Indisulam leads to inhibition of cancer stem cells. Our data identify existing clinical drugs that target KRAS4A splicing, and suggest that levels of the minor KRAS4A isoform in human tumors can be a biomarker of sensitivity to some existing cancer therapeutics.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , RNA-Binding Proteins/metabolism , A549 Cells , Animals , Blotting, Western , Cell Proliferation , Flow Cytometry , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Polymerase Chain Reaction , Proto-Oncogene Proteins p21(ras)/genetics , RNA-Binding Proteins/genetics
14.
Oncogene ; 39(36): 5839-5854, 2020 09.
Article in English | MEDLINE | ID: mdl-32796959

ABSTRACT

Identification of pro-metastatic genomic alterations is urgently needed to help understand and prevent the fatal course of prostate cancer. Here, we found that the transcription factor EGR3, located at chromosome 8p21.3, is a critical metastasis suppressor. Aberrant deletion of EGR3 was found in up to 59.76% (deep deletions, 16.87%; shallow deletions, 42.89%) of prostate cancer patients. In informatics analysis, EGR3 loss was associated with prostate cancer progression and low survival rates. EGR3 expression inversely correlated with the expressions of epithelial-to-mesenchymal transition (EMT) and metastasis-related gene sets in prostate cancer tissues. In prostate cancer cells, EGR3 blocked the EMT process and suppressed cell migration and invasion. In a mouse model for cancer metastasis, EGR3 overexpression significantly suppressed bone metastases of PC3 and 22Rv1 prostate cancer cells. Mechanistically, EGR3 transcriptionally activated ZFP36, GADD45B, and SOCS3 genes by directly binding to their promoter regions. The EMT-inhibitory and tumor-suppressive roles of the EGR3 downstream genes were identified through in vitro and in silico analyses. Together, our results showed that EGR3 may be a biomarker to predict clinical outcomes and that it plays an important role in the metastatic progression of prostate cancer.


Subject(s)
Early Growth Response Protein 3/deficiency , Genetic Predisposition to Disease , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Movement , DNA Copy Number Variations , Disease Models, Animal , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Follow-Up Studies , Gene Deletion , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Humans , Male , Mice , Neoplasm Metastasis , Prognosis , Proportional Hazards Models , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Risk Factors , Xenograft Model Antitumor Assays
15.
Cancer Lett ; 489: 41-49, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32522693

ABSTRACT

The mechanism underlying hypoxia-driven chromatin remodeling is a long-lasting question. For the last two decades, this question has been resolved in part. It is now widely agreed that hypoxia dynamically changes the methylation status of histones to control gene expression. Hypoxia-inducible factor (HIF) plays a central role in cellular responses to hypoxia through transcriptional activation of numerous genes. At least in part, the hypoxic regulation of histone methylation is attributed to the HIF-mediated expression of histone modifying enzymes. Protein hydroxylation and histone demethylation have emerged as the oxygen sensing processes because they are catalyzed by a family of 2-oxoglutarate (2OG)-dependent dioxygenases whose activities depend upon the ambient oxygen level. Recently, it has been extensively investigated that the 2OG dioxygenases oxygen-dependently regulate histone methylation. Nowadays, the hypoxic change in the histone methylation status is regarded as an important event to drive malignant behaviors of cancer cells. In this review, we introduced and summarized the cellular processes that govern hypoxia-driven regulation of histone methylation in the context of cancer biology. We also discussed the emerging roles of histone methyltransferases and demethylases in epigenetic response to hypoxia.


Subject(s)
Cell Hypoxia/genetics , DNA Methylation/genetics , Gene Expression Regulation, Neoplastic/genetics , Histones/genetics , Neoplasms/pathology , Animals , Disease Progression , Humans , Neoplasms/genetics
16.
Sci Rep ; 9(1): 10599, 2019 07 22.
Article in English | MEDLINE | ID: mdl-31332228

ABSTRACT

The transcription factor HIF-1 induces the expression of genes that are essential for cell survival and oxygen homeostasis in hypoxic conditions. The prolyl isomerase Pin1 plays a role in the regulation of HIF-1α. However, the mechanism by which Pin1 controls HIF-1α remains controversial. Surprisingly, we here show that a PIN1 transcript downregulates HIF-1α as a long non-coding RNA. Pin1-silencing siRNAs augmented the hypoxia-induced expression of HIF-1α, thereby upregulating the expression of HIF-1 target genes. However, the overexpression of Pin1 protein did not inhibit the hypoxic expression of HIF-1α. Pin1 restoration in Pin1-depleted cells also failed to reverse the induction of HIF-1α by Pin1 knockdown. Unexpectedly, HIF-1α was found to be induced by both siRNAs for PIN1 transcript variants 1/2 and that for PIN1 transcript variants 2/3, indicating that the PIN1 transcript variant 2 (PIN1-v2) is responsible for HIF-1α induction. Mechanistically, PIN1-v2, which is classified as a long non-coding RNA due to early termination of translation, was evaluated to inhibit the transcription of HIF1A gene. In conclusion, PIN1-v2 may function in balancing the HIF-1-driven gene expression under hypoxia.


Subject(s)
Hypoxia-Inducible Factor 1/metabolism , Hypoxia/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , RNA, Long Noncoding/metabolism , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , Humans , Hypoxia-Inducible Factor 1/physiology , NIMA-Interacting Peptidylprolyl Isomerase/physiology , RNA, Long Noncoding/physiology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
17.
J Thorac Oncol ; 14(7): 1223-1232, 2019 07.
Article in English | MEDLINE | ID: mdl-30959120

ABSTRACT

INTRODUCTION: Despite adoption of molecular biomarkers in the management of NSCLC, the recently adopted eighth edition of the TNM staging system utilized only clinicopathologic characteristics and validated improvement in risk stratification of early-stage disease has remained elusive. We therefore evaluated the integration of a clinically validated molecular prognostic classifier into conventional staging. METHODS: A novel staging system, the TNMB (with the B denoting biology) system, which integrates a 14-gene molecular prognostic classifier into the eighth edition of the TNM staging system, was developed by using data from 321 patients with NSCLC at the University of California, San Francisco. The TNMB staging system was subsequently validated in an independent, multicenter cohort of 1373 patients, and its implementation was compared with adoption of the seventh and eighth edition staging systems utilizing metrics of reclassification. RESULTS: Compared with staging according to the eighth edition of the TNM system, the TNMB staging system enhanced the identification of high-risk patients, with a net reclassification improvement of 0.33 (95% confidence interval [CI]: 0.24-0.41). It better predicted differences in survival, with a relative integrated discrimination improvement of 22.1% (95% CI: 8.8%-35.3%), and it improved agreement between observed and predicted survival, with a decrease in the reclassification calibration statistic of from 39 to 21. The seventh and eighth editions failed to change the net reclassification improvement (0.01 [95% CI: -0.04 to 0.03] and 0.03 [95% CI: 0.00 to 0.06], respectively) or relative integrated discrimination improvement (2.1% [95% CI: -5.8 to 9.9] and -2.5% [95% CI: -17.6 to 12.4], respectively); in addition, the eighth edition worsened calibration, with an increase in the reclassification calibration statistic from 23 to 25. CONCLUSIONS: Incorporation of a molecular prognostic classifier significantly improved identification of high-risk patients and survival predictions compared with when conventional staging is used. The TNMB staging system may lead to improved survival of early-stage disease through more effective application of adjuvant therapy.


Subject(s)
Adenocarcinoma of Lung/pathology , Biomarkers, Tumor/genetics , Carcinoma, Large Cell/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/pathology , Lung Neoplasms/pathology , Mutation , Adenocarcinoma of Lung/classification , Adenocarcinoma of Lung/genetics , Aged , Carcinoma, Large Cell/classification , Carcinoma, Large Cell/genetics , Carcinoma, Non-Small-Cell Lung/classification , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/classification , Carcinoma, Squamous Cell/genetics , Female , Follow-Up Studies , Humans , Lung Neoplasms/classification , Lung Neoplasms/genetics , Male , Neoplasm Staging , Retrospective Studies , Survival Rate
18.
Oncogene ; 38(19): 3651-3666, 2019 05.
Article in English | MEDLINE | ID: mdl-30659265

ABSTRACT

Oxygen is an indispensable element for cell survival and maintenance. Eukaryotic cells are equipped with a series of signaling pathways that cope with hypoxia. The dioxygenase factor inhibiting HIF (FIH) is an oxygen sensor that regulates the transcriptional activity of hypoxia-inducible factor (HIF) through asparaginyl hydroxylation. Given that HACE1 was detected as an FIH-interacting protein in a previous proteomics study, we tested whether the E3 ubiquitin ligase HACE1 is a substrate for FIH. FIH interacted with HACE1, in cells and in vitro, and was determined to hydroxylate HACE1 at the N191 residue within the ankyrin repeat domain. Hydroxylation disrupted the physical association between HACE1 and its representative target, Rac1. Under hypoxic conditions, HACE1 is less hydroxylated due to the inactivation of FIH, and subsequently functions to ubiquitinate the active form of Rac1, leading to the proteasomal degradation of Rac1. Since Rac1 stimulates cell movement, HACE1 inhibits cell migration and invasion in breast cancer by removing active Rac1. Such an effect of HACE1 is reinforced under hypoxia because HACE1 escapes from FIH-mediated hydroxylation. In clinical datasets, HACE1 downregulation is associated with poor outcomes in patients with breast cancer. Taken together, FIH is likely to act as an oxygen sensor that determines oxygen-dependent cancer progression.


Subject(s)
Breast Neoplasms/pathology , Mixed Function Oxygenases/metabolism , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , rac1 GTP-Binding Protein/metabolism , Ankyrin Repeat , Asparagine/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Female , Humans , Hydroxylation , Oxygen/metabolism , Tumor Hypoxia , Ubiquitin-Protein Ligases/genetics , Ubiquitination , rac1 GTP-Binding Protein/genetics
19.
Cell Biol Int ; 42(3): 303-312, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29047187

ABSTRACT

The KN motif and ankyrin repeat domain-containing protein (KANK) family is involved in actin cytoskeleton organization and cell motility. Compared with other KANK members, the biological function of KANK3 is not clear. Here, we identified KANK3 as a new substrate for the oxygen sensor hypoxia-inducible factor 1-alpha inhibitor (HIF1AN), which hydroxylates HIF-1/2α and other ankyrin repeat domain-containing proteins at asparagine residues. An in vitro hydroxylation assay clearly demonstrated asparaginyl hydroxylation of KANK3 by HIF1AN, and mass spectroscopic analysis revealed that KANK3 is hydroxylated at three asparagine residues within the ankyrin repeat domain. Bioinformatics analysis revealed that KANK3 downregulation is correlated with a poor prognosis in several types of cancers, including hepatocellular carcinoma (HCC). In HCC cells, KANK3 knockdown enhanced cell migration and invasion, while its overexpression inhibited these cell behaviors. Interestingly, such effects of KANK3 were not observed under hypoxic conditions, suggesting oxygen-dependent activity of KANK3. Based on these data, we propose that KANK3 acts as a tumor suppressor to control cancer behavior in an oxygen-dependent manner.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carrier Proteins/metabolism , Liver Neoplasms/metabolism , Mixed Function Oxygenases/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Ankyrin Repeat , Asparagine/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carrier Proteins/genetics , Cell Hypoxia/physiology , Cell Line, Tumor , Down-Regulation , Genes, Tumor Suppressor , HEK293 Cells , Hep G2 Cells , Humans , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mixed Function Oxygenases/genetics , Oxygen/metabolism , Repressor Proteins/genetics
20.
Oncotarget ; 8(61): 103744-103757, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29262597

ABSTRACT

The mechanism of Sonic Hedgehog (Shh) pathway activation in non-small cell lung cancer (NSCLC) is poorly described. Using an antibody against the Shh C-terminal domain, we found a small population of Shh-positive (Shh+) cells in NSCLC cells. The objective of this study was to characterize these Shh+ cells. Shh+ and Shh- cells were sorted by using Fluorescence Activated Cell Sorting (FACS) on 12 commercial NSCLC cell lines. Functional analyses on sorted cells were performed with gene expression assays (qRT-PCR and microarray) and cells were treated with cytotoxic chemotherapy and a targeted inhibitor of Shh signaling (GDC0449). We used in vivo models of nude mice inoculated with Shh+ and Shh- sorted cells and drug-treated cells. Finally, we confirmed our results in fresh human NSCLC samples (n=48) paired with normal lung tissue. We found that Shh+ cells produced an uncleaved, full-length Shh protein detected on the membranes of these cells. Shh+ cells exerted a paracrine effect on Shh- cells, inducing their proliferation and migration. Shh+ cells were chemo-resistant and showed features of cancer stem cells (CSCs) in vitro and in vivo. Pharmacological inhibition of the Shh pathway suppressed their CSC features. A high percentage of Shh+ cells was associated with poor prognosis in early-stage NSCLC patients. In conclusion, we describe for the first time the presence of an abnormal membrane-bound full-length Shh protein in human cancer cells that allows the identification of CSCs in vitro and in vivo.

SELECTION OF CITATIONS
SEARCH DETAIL
...