Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(7)2023 Mar 24.
Article in English | MEDLINE | ID: mdl-37047130

ABSTRACT

Anti-pigmentation peptides have been developed as alternative skin-lightening agents to replace conventional chemicals that have adverse effects on the skin. However, the maximum size of these peptides is often limited by their low skin and cell penetration. To address this issue, we used our intra-dermal delivery technology (IDDT) platform to identify peptides with hypo-pigmenting and high cell-penetrating activity. Using our cell-penetrating peptides (CPPs) from the IDDT platform, we identified RMNE1 and its derivative RMNE3, "DualPep-Shine", which showed levels of α-Melanocyte stimulating hormone (α-MSH)-induced melanin inhibition comparable to the conventional tyrosinase inhibitor, Kojic acid. In addition, DualPep-Shine was delivered into the nucleus and regulated the gene expression levels of melanogenic enzymes by inhibiting the promoter activity of microphthalmia-associated transcription factor-M (MITF-M). Using a 3D human skin model, we found that DualPep-Shine penetrated the lower region of the epidermis and reduced the melanin content in a dose-dependent manner. Furthermore, DualPep-Shine showed high safety with little immunogenicity, indicating its potential as a novel cosmeceutical ingredient and anti-pigmentation therapeutic agent.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Cell-Penetrating Peptides , Melanins , Melanocytes , Microphthalmia-Associated Transcription Factor , Nerve Tissue Proteins , Skin Lightening Preparations , Skin Pigmentation , Transcription, Genetic , Melanins/antagonists & inhibitors , Skin Pigmentation/drug effects , Microphthalmia-Associated Transcription Factor/genetics , Transcription, Genetic/drug effects , alpha-MSH/antagonists & inhibitors , alpha-MSH/metabolism , Humans , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Skin Lightening Preparations/chemistry , Skin Lightening Preparations/pharmacology , Melanoma, Experimental , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/pharmacology , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Keratinocytes/drug effects , Keratinocytes/metabolism , Melanocytes/drug effects , Melanocytes/metabolism , Epidermis/drug effects , Epidermis/metabolism
2.
J Physiol Biochem ; 79(1): 83-105, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36194366

ABSTRACT

Long noncoding RNAs (lncRNAs) are emerging regulators of vascular diseases, yet their role in diabetic vascular calcification/aging remains poorly understood. In this study, we identified a down-expressed lncRNA SNHG1 in high glucose (HG)-induced vascular smooth muscle cells (HA-VSMCs), which induced excessive autophagy and promoted HA-VSMCs calcification/senescence. Overexpression of SNHG1 alleviated HG-induced HA-VSMCs calcification/senescence. The molecular mechanisms of SNHG1 in HA-VSMCs calcification/senescence were explored by RNA pull-down, RNA immunoprecipitation, RNA stability assay, luciferase reporter assay, immunoprecipitation and Western blot assays. In one mechanism, SNHG1 directly interacted with Bhlhe40 mRNA 3'-untranslated region and increased Bhlhe40 mRNA stability and expression. In another mechanism, SNHG1 enhanced Bhlhe40 protein SUMOylation by serving as a scaffold to facilitate the binding of SUMO E3 ligase PIAS3 and Bhlhe40 protein, resulting in increased nuclear translocation of Bhlhe40 protein. Moreover, Bhlhe40 suppressed the expression of Atg10, which is involved in the process of autophagosome formation. Collectively, the protective effect of SNHG1 on HG-induced HA-VSMCs calcification/senescence is accomplished by stabilizing Bhlhe40 mRNA and promoting the nuclear translocation of Bhlhe40 protein. Our study could provide a novel approach for diabetic vascular calcification/aging.


Subject(s)
Autophagy-Related Proteins , Basic Helix-Loop-Helix Transcription Factors , MicroRNAs , RNA, Long Noncoding , Vascular Calcification , Humans , Autophagy , Autophagy-Related Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Glucose/metabolism , Homeodomain Proteins , MicroRNAs/metabolism , Molecular Chaperones/metabolism , Molecular Chaperones/pharmacology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Inhibitors of Activated STAT/metabolism , Protein Inhibitors of Activated STAT/pharmacology , RNA, Long Noncoding/metabolism
3.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 30(5): 1435-1439, 2022 Oct.
Article in Chinese | MEDLINE | ID: mdl-36208246

ABSTRACT

OBJECTIVE: To observe the mutation and expression of TCF3 gene in Burkitt's lymphoma (BL), and explore its effect on the proliferation of BL cells and clinical efficacy and prognosis. METHODS: The mutation and expression of TCF3 in tumor tissues from BL patients were observed by the second-generation sequencing and real-time quantitative PCR. The proliferation and apoptosis of lymphoma cells after TCF3 knocked down were observed by siRNA interference technique and CCK-8 method. Survival analysis was used to observe the relationship between TCF3 mutation and the treatment efficacy and prognosis of BL patients. RESULTS: There were high frequency mutation rate (mutation rate was 23.7%) and high expression of TCF3 in BL patients. After TCF3 knocked down, cell proliferation was inhibited and apoptosis was promoted. In TCF3-siRNA group and control group, the cell proliferation rate at 48 h was (50.2±5.9)% and (96.6±11.4)%, and apoptosis rate was 30.1% and 1.5%, respectively, which showed significantly different between the two groups (P<0.001, P=0.005). The complete remission rate of patients with TCF3 mutation was low. The complete remission rate of mutant group and wild-type group was 44.4% and 82.8%, respectively (P=0.023). The 2-year progression-free survival rate and overall survival rate of the patients with TCF3 mutation was 55.6% and 61.0%, respectively, which was lower than 83.2% and 85.2% of the patients without mutation, but the differences were not statistically significant. CONCLUSION: There are mutation and abnormal expression of TCF3 in patients with BL. Patients with TCF3 mutations have low remission rate and poor prognosis.


Subject(s)
Burkitt Lymphoma , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Basic Helix-Loop-Helix Transcription Factors/therapeutic use , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Humans , Prognosis , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use
4.
J Food Biochem ; 46(12): e14456, 2022 12.
Article in English | MEDLINE | ID: mdl-36226991

ABSTRACT

Saponins from bitter melon (BMS) exert potential bioactivities and pharmacological activities, including anti-oxidation and lifespan extension. However, the exact mechanisms of BMS in response to oxidative stress remain unknown. Results demonstrated that bitter melon saponins could strengthen locomotive activities (body bend and head thrashing) accompanied by delaying the muscle fiber damage with age in Caenorhabditis elegans. In addition, BMS inhibited the ROS accumulation, improved the activities of antioxidant enzymes like SOD (by 57.90% and 94.34% for 100 µg/ml and 200 µg/ml BMS, respectively) and CAT (by 51.45% and 56.91% for 100 µg/ml and 200 µg/ml BMS, respectively), and extend the lifespan of N2 and CL2006 worms under paraquat-induced oxidative stress. Mechanism study suggested that BMS modulated the mRNA expressions of oxidation-related regulators, like the upregulation of cat-1, hsf-1, sir-2.1, and hlh-30. Furthermore, gene-deficient mutants verified that IIS (insulin/insulin-like growth factor-1 signaling) pathway linked with sir-2.1 and hlh-30 factors were involved in the BMS's lifespan-extension effects under oxidative stress. In general, this study supplemented the explanation of BMS in promoting oxidation-resistance and lifespan-extension activities, which could be served as a potential candidate for anti-aging. PRACTICAL APPLICATIONS: Our previous studies have suggested that saponins from bitter melon exhibited fat-lowering activity in C. elegans. However, little was known about the mechanism underlying the anti-oxidation effects of BMS in C. elegans. Current results indicated that the IIS pathway linked with sir-2.1 and hlh-30 transcriptional factors jointly to increase the lifespan in BMS' responses to oxidative stress. Our findings are beneficial to understand the main nutritional ingredients in bitter melon, which are ideal and expected in functional foods for aging.


Subject(s)
Caenorhabditis elegans Proteins , Momordica charantia , Saponins , Sirtuins , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Saponins/pharmacology , Oxidative Stress , Aging , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Sirtuins/metabolism , Sirtuins/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/pharmacology
5.
Bioengineered ; 13(4): 10827-10842, 2022 04.
Article in English | MEDLINE | ID: mdl-35470736

ABSTRACT

The effect of long intergenic noncoding RNA 01315 (LINC01315) on colorectal cancer has widely been proved. Nevertheless, how LINC01315 functions in the stemness of colorectal cancer and whether LINC01315 exists in colorectal cancer stem-like cell-derived exosomes remain dim, which are thus investigated in this research. CD133+/CD44+ colorectal cancer stem cells were sorted and verified through flow cytometry. Exosomes derived from CD133+/CD44+ colorectal cancer stem cells were collected. The viability, proliferation, stemness and migration of CD133+/CD44+, CD133-/CD44-, and colorectal cancer cells after transfection or the co-culture with exosomes were detected by MTT, colony formation, spheroid, and wound healing assays, respectively. Expressions of LINC01315, BCL-2, Bax, cleaved caspase-3, MMP-9, E-cadherin, and vimentin in cells or exosomes were analyzed using western blot or qRT-PCR. Genes interacted with LINC01315 in colorectal cancer were predicted by bioinformatics analysis. The results showed that LINC01315 was high-expressed in CD133+/CD44+ colorectal cancer stem cells and exosomes. Compared with colorectal cancer cells, the viability, proliferation, stemness, and migration of CD133+/CD44+ cancer cells were stronger, while these of CD133-/CD44- cancer cells were weaker. Besides, LINC01315 silencing decreased the viability, proliferation, stemness, and migration of CD133+/CD44+ cancer cells, while sh-LINC01315 inhibited the promotive effects of CD133+/CD44+ cancer cell-derived exosomes on the viability, proliferation, stemness, and migration of colorectal cancer cells. LINC01315 was also found to be correlated with DPEP1, KRT23, ASCL2, AXIN2, and DUSP4 in colorectal cancer. In conclusion, colorectal cancer stem cell-derived exosomal LINC01315 promotes the proliferation, migration, and stemness of colorectal cancer cells.


Subject(s)
Colorectal Neoplasms , RNA, Long Noncoding , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Humans , Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/metabolism
6.
Eur J Neurosci ; 55(3): 675-693, 2022 02.
Article in English | MEDLINE | ID: mdl-35001440

ABSTRACT

Substance use disorders are associated with disruptions to both circadian rhythms and cellular metabolic state. At the molecular level, the circadian molecular clock and cellular metabolic state may be interconnected through interactions with the nicotinamide adenine dinucleotide (NAD+ )-dependent deacetylase, sirtuin 1 (SIRT1). In the nucleus accumbens (NAc), a region important for reward, both SIRT1 and the circadian transcription factor neuronal PAS domain protein 2 (NPAS2) are highly enriched, and both are regulated by the metabolic cofactor NAD+ . Substances of abuse, like cocaine, greatly disrupt cellular metabolism and promote oxidative stress; however, their effects on NAD+ in the brain remain unclear. Interestingly, cocaine also induces NAc expression of both NPAS2 and SIRT1, and both have independently been shown to regulate cocaine reward in mice. However, whether NPAS2 and SIRT1 interact in the NAc and/or whether together they regulate reward is unknown. Here, we demonstrate diurnal expression of Npas2, Sirt1 and NAD+ in the NAc, which is altered by cocaine-induced upregulation. Additionally, co-immunoprecipitation reveals NPAS2 and SIRT1 interact in the NAc, and cross-analysis of NPAS2 and SIRT1 chromatin immunoprecipitation sequencing reveals several reward-relevant and metabolic-related pathways enriched among shared gene targets. Notably, NAc-specific Npas2 knock-down or a functional Npas2 mutation in mice attenuates SIRT1-mediated increases in cocaine preference. Together, our data reveal an interaction between NPAS2 and SIRT1 in the NAc, which may serve to integrate cocaine's effects on circadian and metabolic factors, leading to regulation of drug reward.


Subject(s)
Cocaine , Nucleus Accumbens , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Circadian Rhythm/physiology , Cocaine/pharmacology , Mice , Mice, Inbred C57BL , NAD/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Reward , Sirtuin 1/genetics , Sirtuin 1/metabolism , Transcription Factors/metabolism
7.
Cancer Biol Med ; 19(6)2021 12 22.
Article in English | MEDLINE | ID: mdl-34931765

ABSTRACT

OBJECTIVE: In various cancers, migration and invasion inhibitory protein (MIIP) is expressed at low level and is involved in cancer pathogenesis. Herein, we sought to explore the function of MIIP in clear cell renal cell carcinoma (ccRCC). METHODS: CCK-8, colony formation, cell cycle, and endothelial cell tube formation assays were performed to evaluate the roles of MIIP in ccRCC proliferation and angiogenesis. To explore the underlying mechanism, we conducted RNA-sequencing, GSEA, qRT-PCR, Western blot, ELISA, cell transfection, coimmunoprecipitation, and ubiquitination assays in ccRCC cell lines. Furthermore, xenograft tumor growth in nude mice, and Ki-67 and CD31 staining in xenograft tissues were examined. Finally, the association of MIIP expression with clinical pathology and the expression status of HIF-2α and cysteine-rich 61 (CYR61) were further analyzed in human RCC tissues through Western blot and immunohistochemistry. RESULTS: Both in vitro and in vivo functional experiments indicated that forced expression of MIIP inhibited ccRCC proliferation and angiogenesis, whereas silencing MIIP either in normal HK-2 cells or in ccRCC cells had the opposite effect (P < 0.05). Mechanistically, CYR61 was identified as a gene significantly downregulated by MIIP overexpression, and was required for the suppressive role of MIIP in ccRCC. MIIP was found to promote HSP90 acetylation and thus impair its chaperone function toward HIF-2α. Consequently, RACK1 binds HIF-2α and causes its ubiquitination and proteasomal degradation, thus decreasing the transcription of its target, CYR61. Finally, analyses of clinical samples demonstrated that MIIP is significantly downregulated in cancer vs. normal tissues in RCC cases, and its expression is negatively associated with histological grade, metastasis, the prognosis of patients with RCC, and the expression of HIF-2α and CYR61 (P < 0.05). CONCLUSIONS: MIIP is a novel tumor suppressor in ccRCC via negative regulation of HIF-2α-CYR61 axis.


Subject(s)
Carcinoma, Renal Cell , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Neoplasms , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Proliferation , Cysteine/genetics , Cysteine/metabolism , Cysteine/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, Nude , Neoplastic Processes
8.
Biofouling ; 35(8): 900-921, 2019 09.
Article in English | MEDLINE | ID: mdl-31617758

ABSTRACT

Antimicrobial peptides (AMPs) are short, positively charged host defense peptides, found in various life forms from microorganisms to humans. AMPs are gaining more attention as substitutes for antibiotics in order to combat the risk posed by multi-drug- resistant pathogens. The nematode Caenorhabditis elegans relies solely on its innate immune defense to cope with its challenging life-style. Bacterial infection in C. elegans leads to induction of antimicrobial proteins, defensins, nemapores, cecropins, and neuropeptide-like proteins, which act to limit bacterial proliferation. This study reports how the C. elegans recombinant antibacterial factor (ABF-1) rapidly inhibited bacterial growth (Salmonella Typhi, Klebsiella pneumonia, Shigella sonnei and Vibrio alginolyticus). The ABF-1 exposure on S. Typhi, showed differential regulation in cell-cycle, DNA repair mechanism, membrane stability, and stress related proteins. The exogenous supply of ABF-1 protein has extended C. elegans survival by reducing the bacterial colony forming units on the nematode intestine. Together, these findings indicate the valuable and potential therapeutic applications of ABF-1 protein as antimicrobial agents against intracellular pathogens.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Caenorhabditis elegans Proteins/pharmacology , Caenorhabditis elegans , Recombinant Proteins/pharmacology , Salmonella typhi/drug effects , Animals , Antimicrobial Cationic Peptides/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/genetics , Humans , Intestines/microbiology , Microbial Sensitivity Tests , Recombinant Proteins/genetics , Survival Analysis
9.
Acta Neuropathol Commun ; 6(1): 121, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30409172

ABSTRACT

Pathologic, biochemical and genetic evidence indicates that accumulation and aggregation of amyloid ß-proteins (Aß) is a critical factor in the pathogenesis of Alzheimer's disease (AD). Several therapeutic interventions attempting to lower Aß have failed to ameliorate cognitive decline in patients with clinical AD significantly, but most such approaches target only one or two facets of Aß production/clearance/toxicity and do not consider the heterogeneity of human Aß species. As synaptic dysfunction may be among the earliest deficits in AD, we used hippocampal long-term potentiation (LTP) as a sensitive indicator of the early neurotoxic effects of Aß species. Here we confirmed prior findings that soluble Aß oligomers, much more than fibrillar amyloid plaque cores or Aß monomers, disrupt synaptic function. Interestingly, not all (84%) human AD brain extracts are able to inhibit LTP and the degree of LTP impairment by AD brain extracts does not correlate with Aß levels detected by standard ELISAs. Bioactive AD brain extracts also induce neurotoxicity in iPSC-derived human neurons. Shorter forms of Aß (including Aß1-37, Aß1-38, Aß1-39), pre-Aß APP fragments (- 30 to - 1) and N-terminally extended Aßs (- 30 to + 40) each showed much less synaptotoxicity than longer Aßs (Aß1-42 - Aß1-46). We found that antibodies which target the N-terminus, not the C-terminus, efficiently rescued Aß oligomer-impaired LTP and oligomer-facilitated LTD. Our data suggest that preventing soluble Aß oligomer formation and targeting their N-terminal residues with antibodies could be an attractive combined therapeutic approach.


Subject(s)
Alzheimer Disease/pathology , Hippocampus/pathology , Synapses/pathology , Synapses/physiology , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/immunology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Animals , Antibodies/pharmacology , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Disease Models, Animal , Female , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Long-Term Potentiation/drug effects , Long-Term Potentiation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Neural Inhibition/drug effects , Neural Inhibition/genetics , Peptide Fragments/pharmacology , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
10.
Dev Dyn ; 247(8): 965-975, 2018 08.
Article in English | MEDLINE | ID: mdl-29770538

ABSTRACT

BACKGROUND: In the developing mouse embryo, the bHLH transcription factor Neurog2 is transiently expressed by retinal progenitor cells and required for the initial wave of neurogenesis. Remarkably, another bHLH factor, Ascl1, normally not present in the embryonic Neurog2 retinal lineage, can rescue the temporal phenotypes of Neurog2 mutants. RESULTS: Here we show that Neurog2 simultaneously promotes terminal cell cycle exit and retinal ganglion cell differentiation, using mitotic window labeling and integrating these results with retinal marker quantifications. We also analyzed the transcriptomes of E12.5 GFP-expressing cells from Neurog2GFP/+ , Neurog2GFP/GFP , and Neurog2Ascl1KI/GFP eyes, and validated the most significantly affected genes using qPCR assays. CONCLUSIONS: Our data support the hypothesis that Neurog2 acts at the top of a retinal bHLH transcription factor hierarchy. The combined expression levels of these downstream factors are sufficiently induced by ectopic Ascl1 to restore RGC genesis, highlighting the robustness of this gene network during retinal ganglion cell neurogenesis. Developmental Dynamics 247:965-975, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Cycle , Nerve Tissue Proteins/physiology , Neurogenesis , Retinal Ganglion Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Cell Differentiation/drug effects , Embryo, Mammalian , Mice , Transcriptome/drug effects
11.
ACS Chem Neurosci ; 7(9): 1188-91, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27504748

ABSTRACT

The ability to assess oxygenation within living cells is much sought after to more deeply understand normal and pathological cell biology. Hypoxia Red manufactured by Enzo Life Sciences is advertised as a novel hypoxia detector dependent on nitroreducatase activity. We sought to use Hypoxia Red in primary neuronal cultures to test cell-to-cell metabolic variability in response to hypoxic stress. Neurons treated with 90 min of hypoxia were labeled with Hypoxia Red. We observed that, even under normoxic conditions neurons expressed fluorescence robustly. Analysis of the chemical reactions and biological underpinnings of this method revealed that the high uptake and reduction of the dye is due to active nitroreductases in normoxic cells that are independent of oxygen availability.


Subject(s)
Cell Hypoxia/physiology , Neurons/metabolism , Nitroreductases/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Cell Count , Cell Hypoxia/drug effects , Cells, Cultured , Embryo, Mammalian , Glucose/deficiency , Microtubule-Associated Proteins/metabolism , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Oxygen , Prosencephalon/cytology , Rats , Rats, Sprague-Dawley , Subcellular Fractions/metabolism , Tubulin/metabolism
12.
Int J Mol Sci ; 16(11): 26667-76, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26561805

ABSTRACT

Protein transduction using cell-penetrating peptides (CPPs) is useful for the delivery of large protein molecules, including some transcription factors. This method is safer than gene transfection methods with a viral vector because there is no risk of genomic integration of the exogenous DNA. Recently, this method was reported as a means for the induction of induced pluripotent stem (iPS) cells, directing the differentiation into specific cell types and supporting gene editing/correction. Furthermore, we developed a direct differentiation method to obtain a pancreatic lineage from mouse and human pluripotent stem cells via the protein transduction of three transcription factors, Pdx1, NeuroD, and MafA. Here, we discuss the possibility of using CPPs as a means of directing the differentiation of iPS cells and other stem cell technologies.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/pharmacology , Cell-Penetrating Peptides/pharmacology , Homeodomain Proteins/pharmacology , Induced Pluripotent Stem Cells/drug effects , Insulin-Secreting Cells/drug effects , Maf Transcription Factors, Large/pharmacology , Nerve Tissue Proteins/pharmacology , Trans-Activators/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/metabolism , Cellular Reprogramming/drug effects , Cinnamates/pharmacology , Gene Expression , Glucagon-Like Peptide 1/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Niacinamide/pharmacology , Trans-Activators/genetics , Trans-Activators/metabolism , Tretinoin/pharmacology , Veratrum Alkaloids/pharmacology
13.
Eur J Clin Invest ; 44(10): 989-99, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25208310

ABSTRACT

BACKGROUND: Cardiovascular patients suffer from reduced blood flow leading to ischaemia and impaired tissue metabolism. Unfortunately, an increasing group of elderly patients cannot be treated with current revascularization methods. Thus, new treatment strategies are urgently needed. Hypoxia-inducible factors (HIFs) upregulate the expression of angiogenic mediators together with genes involved in energy metabolism and recovery of ischaemic tissues. Especially, HIF-2α is a novel factor, and only limited information is available about its therapeutic potential. METHODS: Gene transfers with adenoviral HIF-1α and HIF-2α were performed into the mouse heart and rabbit ischaemic hindlimbs. Angiogenesis was evaluated by histology. Left ventricle function was analysed with echocardiography. Perfusion in rabbit skeletal muscles and energy recovery after electrical stimulation-induced exercise were measured with ultrasound and (31)P-magnetic resonance spectroscopy ((31)P-MRS), respectively. RESULTS: HIF-1α and HIF-2α gene transfers increased capillary size up to fivefold in myocardium and ischaemic skeletal muscles. Perfusion in skeletal muscles was increased by fourfold without oedema. Especially, AdHIF-1α enhanced the recovery of ischaemic muscles from electrical stimulation-induced energy depletion. Special characteristic of HIF-2α gene transfer was a strong capillary growth in muscle connective tissue and that HIF-2α gene transfer maintained left ventricle function. CONCLUSIONS: We conclude that both AdHIF-1α and AdHIF-2α gene transfers induced beneficial angiogenesis in vivo. Transient moderate increases in angiogenesis improved energy recovery after exercise in ischaemic muscles. This study shows for the first time that a moderate increase in angiogenesis is enough to improve tissue energy metabolism, which is potentially a very useful feature for cardiovascular gene therapy.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Muscle, Skeletal/metabolism , Neovascularization, Physiologic/drug effects , Animals , Capillaries/physiology , Coronary Vessels/physiology , Gene Expression/physiology , Gene Transfer Techniques , Genetic Therapy/methods , Hindlimb/blood supply , Ischemia/physiopathology , Ischemia/therapy , Mice, Inbred C57BL , Muscle, Skeletal/blood supply , Myocardium/metabolism , Rabbits
14.
Cell Physiol Biochem ; 33(3): 847-58, 2014.
Article in English | MEDLINE | ID: mdl-24685606

ABSTRACT

BACKGROUND: Neurogenin2 (Ngn2) is a proneural gene that directs neuronal differentiation of progenitor cells during development. Here, we investigated whether Ngn2 can reprogram MSCs to adopt a neural precursor fate and enhance the therapeutic effects of MSCs after experimental stroke. METHODS: In vitro, MSCs were transfected with lenti-GFP or lenti-Ngn2. Following neuronal induction, cells were identified by immunocytochemistry, Western blot and electrophysiological analyses. In a stroke model induced by transient right middle cerebral artery occlusion (MCAO), PBS, GFP-MSCs or Ngn2-MSCs were injected 1 day after MCAO. Behavioral tests, neurological and immunohistochemical assessments were performed. RESULTS: In vitro, Ngn2-MSCs expressed neural stem cells markers (Pax6 and nestin) and lost the potential to differentiate into mesodermal cell types. Following neural induction, Ngn2-MSCs expressed higher levels of neuron-specific proteins MAP2, Tuj1 and NeuN, and also expressed voltage-gated Na+ channel, which was absent in GFP-MSCs. In vivo, after transplantation, Ngn2-MSCs significantly reduced apoptotic cells, decreased infarct volume, and increased the expression of VEGF and BDNF. Finally, Ngn2-MSCs treated animals showed the highest functional recovery among the three groups. CONCLUSIONS: Ngn2 was sufficient to convert MSCs into a neural precursor fate and transplantation of Ngn2-MSCs was advantageous for the treatment of stroke rats.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/pharmacology , Cell Differentiation/drug effects , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Nerve Tissue Proteins/pharmacology , Neural Stem Cells/metabolism , Stroke/therapy , Allografts , Animals , Antigens, Differentiation/biosynthesis , Male , Mesenchymal Stem Cells/pathology , Neural Stem Cells/pathology , Rats , Rats, Sprague-Dawley , Stroke/metabolism , Stroke/pathology
15.
Glia ; 61(2): 225-39, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23047160

ABSTRACT

Glioblastoma multiform (GBM) are devastating brain tumors containing a fraction of multipotent stem-like cells which are highly tumorigenic. These cells are resistant to treatments and are likely to be responsible for tumor recurrence. One approach to eliminate GBM stem-like cells would be to force their terminal differentiation. During development, neurons formation is controlled by neurogenic transcription factors such as Ngn1/2 and NeuroD1. We found that in comparison with oligodendrogenic genes, the expression of these neurogenic genes is low or absent in GBM tumors and derived cultures. We thus explored the effect of overexpressing these neurogenic genes in three CD133(+) Sox2(+) GBM stem-like cell cultures and the U87 glioma line. Introduction of Ngn2 in CD133(+) cultures induced massive cell death, proliferation arrest and a drastic reduction of neurosphere formation. Similar effects were observed with NeuroD1. Importantly, Ngn2 effects were accompanied by the downregulation of Olig2, Myc, Shh and upregulation of Dcx and NeuroD1 expression. The few surviving cells adopted a typical neuronal morphology and some of them generated action potentials. These cells appeared to be produced at the expense of GFAP(+) cells which were radically reduced after differentiation with Ngn2. In vivo, Ngn2-expressing cells were unable to form orthotopic tumors. In the U87 glioma line, Ngn2 could not induce neuronal differentiation although proliferation in vitro and tumoral growth in vivo were strongly reduced. By inducing cell death, cell cycle arrest or differentiation, this work supports further exploration of neurogenic proteins to oppose GBM stem-like and non-stem-like cell growth.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/pharmacology , Brain Neoplasms/pathology , Cell Differentiation , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Transcription Factors/pharmacology , AC133 Antigen , Antigens, CD/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Death , Flow Cytometry , Glial Fibrillary Acidic Protein/metabolism , Glycoproteins/metabolism , Hedgehog Proteins/metabolism , Humans , Neoplastic Stem Cells/drug effects , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Oligodendrocyte Transcription Factor 2 , Oncogene Protein p55(v-myc)/metabolism , Peptides/metabolism , SOXB1 Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured
16.
Biochem Biophys Res Commun ; 427(3): 531-6, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23022191

ABSTRACT

Olig2 protein, a member of the basic helix-loop-helix transcription factor family, was introduced into the mouse embryonic carcinoma cell line P19 for induction of motor neuron differentiation. We show that Olig2 protein has the ability to permeate the cell membrane without the addition of a protein transduction domain (PTD), similar to other basic helix-loop-helix transcription factors such as MyoD and NeuroD2. Motor neuron differentiation was evaluated for the elongation of neurites and the expression of choline acetyltransferase (ChAT) mRNA, a differentiation marker of motor neurons. By addition of Olig2 protein, motor neuron differentiation was induced in P19 cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Motor Neurons/cytology , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Cell Line, Tumor , Choline O-Acetyltransferase/biosynthesis , Mice , Motor Neurons/drug effects , Motor Neurons/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Neurites/metabolism , Neurites/physiology , Neurogenesis/drug effects , Oligodendrocyte Transcription Factor 2 , Protein Structure, Tertiary , Protein Transport , RNA, Messenger/biosynthesis
17.
Transplant Proc ; 42(6): 2071-4, 2010.
Article in English | MEDLINE | ID: mdl-20692411

ABSTRACT

BACKGROUND: It has been reported that the human pancreatic nonendocrine fraction, which remains after islet isolation, can be differentiated toward beta cells. However, the optimal method to accomplish this goal has not been established. In this study, we introduced the human neurogenic differentiation 1 (NeuroD1) gene into human nonendocrine pancreatic epithelial cells (NEPECs) and promoted insulin-producing cells in vitro. METHODS: The human pancreatic nonislet fractions were obtained from brain-dead donors and cultured in suspension for 2-3 days followed by culture with G418 for 4 days. These cells (NEPECs) were then plated on dishes. The NEPECs spread into a cell monolayer within 7 days and all of the cells were cytokeratin-19 (CK19) positive. Seven days after plating, plasmids encoding human NeuroD1 gene under human CK19 promoter were transfected 3 times every other day (termed NEPEC+ND). Seven days after starting induction, these cells were characterized. RESULTS: Seven days after starting the induction of human NeuroD1, NEPEC+ND strongly expressed NeuroD1 and insulin mRNA. The ratio of NeuroD1-positive cells in NEPEC+ND was significantly higher than in NEPEC. Human insulin-positive cells in NEPEC+ND were also significantly greater than in NEPEC. Human insulin and C-peptide levels in culture medium in NEPEC+ND were significantly higher than in NEPEC. CONCLUSIONS: These findings demonstrated that human NeuroD1 under control of the CK19 promoter can induce the differentiation of CK19-positive NEPECs into insulin-producing cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/pharmacology , Cell Differentiation/drug effects , Insulin/metabolism , Nerve Tissue Proteins/pharmacology , Pancreas/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Death , Cell Culture Techniques/methods , Cell Division , DNA Primers , Humans , Insulin/genetics , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/physiology , Keratin-19/genetics , Nerve Tissue Proteins/genetics , Pancreas/cytology , Pancreas/drug effects , Pancreas/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
Arthritis Rheum ; 62(9): 2707-15, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20496369

ABSTRACT

OBJECTIVE: Since nucleus pulposus cells reside under conditions of hypoxia, we determined if the expression of ANK, a pyrophosphate transporter, is regulated by the hypoxia-inducible factor (HIF) proteins. METHODS: Quantitative reverse transcription-polymerase chain reaction and Western blot analyses were used to measure ANK expression in nucleus pulposus cells from rats and humans. Transfections were performed to determine the effect of HIF-1/2 on ANK promoter activity. RESULTS: ANK was expressed in embryonic and mature rat discs. Oxygen-dependent changes in ANK expression in nucleus pulposus cells were minimal. However, silencing of HIF-1α and HIF-2α resulted in increased ANK expression and up-regulation of promoter activity. HIF-mediated suppression of ANK was validated by measuring promoter activity in HIF-1ß-null embryonic fibroblasts. Under conditions of hypoxia, there was induction of promoter activity in the null cells as compared with the wild-type cells. Overexpression of HIF-1α and HIF-2α in nucleus pulposus cells resulted in a significant suppression of ANK promoter activity. Since the ANK promoter contains 2 hypoxia-responsive elements (HREs), we performed site-directed mutagenesis and measured promoter activity. We found that HIF-1 can bind to either of the HREs and can suppress promoter activity; in contrast, HIF-2 was required to bind to both HREs in order to suppress activity. Finally, analysis of human nucleus pulposus tissue showed that while ANK was expressed in normal tissue, there was increased expression of ANK along with alkaline phosphatase in the degenerated state. CONCLUSION: Both HIF-1 and HIF-2 serve as negative regulators of ANK expression in the disc. We propose that baseline expression of ANK in the disc serves to prevent mineral formation under physiologic conditions.


Subject(s)
Ankyrins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcinosis/metabolism , Hypoxia-Inducible Factor 1/metabolism , Intervertebral Disc/metabolism , Adult , Aged , Aged, 80 and over , Animals , Ankyrins/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Blotting, Western , Calcinosis/chemically induced , Calcinosis/pathology , Cell Hypoxia/physiology , Cells, Cultured , Embryo, Mammalian/cytology , Fluorescent Antibody Technique, Indirect , Gene Expression/drug effects , Gene Silencing , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit , Intervertebral Disc/drug effects , Intervertebral Disc/pathology , Mice , Mice, Knockout , Middle Aged , RNA, Messenger/metabolism , Rats
19.
Stem Cells ; 27(9): 2238-46, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19522012

ABSTRACT

Nurr1 is a transcription factor specific for the development and maintenance of the midbrain dopamine (DA) neurons. Exogenous Nurr1 in neural precursor (NP) cells induces the differentiation of DA neurons in vitro that are capable of reversing motor dysfunctions in a rodent model for Parkinson disease. The promise of this therapeutic approach, however, is unclear due to poor cell survival and phenotype loss of DA cells after transplantation. We herein demonstrate that Nurr1 proteins undergo ubiquitin-proteasome-system-mediated degradation in differentiating NP cells. The degradation process is activated by a direct Akt-mediated phosphorylation of Nurr1 proteins and can be prevented by abolishing the Akt-target sequence in Nurr1 (Nurr1(Akt)). Overexpression of Nurr1(Akt) in NP cells yielded DA neurons in which Nurr1 protein levels were maintained for prolonged periods. The sustained Nurr1 expression endowed the Nurr1(Akt)-induced DA neurons with resistance to toxic stimuli, enhanced survival, and sustained DA phenotypes in vitro and in vivo after transplantation.


Subject(s)
Dopamine/metabolism , Neurons/cytology , Neurons/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Blotting, Western , Butadienes/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cell Survival/genetics , Cell Survival/physiology , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Humans , Immunoprecipitation , Mesencephalon/cytology , Morpholines/pharmacology , Nitriles/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Protein Stability/drug effects , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
20.
BJU Int ; 102(3): 358-63, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18394010

ABSTRACT

OBJECTIVE: To determine the role of hypoxia-inducible factor-2alpha (HIF2alpha) on the sensitivity of renal cell carcinoma (RCC) cell lines to ionizing radiation and to determine if the mTOR antagonist, rapamycin, could decrease HIF2alpha protein levels. MATERIALS AND METHODS: Cell lines expressing stable short-hairpin RNAs (shRNAs) encoding HIF2alpha shRNAs or an empty vector were transfected with a hypoxia responsive element (HRE)-driven firefly luciferase reporter gene. Two separate paired cell lines were assayed for their response to increasing doses of ionizing radiation. Proliferation and cell cycle kinetics were compared for cell lines expressing HIF2alpha shRNAs and empty vectors. The effect of an mTOR antagonist, rapamycin on HIF1alpha and HIF2alpha proteins levels was also assessed. RESULTS: We confirmed that the 786-O RCC lines with stably integrated shRNAs against HIF2alpha had decreased activation of a plasmid with a HRE-driven firefly luciferase reporter gene. Lines from two separate cell clones with decreased HIF2alpha levels showed a significant increase in radiation sensitivity and an increase in G2 cell cycle arrest. Rapamycin, while effective in decreasing HIF1alpha protein levels, did not affect HIF2alpha levels in either of the RCC cell lines. CONCLUSIONS: These results show that decreasing levels of HIF2alpha leads to an increased sensitivity to ionizing radiation. This finding may explain in part, the known resistance of RCC to radiation therapy. Although mTOR antagonists are approved for the treatment of RCC, these agents do not decrease HIF2alpha levels and therefore might not be effective in enhancing the radio-sensitivity of these tumours.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/pharmacology , Carcinoma, Renal Cell/radiotherapy , Kidney Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Sirolimus/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Flow Cytometry , G2 Phase/drug effects , Genes, Reporter , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Luciferases/metabolism , Protein Kinases/drug effects , Radiation Tolerance , Radiation-Sensitizing Agents/metabolism , TOR Serine-Threonine Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...