Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cancers (Basel) ; 16(3)2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38339252

ABSTRACT

Ovarian cancer (OCa) is the most lethal form of gynecologic cancer, and the tumor heterogeneities at the molecular, cellular, and tissue levels fuel tumor resistance to standard therapies and pose a substantial clinical challenge. Here, we tested the hypothesis that the heightened basal endoplasmic reticulum stress (ERS) observed in OCa represents an exploitable vulnerability and may overcome tumor heterogeneity. Our recent studies identified LIPA as a novel target to induce ERS in cancer cells using the small molecule ERX-41. However, the role of LIPA and theutility of ERX-41 to treat OCa remain unknown. Expression analysis using the TNMplot web tool, TCGA data sets, and immunohistochemistry analysis using a tumor tissue array showed that LIPA is highly expressed in OCa tissues, compared to normal tissues. ERX-41 treatment significantly reduced the cell viability and colony formation ability and promoted the apoptosis of OCa cells. Mechanistic studies revealed a robust and consistent induction of ERS markers, including CHOP, elF2α, PERK, and ATF4, upon ERX-41 treatment. In xenograft and PDX studies, ERX-41 treatment resulted in a significant reduction in tumor growth. Collectively, our results suggest that ERX-41 is a novel therapeutic agent that targets the LIPA with a unique mechanism of ERS induction, which could be exploited to treat heterogeneity in OCa.

2.
Breast Cancer Res Treat ; 200(1): 151-162, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37199805

ABSTRACT

PURPOSE: Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer. Oncogenic PELP1 is frequently overexpressed in TNBC, and it has been demonstrated that PELP1 signaling is essential for TNBC progression. The therapeutic utility of targeting PELP1 in TNBC, however, remains unknown. In this study, we investigated the effectiveness of SMIP34, a recently developed PELP1 inhibitor for the treatment of TNBC. METHODS: To ascertain the impact of SMIP34 treatment, we used seven different TNBC models for testing cell viability, colony formation, invasion, apoptosis, and cell cycle analysis. Western blotting and RT-qPCR were used to determine the mechanistic insights of SMIP34 action. Using xenograft and PDX tumors, the ability of SMIP34 in suppressing proliferation was examined both ex vivo and in vivo. RESULTS: TNBC cells' viability, colony formation, and invasiveness were all decreased by SMIP34 in in vitro cell-based assays, while apoptosis was increased. SMIP34 treatment promoted the degradation of PELP1 through the proteasome pathway. RT-qPCR analyses confirmed that SMIP34 treatment downregulated PELP1 target genes. Further, SMIP34 treatment substantially downregulated PELP1 mediated extranuclear signaling including ERK, mTOR, S6 and 4EBP1. Mechanistic studies confirmed downregulation of PELP1 mediated ribosomal biogenesis functions including downregulation of cMyc and Rix complex proteins LAS1L, TEX-10, and SENP3. The proliferation of TNBC tumor tissues was decreased in explant experiments by SMIP34. Additionally, SMIP34 treatment markedly decreased tumor progression in both TNBC xenograft and PDX models. CONCLUSIONS: Together, these findings from in vitro, ex vivo, and in vivo models show that SMIP34 may be a useful therapeutic agent for inhibiting PELP1 signaling in TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Cell Line, Tumor , Cell Proliferation , Co-Repressor Proteins , Cysteine Endopeptidases/metabolism , Signal Transduction , Transcription Factors , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
3.
Cancer Res ; 82(20): 3830-3844, 2022 10 17.
Article in English | MEDLINE | ID: mdl-35950923

ABSTRACT

Most patients with estrogen receptor alpha-positive (ER+) breast cancers initially respond to treatment but eventually develop therapy resistance with disease progression. Overexpression of oncogenic ER coregulators, including proline, glutamic acid, and leucine-rich protein 1 (PELP1), are implicated in breast cancer progression. The lack of small molecules that inhibits PELP1 represents a major knowledge gap. Here, using a yeast-two-hybrid screen, we identified novel peptide inhibitors of PELP1 (PIP). Biochemical assays demonstrated that one of these peptides, PIP1, directly interacted with PELP1 to block PELP1 oncogenic functions. Computational modeling of PIP1 revealed key residues contributing to its activity and facilitated the development of a small-molecule inhibitor of PELP1, SMIP34, and further analyses confirmed that SMIP34 directly bound to PELP1. In breast cancer cells, SMIP34 reduced cell growth in a dose-dependent manner. SMIP34 inhibited proliferation of not only wild-type (WT) but also mutant (MT) ER+ and therapy-resistant breast cancer cells, in part by inducing PELP1 degradation via the proteasome pathway. RNA sequencing analyses showed that SMIP34 treatment altered the expression of genes associated with estrogen response, cell cycle, and apoptosis pathways. In cell line-derived and patient-derived xenografts of both WT and MT ER+ breast cancer models, SMIP34 reduced proliferation and significantly suppressed tumor progression. Collectively, these results demonstrate SMIP34 as a first-in-class inhibitor of oncogenic PELP1 signaling in advanced breast cancer. SIGNIFICANCE: Development of a novel inhibitor of oncogenic PELP1 provides potential therapeutic avenues for treating therapy-resistant, advanced ER+ breast cancer.


Subject(s)
Breast Neoplasms , Co-Repressor Proteins , Transcription Factors , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/metabolism , Estrogen Receptor alpha/genetics , Estrogens , Female , Glutamic Acid , Humans , Leucine , Proline , Proteasome Endopeptidase Complex , Receptors, Estrogen/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
4.
Carbohydr Polym ; 294: 119791, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-35868798

ABSTRACT

Chitosan (CS) was modified with two fatty acids, i.e., capric acid (CA) and palmitic acid (PA). Particle size (315.8 nm), zeta potential (31.8 mV), and viscosity (29.4 mPa.s) of CS-PA nanogels were lower than CS-CA nanogels (793.2 nm, 53.3 mV, and 70.7 mPa.s). First, hempseed oil-in-water Pickering nanoemulsions were stabilized by CS-based particles/maltodextrin (MD). Then, the emulsions were dried using an electrostatic collector-equipped spray dryer. The D50 of re-dispersed emulsion powders with CS-PA/MD coating was 936 nm. According to the FE-SEM images, oil coated with CS-PA/MD showed higher porosity and C/O ratio at the particle surface compared to the CS-CA/MD coating leading to more oil leakage. In addition, the crystallinity of hempseed oil coated with CS-PA/MD was higher than the one coated with CS-CA/MD. These findings showed that submicron Pickering emulsion powders could be achieved by targeted modification of CS and using a spray dryer equipped with an electrostatic collector.


Subject(s)
Chitosan , Cannabis , Emulsions , Nanogels , Particle Size , Plant Extracts , Powders , Static Electricity
5.
Breast Cancer Res ; 24(1): 26, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35395812

ABSTRACT

BACKGROUND: Methyltransferase SETDB1 is highly expressed in breast cancer (BC), however, the mechanisms by which SETDB1 promotes BC progression to endocrine therapy resistance remains elusive. In this study, we examined the mechanisms by which SETDB1 contribute to BC endocrine therapy resistance. METHODS: We utilized therapy sensitive (MCF7 and ZR75), therapy resistant (MCF7-TamR, MCF7-FR, MCF7-PELP1cyto, MCF7-SETDB1) estrogen receptor alpha positive (ER+)BC models and conducted in vitro cell viability, colony formation, 3-dimensional cell growth assays to investigate the role of SETDB1 in endocrine resistance. RNA-seq of parental and SETDB1 knock down ER+ BC cells was used to identify unique pathways. SETDB1 interaction with PELP1 was identified by yeast-two hybrid screen and confirmed by immunoprecipitation and GST-pull down assays. Mechanistic studies were conducted using Western blotting, reporter gene assays, RT-qPCR, and in vitro methylation assays. Xenograft assays were used to establish the role of PELP1 in SETDB1 mediated BC progression. RESULTS: RNA-seq analyses showed that SETDB1 regulates expression of a subset of estrogen receptor (ER) and Akt target genes that contribute to endocrine therapy resistance. Importantly, using yeast-two hybrid screen, we identified ER coregulator PELP1 as a novel interacting protein of SETDB1. Biochemical analyses confirmed SETDB1 and PELP1 interactions in multiple BC cells. Mechanistic studies confirmed that PELP1 is necessary for SETDB1 mediated Akt methylation and phosphorylation. Further, SETDB1 overexpression promotes tamoxifen resistance in BC cells, and PELP1 knockdown abolished these effects. Using xenograft model, we provided genetic evidence that PELP1 is essential for SETDB1 mediated BC progression in vivo. Analyses of TCGA datasets revealed SETDB1 expression is positively correlated with PELP1 expression in ER+ BC patients. CONCLUSIONS: This study suggests that the PELP1/SETDB1 axis play an important role in aberrant Akt activation and serves as a novel target for treating endocrine therapy resistance in breast cancer.


Subject(s)
Breast Neoplasms , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Co-Repressor Proteins/pharmacology , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/pharmacology , Humans , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Saccharomyces cerevisiae/metabolism , Tamoxifen/pharmacology , Transcription Factors/genetics
6.
Commun Biol ; 4(1): 1235, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34716410

ABSTRACT

Histone deacetylase inhibitors (HDACi) are identified as novel therapeutic agents, however, recent clinical studies suggested that they are marginally effective in treating triple negative breast cancer (TNBC). Here, we show that first-in-class Leukemia Inhibitory Factor Receptor (LIFRα) inhibitor EC359 could enhance the therapeutic efficacy of HDACi against TNBC. We observed that both targeted knockdown of LIFR with CRISPR or treatment with EC359 enhanced the potency of four different HDACi in reducing cell viability, cell survival, and enhanced apoptosis compared to monotherapy in TNBC cells. RNA-seq studies demonstrated oncogenic/survival signaling pathways activated by HDACi were attenuated by the EC359 + HDACi therapy. Importantly, combination therapy potently inhibited the growth of TNBC patient derived explants, cell derived xenografts and patient-derived xenografts in vivo. Collectively, our results suggest that targeted inhibition of LIFR can enhance the therapeutic efficacy of HDACi in TNBC.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Leukemia Inhibitory Factor Receptor alpha Subunit/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Survival/drug effects , Female , Histone Deacetylase Inhibitors/administration & dosage , Mice , Mice, SCID
7.
Neurooncol Adv ; 3(1): vdab099, 2021.
Article in English | MEDLINE | ID: mdl-34485908

ABSTRACT

BACKGROUND: Glioblastomas (GBMs) are the most lethal primary brain tumors. Estrogen receptor ß (ESR2/ERß) function as a tumor suppressor in GBM, however, ERß expression is commonly suppressed during glioma progression. In this study, we examined whether drugs that reverse epigenetic modifications will enhance ERß expression and augment ERß agonist-mediated tumor suppression. METHODS: We tested the utility of epigenetic drugs which act as an inhibitor of histone deacetylases (HDACs), histone methylases, and BET enzymes. Mechanistic studies utilized RT-qPCR, chromatin immunoprecipitation (ChIP), and western blotting. Cell viability, apoptosis, colony formation, and invasion were measured using in vitro assays. An orthotopic GBM model was used to test the efficacy of in vivo. RESULTS: Of all inhibitors tested, HDACi (panobinostat and romidepsin) showed the potential to increase the expression of ERß in GBM cells. Treatment with HDACi uniquely upregulated ERß isoform 1 expression that functions as a tumor suppressor but not ERß isoform 5 that drives oncogenic functions. Further, combination therapy of HDACi with the ERß agonist, LY500307, potently reduced cell viability, invasion, colony formation, and enhanced apoptosis. Mechanistic studies showed that HDACi induced ERß is functional, as it enhanced ERß reporter activities and ERß target genes expression. ChIP analysis confirmed alterations in the histone acetylation at the ERß and its target gene promoters. In orthotopic GBM model, combination therapy of panobinostat and LY500307 enhanced survival of tumor-bearing mice. CONCLUSIONS: Our results suggest that the combination therapy of HDACi and LY500307 provides therapeutic utility in overcoming the suppression of ERß expression that commonly occurs in GBM progression.

8.
Cell Death Discov ; 7(1): 216, 2021 Aug 16.
Article in English | MEDLINE | ID: mdl-34400617

ABSTRACT

Endometrial cancer (EC) is the fourth most common cancer in women. Advanced-stage EC has limited treatment options with a poor prognosis. There is an unmet need for the identification of actionable drivers for the development of targeted therapies in EC. Leukemia inhibitory factor receptor (LIFR) and its ligand LIF play a major role in cancer progression, metastasis, stemness, and therapy resistance. However, little is known about the functional significance of the LIF/LIFR axis in EC progression. In this study using endometrial tumor tissue arrays, we identified that expression of LIF, LIFR is upregulated in EC. Knockout of LIFR using CRISPR/Cas9 in two different EC cells resulted in a significant reduction of their cell viability and cell survival. In vivo studies demonstrated that LIFR-KO significantly reduced EC xenograft tumor growth. Treatment of established and primary patient-derived EC cells with a novel LIFR inhibitor, EC359 resulted in the reduction of cell viability with an IC50 in the range of 20-100 nM and induction of apoptosis. Further, treatment with EC359 reduced the spheroid formation of EC cancer stem cells and reduced the levels of cancer stem cell markers SOX2, OCT4, NANOG, and Axin2. Mechanistic studies demonstrated that EC359 treatment attenuated the activation of LIF-LIFR driven pathways, including STAT3 and AKT/mTOR signaling in EC cells. Importantly, EC359 treatment resulted in a significant reduction of the growth of EC patient-derived explants ex vivo, EC cell line-derived xenografts, and patient-derived xenografts in vivo. Collectively, our work revealed the oncogenic potential of the LIF/LIFR axis in EC and support the utility of LIFR inhibitor, EC359, as a novel targeted therapy for EC via the inhibition of LIF/LIFR oncogenic signaling.

9.
Eur J Cancer ; 154: 11-20, 2021 09.
Article in English | MEDLINE | ID: mdl-34217908

ABSTRACT

BACKGROUND: CDK4/6 inhibitors modulate immune response in breast cancer. This phase I/II trial was designed to test the safety and efficacy of palbociclib, pembrolizumab and letrozole in women with hormone receptor positive (HR+) human epidermal growth factor receptor 2 negative (HER2-) metastatic breast cancer (MBC). PATIENTS AND METHODS: Women with stage IV HR+ HER2- MBC were enrolled and treated with palbociclib, pembrolizumab and letrozole. Primary end-points were safety, tolerability and efficacy. RESULTS: Between November 2016 and July 2020, 23 patients were enrolled with 20 evaluable for response, including 4 patients in cohort 1 and 16 patients in cohort 2. Cohort 1 median age was 48 years (33-70) and cohort 2 median age was 55 (37-75). Cohort 1 closed early due to limited accrual. Grade III-IV adverse events were neutropenia (83%), leucopaenia (65%), thrombocytopenia (17%) and elevated liver enzymes (17%). In cohort 1, 50% achieved a partial response (PR) and 50% had stable disease (SD). In cohort 2, 31% achieved complete response (CR), 25% had PR and 31% had SD by Response Evaluation Criteria in Solid Tumours version 1.1. Median progression-free survival was 25.2 months (95% confidence interval [CI] 5.3, not reached) and median overall survival was 36.9 months (95% CI 36.9, not reached) in cohort 2 with a median follow-up of 24.8 months (95% CI 17.1, not reached). A correlative immune biomarker analysis was published separately. CONCLUSION: The combination of palbociclib, pembrolizumab and letrozole is well tolerated, and a complete response rate of 31% was identified in HR+ MBC patients who received this combination as front-line therapy. Confirmatory trials are required to better understand the immune-priming effects of CDK4/6 inhibitors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Female , Humans , Immune Checkpoint Inhibitors/administration & dosage , Letrozole/administration & dosage , Middle Aged , Piperazines/administration & dosage , Pyridines/administration & dosage , Receptors, Estrogen/analysis
10.
Parasit Vectors ; 12(1): 510, 2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31666095

ABSTRACT

BACKGROUND: Apicomplexan parasites of the genus Cryptosporidium infect a wide range of animal species as well as humans. Cryptosporidium spp. can cause life threatening diarrhea especially in young animals, children, immunocompromised patients and malnourished individuals. Asymptomatic cryptosporidial infections in animals can also occur, making these animals potential reservoirs of infection. METHODS: In the present study, a molecular survey of Cryptosporidium spp. in ruminants that were slaughtered for human consumption in Yazd Province, located in central Iran was conducted. Faeces were collected per-rectum from 484 animals including 192 cattle, 192 sheep and 100 goats. DNA was extracted from all samples and screened for Cryptosporidium by PCR amplification of the 18S rRNA gene. Positives were Sanger sequenced and further subtyped by sequence analysis of the 60 kDa glycoprotein (gp60) locus. RESULTS: In total, Cryptosporidium spp. were detected in 22 animals: C. andersoni and C. bovis in seven and two cattle faecal samples, respectively, C. ubiquitum in five sheep, and C. xiaoi in six sheep and two goat samples, respectively. To our knowledge, this study provides for the first time, molecular information concerning Cryptosporidium species infecting goats in Iran, and is also the first report of C. ubiquitum and C. xiaoi from ruminants in Iran. CONCLUSION: The presence of potentially zoonotic species of Cryptosporidium in ruminants in this region may suggest that livestock could potentially contribute to human cryptosporidiosis, in particular among farmers and slaughterhouse workers, in the area. Further molecular studies on local human populations are required to more accurately understand the epidemiology and transmission dynamics of Cryptosporidium spp. in this region.


Subject(s)
Cryptosporidiosis/enzymology , Cryptosporidiosis/parasitology , Cryptosporidium/classification , Age Factors , Animals , Cattle , Cryptosporidiosis/epidemiology , Cryptosporidium/genetics , DNA, Protozoan/isolation & purification , Disease Reservoirs/parasitology , Feces/parasitology , Genotyping Techniques , Goats , Humans , Immunocompromised Host , Iran/epidemiology , Malnutrition/complications , Prevalence , RNA, Ribosomal, 18S/genetics , Sheep , Zoonoses/parasitology
11.
Infect Genet Evol ; 75: 104021, 2019 11.
Article in English | MEDLINE | ID: mdl-31494270

ABSTRACT

Giardia duodenalis is one of the most common intestinal parasites in humans as well as livestock and wildlife. It is of both public and veterinary health importance in developing nations. A molecular survey of Giardia duodenalis assemblages in ruminants from Yazd Province, Iran was conducted on 484 animal faecal samples collected per rectum from slaughtered ruminants including 192 cattle, 192 sheep and 100 goats from June to November 2017. Species-specific and assemblage-specific PCRs for assemblages A, B and E at the triose phosphate isomerase (tpi) gene were performed, and samples positive for Giardia were confirmed by sequencing. In total, 25 (5.16%) of examined faecal samples including eight cattle (4.2%), twelve sheep (6.2%) and five goats (5%) were infected with G. duodenalis. Assemblage-specific PCR detected G. duodenalis assemblage E in seven faecal samples (six in sheep and one in a goat). Assemblages A and B were not detected. This study provides the first insight into Giardia infection in slaughtered livestock in Iran. Although the prevalence of infection with Giardia in this hot-arid area of Iran was low, educating people about direct contact with livestock such as farmers and abattoirs workers about this zoonotic infection is important.


Subject(s)
Cattle Diseases/epidemiology , Cattle Diseases/parasitology , Giardia lamblia/genetics , Giardiasis/veterinary , Goat Diseases/epidemiology , Goat Diseases/parasitology , Sheep Diseases/epidemiology , Sheep Diseases/parasitology , Animals , Cattle , Giardia lamblia/classification , Goats , Iran/epidemiology , Molecular Typing , Prevalence , Sheep
12.
Cell Regen ; 7(1): 1-6, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30671223

ABSTRACT

Myocardial infarction leads to the loss of a huge number of cardiomyocytes and the reparatory response to this phenomenon is scar tissue formation, which impairs heart function. Direct reprogramming technology offers an alternative strategy for the generation of functional cardiomyocytes not only in vitro, but also in vivo in the site of injury. Results have demonstrated cardiac tissue regeneration and improvement in heart function after myocardial infarction following local injection of vectors encoding reprogramming transcription factors or miRNAs. This shows the great potential of cardiac reprogramming technology for heart regeneration. However, in addition to cardiomyocytes, other cell types, including endothelial cells and smooth muscle cells are also required to be generated in the damaged area in order to achieve complete cardiac tissue regeneration. To this aim induced proliferative/expandable cardiovascular progenitor cells (iCPCs) appear to be an appropriate cell source, which is capable of differentiation into three cardiovascular lineages both in vitro and in vivo. In this regard, this study goes over in vitro and in vivo cardiac reprogramming technology and specifically deals with cardiac progenitor reprogramming and its potential for heart regeneration.

13.
J Mol Cell Cardiol ; 108: 61-72, 2017 07.
Article in English | MEDLINE | ID: mdl-28502796

ABSTRACT

Replacing dying or diseased cells of a tissue with new ones that are converted from patient's own cells is an attractive strategy in regenerative medicine. In vivo reprogramming is a novel strategy that can circumvent the hurdles of autologous/allogeneic cell injection therapies. Interestingly, studies have demonstrated that direct injection of cardiac transcription factors or specific miRNAs into the infarct border zone of murine hearts following myocardial infarction converts resident cardiac fibroblasts into functional cardiomyocytes. Moreover, in vivo cardiac reprogramming not only drives cardiac tissue regeneration, but also improves cardiac function and survival rate after myocardial infarction. Thanks to the influence of cardiac microenvironment and the same developmental origin, cardiac fibroblasts seem to be more amenable to reprogramming toward cardiomyocyte fate than other cell sources (e.g. skin fibroblasts). Thus, reprogramming of cardiac fibroblasts to functional induced cardiomyocytes in the cardiac environment holds great promises for induced regeneration and potential clinical purposes. Application of small molecules in future studies may represent a major advancement in this arena and pharmacological reprogramming would convey reprogramming technology to the translational medicine paradigm. This study reviews accomplishments in the field of in vitro and in vivo mouse cardiac reprogramming and then deals with strategies for the enhancement of the efficiency and quality of the process. Furthermore, it discusses challenges ahead and provides suggestions for future research. Human cardiac reprogramming is also addressed as a foundation for possible application of in vivo cardiac reprogramming for human heart regeneration in the future.


Subject(s)
Cellular Reprogramming , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Regeneration , Animals , Cell Transdifferentiation/genetics , Cellular Microenvironment , Humans , In Vitro Techniques , Models, Animal , Regenerative Medicine , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Tissue Cell ; 48(5): 475-87, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27514850

ABSTRACT

Direct reprogramming of specialized cells into other cell types has revolutionized the fields of stem cell, differentiation, and regenerative medicine. Direct reprogramming technology can convert various differentiated cell types to other fates by the forced expression of lineage-specific transcription factors. In addition to this approach, transdifferentiation can be induced in somatic cells by a method named cell-activation and signaling-directed (CASD) lineage conversion, which uses pluripotency reprogramming factors in combination with specific differentiation signals. This approach is capable of generating tissue-specific progenitors in addition to functional mature cells through a challenging transitory (pluripotent or non-pluripotent) state. Interestingly, the CASD lineage conversion has been accomplished by using small molecules and growth factors in a chemical-only paradigm. This approach will have a substantial positive impact on the field and bring reprogramming technology into the translational pipeline. From the regenerative medicine perspective, in future, new therapies might be designed based on the patient's own cells that are directly reprogrammed into the desired cell types in vitro and in situ. This review provides an overview of direct reprogramming to different lineages and describes what is known about the cell-activation and signaling-directed transdifferentiation.


Subject(s)
Cell Differentiation/genetics , Cell Engineering , Cell Lineage/genetics , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Cell Transdifferentiation/genetics , Fibroblasts/cytology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Organ Specificity/genetics , Signal Transduction/genetics
15.
Int J Stem Cells ; 9(1): 9-20, 2016 May 30.
Article in English | MEDLINE | ID: mdl-27426081

ABSTRACT

Forced expression of lineage-specific transcription factors in somatic cells can result in the generation of different cell types in a process named direct reprogramming, bypassing the pluripotent state. However, the introduction of transgenes limits the therapeutic applications of the produced cells. Numerous small-molecules have been introduced in the field of stem cell biology capable of governing self-renewal, reprogramming, transdifferentiation and regeneration. These chemical compounds are versatile tools for cell fate conversion toward desired outcomes. Cell fate conversion using small-molecules alone (chemical reprogramming) has superiority over arduous traditional genetic techniques in several aspects. For instance, rapid, transient, and reversible effects in activation and inhibition of functions of specific proteins are of the profits of small-molecules. They are cost-effective, have a long half-life, diversity on structure and function, and allow for temporal and flexible regulation of signaling pathways. Additionally, their effects could be adjusted by fine-tuning concentrations and combinations of different small-molecules. Therefore, chemicals are powerful tools in cell fate conversion and study of stem cell and chemical biology in vitro and in vivo. Moreover, transgene-free and chemical-only transdifferentiation approaches provide alternative strategies for the generation of various cell types, disease modeling, drug screening, and regenerative medicine. The current review gives an overview of the recent findings concerning transdifferentiation by only small-molecules without the use of transgenes.

16.
Differentiation ; 92(1-2): 35-40, 2016.
Article in English | MEDLINE | ID: mdl-27056282

ABSTRACT

Hundreds of transcription factors (TFs) are expressed and work in each cell type, but the identity of the cells is defined and maintained through the activity of a small number of core TFs. Existing reprogramming strategies predominantly focus on the ectopic expression of core TFs of an intended fate in a given cell type regardless of the state of native/somatic gene regulatory networks (GRNs) of the starting cells. Interestingly, an important point is that how much products of the reprogramming, transdifferentiation and differentiation (programming) are identical to their in vivo counterparts. There is evidence that shows that direct fate conversions of somatic cells are not complete, with target cell identity not fully achieved. Manipulation of core TFs provides a powerful tool for engineering cell fate in terms of extinguishment of native GRNs, the establishment of a new GRN, and preventing installation of aberrant GRNs. Conventionally, core TFs are selected to convert one cell type into another mostly based on literature and the experimental identification of genes that are differentially expressed in one cell type compared to the specific cell types. Currently, there is not a universal standard strategy for identifying candidate core TFs. Remarkably, several biological computational platforms are developed, which are capable of evaluating the fidelity of reprogramming methods and refining existing protocols. The current review discusses some deficiencies of reprogramming technologies in the production of a pure population of authentic target cells. Furthermore, it reviews the role of computational approaches (e.g. CellNet, KeyGenes, Mogrify, etc.) in improving (re)programming methods and consequently in regenerative medicine and cancer therapeutics.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Computational Biology/methods , Gene Regulatory Networks , Neoplasms/therapy , Regenerative Medicine , Animals , Humans , Neoplasms/genetics , Neoplasms/metabolism
17.
Cell Regen ; 4: 10, 2015.
Article in English | MEDLINE | ID: mdl-26566431

ABSTRACT

Induced pluripotent stem cells are powerful tools for disease modeling, drug screening, and cell transplantation therapies. These cells can be generated directly from somatic cells by ectopic expression of defined factors through a reprogramming process. However, pluripotent reprogramming is an inefficient process because of various defined and unidentified barriers. Recent studies dissecting the molecular mechanisms of reprogramming have methodically improved the quality, ease, and efficiency of reprogramming. Different strategies have been applied for enhancing reprogramming efficiency, including depletion/inhibition of barriers (p53, p21, p57, p16(Ink4a)/p19(Arf), Mbd3, etc.), overexpression of enhancing genes (e.g., FOXH1, C/EBP alpha, UTF1, and GLIS1), and administration of certain cytokines and small molecules. The current review provides an in-depth overview of the cutting-edge findings regarding distinct barriers of reprogramming to pluripotency and strategies to enhance reprogramming efficiency. By incorporating the mechanistic insights from these recent findings, a combined method of inhibition of roadblocks and application of enhancing factors may yield the most reliable and effective approach in pluripotent reprogramming.

18.
Dent Res J (Isfahan) ; 11(2): 244-50, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24932197

ABSTRACT

BACKGROUND: Dental pulp stem cells can be used in regenerative endodontic therapy. The aim of this study was to introduce an efficient method for dental pulp stem cells isolation. MATERIALS AND METHODS: In this in-vitro study, 60 extracted human third molars were split and pulp tissue was extracted. Dental pulp stem cells were isolated by the following three different methods: (1) digestion of pulp by collagenase/dispase enzyme and culture of the released cells; (2) outgrowth of the cells by culture of undigested pulp pieces; (3) digestion of pulp tissue pieces and fixing them. The cells were cultured in minimum essential medium alpha modification (αMEM) medium supplemented with 20% fetal bovine serum(FBS) in humid 37°C incubator with 5% CO 2. The markers of stem cells were studied by reverse transcriptase polymerase chain reaction (PCR). The student t-test was used for comparing the means of independent groups. P <0.05 was considered as significant. RESULTS: The results indicated that by the first method a few cell colonies with homogenous morphology were detectable after 4 days, while in the outgrowth method more time was needed (10-12 days) to allow sufficient numbers of heterogeneous phenotype stem cells to migrate out of tissue. Interestingly, with the improved third method, we obtained stem cells successfully with about 60% efficiency after 2 days. The results of RT-PCR suggested the expression of Nanog, Oct-4, and Nucleostemin markers in the isolated cells from dental pulps. CONCLUSION: This study proposes a new method with high efficacy to obtain dental pulp stem cells in a short time.

19.
Clin Breast Cancer ; 7(10): 775-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-18021478

ABSTRACT

BACKGROUND: The frequency of aromatase inhibitor (AI)-associated arthralgia and/or bone pain in clinical practice is not known. PATIENTS AND METHODS: Fifty-six consecutive patients with breast cancer not on clinical trials who were receiving AIs in a clinical practice were interviewed regarding occurrence of worsening or new arthralgia and/or bone pain after starting AI therapy. The occurrence, character, severity, and resolution of pain were evaluated. RESULTS: Arthralgia and/or bone pain was reported in 61% of patients. It was severe in 30%, continuous in 41%, central in 50%, peripheral in 79%, and resulted in discontinuation of the drug in 20% of patients. Effective therapies in controlling pain were acetaminophen, 29%; nonsteroidal anti-inflammatory drugs, 50%; opiates, 18%, and glucosamine in 15% of patients. Despite this, 20% of patients discontinued AI therapy because of pain. CONCLUSION: Aromatase inhibitor-associated pain is more frequent in patients not in clinical trials than previously appreciated in clinical trials. Improved patient education is needed, and prompt therapeutic management of pain is required to ensure continued drug treatment and improved quality of life.


Subject(s)
Aromatase Inhibitors/adverse effects , Arthralgia/chemically induced , Arthralgia/epidemiology , Breast Neoplasms/drug therapy , Pain/chemically induced , Pain/epidemiology , Aromatase Inhibitors/therapeutic use , Bone and Bones/pathology , Clinical Trials as Topic , Female , Humans
20.
Cancer ; 101(12): 2727-36, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15526319

ABSTRACT

BACKGROUND: Cytokines have been implicated in diverse processes that are relevant to pancreatic carcinoma, including cachexia, asthenia, and tumor growth. The objective of this study was to examine the association between serum levels of proinflammatory and antiinflammatory or angiogenic cytokines and the outcomes of patients with pancreatic carcinoma. METHODS: Serum cytokine levels were measured by enzyme-linked immunosorbent assay from 51 patients with pancreatic carcinoma and from 48-62 healthy volunteers. Cytokine levels were compared with disease manifestations and overall survival. RESULTS: Circulating levels of vascular endothelial growth factor, tumor necrosis factor alpha, interleukin-1alpha (IL-1alpha), and IL-1beta were not elevated significantly in patients with pancreatic carcinoma, but levels of IL-6, IL-8, IL-10, and IL-1 receptor antagonist (IL-1RA) were elevated significantly (P <0.05). Cytokine levels were dichotomized based on an analysis of null Martingale residuals. Patients who had IL-6 levels > 5.2 pg/mL or IL-10 levels >9.8 pg/mL had significantly worse survival compared with patients who had lower IL-6 or IL-10 levels (P <0.05). IL-8 levels were not associated with survival differences. Patients who had IL-1RA levels <159 pg/mL had significantly worse survival compared with patients who had higher IL-1RA levels (P <0.05). Higher IL-6, IL-10, and IL-8 levels were associated with poor performance status and/or weight loss. In multivariate analysis, only T4 tumors and high IL-6 levels were selected as independent prognostic factors for poor survival. CONCLUSIONS: Circulating levels of several cytokines were high in patients with pancreatic carcinoma, and their association with weight loss and poor performance status suggested that they may be involved in these disease manifestations. Furthermore, serum cytokine levels, in particular IL-6, may be a useful prognostic marker.


Subject(s)
Carcinoma/blood , Cytokines/blood , Pancreatic Neoplasms/blood , Adult , Aged , Carcinoma/mortality , Female , Humans , Interleukin-1/blood , Interleukin-10/blood , Interleukin-6/blood , Interleukin-8/blood , Karnofsky Performance Status , Male , Middle Aged , Multivariate Analysis , Pancreatic Neoplasms/mortality , Prognosis , Receptors, Interleukin-1/antagonists & inhibitors , Survival Rate , Weight Loss
SELECTION OF CITATIONS
SEARCH DETAIL
...