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

Publication year range
1.
Genes Dev ; 35(13-14): 1020-1034, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34168041

ABSTRACT

During mitosis, chromatin condensation is accompanied by a global arrest of transcription. Recent studies suggest transcriptional reactivation upon mitotic exit occurs in temporally coordinated waves, but the underlying regulatory principles have yet to be elucidated. In particular, the contribution of sequence-specific transcription factors (TFs) remains poorly understood. Here we report that Brn2, an important regulator of neural stem cell identity, associates with condensed chromatin throughout cell division, as assessed by live-cell imaging of proliferating neural stem cells. In contrast, the neuronal fate determinant Ascl1 dissociates from mitotic chromosomes. ChIP-seq analysis reveals that Brn2 mitotic chromosome binding does not result in sequence-specific interactions prior to mitotic exit, relying mostly on electrostatic forces. Nevertheless, surveying active transcription using single-molecule RNA-FISH against immature transcripts reveals differential reactivation kinetics for key targets of Brn2 and Ascl1, with transcription onset detected in early (anaphase) versus late (early G1) phases, respectively. Moreover, by using a mitotic-specific dominant-negative approach, we show that competing with Brn2 binding during mitotic exit reduces the transcription of its target gene Nestin Our study shows an important role for differential binding of TFs to mitotic chromosomes, governed by their electrostatic properties, in defining the temporal order of transcriptional reactivation during mitosis-to-G1 transition.


Subject(s)
Mitosis , Neural Stem Cells , Chromatin , Chromosomes/metabolism , Mitosis/genetics , Neural Stem Cells/metabolism , Transcription Factors/metabolism
2.
Genes Dev ; 33(5-6): 310-332, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30804224

ABSTRACT

Whether cell types exposed to a high level of environmental insults possess cell type-specific prosurvival mechanisms or enhanced DNA damage repair capacity is not well understood. BRN2 is a tissue-restricted POU domain transcription factor implicated in neural development and several cancers. In melanoma, BRN2 plays a key role in promoting invasion and regulating proliferation. Here we found, surprisingly, that rather than interacting with transcription cofactors, BRN2 is instead associated with DNA damage response proteins and directly binds PARP1 and Ku70/Ku80. Rapid PARP1-dependent BRN2 association with sites of DNA damage facilitates recruitment of Ku80 and reprograms DNA damage repair by promoting Ku-dependent nonhomologous end-joining (NHEJ) at the expense of homologous recombination. BRN2 also suppresses an apoptosis-associated gene expression program to protect against UVB-, chemotherapy- and vemurafenib-induced apoptosis. Remarkably, BRN2 expression also correlates with a high single-nucleotide variation prevalence in human melanomas. By promoting error-prone DNA damage repair via NHEJ and suppressing apoptosis of damaged cells, our results suggest that BRN2 contributes to the generation of melanomas with a high mutation burden. Our findings highlight a novel role for a key transcription factor in reprogramming DNA damage repair and suggest that BRN2 may impact the response to DNA-damaging agents in BRN2-expressing cancers.


Subject(s)
Apoptosis , DNA End-Joining Repair/genetics , Homeodomain Proteins/metabolism , Melanoma/genetics , Melanoma/physiopathology , Mutation/genetics , POU Domain Factors/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/genetics , Humans , Ku Autoantigen/metabolism , POU Domain Factors/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Binding , Protein Domains , Protein Transport
3.
Am J Hum Genet ; 110(6): 998-1007, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37207645

ABSTRACT

While common obesity accounts for an increasing global health burden, its monogenic forms have taught us underlying mechanisms via more than 20 single-gene disorders. Among these, the most common mechanism is central nervous system dysregulation of food intake and satiety, often accompanied by neurodevelopmental delay (NDD) and autism spectrum disorder. In a family with syndromic obesity, we identified a monoallelic truncating variant in POU3F2 (alias BRN2) encoding a neural transcription factor, which has previously been suggested as a driver of obesity and NDD in individuals with the 6q16.1 deletion. In an international collaboration, we identified ultra-rare truncating and missense variants in another ten individuals sharing autism spectrum disorder, NDD, and adolescent-onset obesity. Affected individuals presented with low-to-normal birth weight and infantile feeding difficulties but developed insulin resistance and hyperphagia during childhood. Except for a variant leading to early truncation of the protein, identified variants showed adequate nuclear translocation but overall disturbed DNA-binding ability and promotor activation. In a cohort with common non-syndromic obesity, we independently observed a negative correlation of POU3F2 gene expression with BMI, suggesting a role beyond monogenic obesity. In summary, we propose deleterious intragenic variants of POU3F2 to cause transcriptional dysregulation associated with hyperphagic obesity of adolescent onset with variable NDD.


Subject(s)
Autism Spectrum Disorder , Neurodevelopmental Disorders , Prader-Willi Syndrome , Adolescent , Humans , Autism Spectrum Disorder/genetics , Hyperphagia/genetics , Hyperphagia/complications , Neurodevelopmental Disorders/genetics , Obesity/complications , Prader-Willi Syndrome/complications , Prader-Willi Syndrome/genetics , Proteins
4.
Glia ; 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39215540

ABSTRACT

Schwann cells are critical for the proper development and function of the peripheral nervous system (PNS), where they form a collaborative relationship with axons. Past studies highlighted that a pair of proteins called the prohibitins play major roles in Schwann cell biology. Prohibitins are ubiquitously expressed and versatile proteins. We have previously shown that while prohibitins play a crucial role in Schwann cell mitochondria for long-term myelin maintenance and axon health, they may also be present at the Schwann cell-axon interface during development. Here, we expand on this, showing that drug-mediated modulation of prohibitins in vitro disrupts myelination and confirming that Schwann cell-specific ablation of prohibitin 2 (Phb2) in vivo results in severe defects in radial sorting and myelination. We show in vivo that Phb2-null Schwann cells cannot effectively proliferate and the transcription factors EGR2 (KROX20), POU3F1 (OCT6), and POU3F2 (BRN2), necessary for proper Schwann cell maturation, are dysregulated. Schwann cell-specific deletion of Jun, a transcription factor associated with negative regulation of myelination, confers partial rescue of the developmental defect seen in mice lacking Schwann cell Phb2. Finally, we identify a pool of candidate PHB2 interactors that change their interaction with PHB2 depending on neuronal signals, and thus are potential mediators of PHB2-associated developmental defects. This work develops our understanding of Schwann cell biology, revealing that Phb2 may modulate the timely expression of transcription factors necessary for proper PNS development, and proposing candidates that may play a role in PHB2-mediated integration of axon signals in the Schwann cell.

5.
Mol Biol (Mosk) ; 55(5): 707-733, 2021.
Article in Russian | MEDLINE | ID: mdl-34671001

ABSTRACT

Direct reprogramming technology allows several specific types of cells, including specialized neurons, to be obtained from readily available autologous somatic cells. It presents unique opportunities for the development of personalized medicine, from in vitro models of hereditary and degenerative neurological diseases to novel neuroregenerative technologies. Over the past decade, a plethora of protocols for primary reprogramming has been published, yet reproducible generation of homogeneous populations of neuronally reprogrammed cells still remains a challenge. All existing protocols, however, use transcription factors that are involved in embryonic neurogenesis. This is presumably be the key issue for obtaining highly efficient and reproducible protocols for ex vivo neurogenesis. Analysis of the functional features of transcription factors in embryonic and adult neurogenesis may not only lead to the improvement of reprogramming protocols, but also, via cell marker analysis, can exactly determine the stage of neurogenesis that a particular protocol will reach. The purpose of this review is to characterize the general factors that play key roles in neurogenesis for the embryonic and adult periods, as well as in cellular reprogramming, and to assess correspondence of cell forms obtained as a result of cellular reprogramming to the ontogenetic series of the nervous system, from pluripotent stem cells to specialized neurons.


Subject(s)
Cellular Reprogramming , Transcription Factors , Cellular Reprogramming/genetics , Neurons , Transcription Factors/genetics
6.
Am J Hum Genet ; 98(2): 363-72, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26833329

ABSTRACT

Genetic studies of intellectual disability and identification of monogenic causes of obesity in humans have made immense contribution toward the understanding of the brain and control of body mass. The leptin > melanocortin > SIM1 pathway is dysregulated in multiple monogenic human obesity syndromes but its downstream targets are still unknown. In ten individuals from six families, with overlapping 6q16.1 deletions, we describe a disorder of variable developmental delay, intellectual disability, and susceptibility to obesity and hyperphagia. The 6q16.1 deletions segregated with the phenotype in multiplex families and were shown to be de novo in four families, and there was dramatic phenotypic overlap among affected individuals who were independently ascertained without bias from clinical features. Analysis of the deletions revealed a ∼350 kb critical region on chromosome 6q16.1 that encompasses a gene for proneuronal transcription factor POU3F2, which is important for hypothalamic development and function. Using morpholino and mutant zebrafish models, we show that POU3F2 lies downstream of SIM1 and controls oxytocin expression in the hypothalamic neuroendocrine preoptic area. We show that this finding is consistent with the expression patterns of POU3F2 and related genes in the human brain. Our work helps to further delineate the neuro-endocrine control of energy balance/body mass and demonstrates that this molecular pathway is conserved across multiple species.


Subject(s)
Homeodomain Proteins/genetics , Intellectual Disability/genetics , Obesity/genetics , POU Domain Factors/genetics , Sequence Deletion , Adolescent , Adult , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Mass Index , Cell Line , Child , Child, Preschool , Chromosomes, Human, Pair 6/genetics , Disease Models, Animal , Energy Metabolism , Female , Homeodomain Proteins/metabolism , Humans , Hypothalamus/metabolism , Male , Middle Aged , Oxytocin/metabolism , POU Domain Factors/metabolism , Pedigree , Phenotype , Repressor Proteins/genetics , Repressor Proteins/metabolism , Young Adult , Zebrafish
7.
Biochem Biophys Res Commun ; 490(3): 901-905, 2017 08 26.
Article in English | MEDLINE | ID: mdl-28648603

ABSTRACT

Eccrine sweat glands regulate body temperature by secreting water and electrolytes. In humans, eccrine sweat glands are ubiquitous in the skin, except in the lips and external genitalia. In mice, eccrine sweat glands are present only in the paw pad. Brn2 is a protein belonging to a large family of transcription factors. A few studies have examined Brn2 in melanoma cells and epidermal keratinocytes. This study investigated changes in the skin in the K5-Brn2 transgenic mouse, which overexpresses Brn2 and contains the keratin 5 promotor. Interestingly, the volume of eccrine sweat glands was reduced markedly in the K5-Brn2 transgenic mouse compared with the wild-type, while the expression of aquaporin 5, important molecule in sweat secretion, was increased in each sweat gland cell, probably to compensate for the reduction in gland development. However, sweating response to a pilocarpine injection in the hind paw was significantly decreased in the K5-Brn2 transgenic mouse compared with the wild-type. The paw epidermis was thicker in the K5-Brn2 transgenic mouse compared with the wild-type. Taken together, eccrine sweat gland development and sweat secretion were suppressed markedly in the K5-Brn2 transgenic mouse. These results may be associated with dominant development of the epidermis by Brn2 overexpression in the paw skin.


Subject(s)
Eccrine Glands/growth & development , Epidermis/growth & development , Nerve Tissue Proteins/genetics , POU Domain Factors/genetics , Up-Regulation , Animals , Eccrine Glands/physiology , Epidermis/physiology , Humans , Keratin-5/genetics , Mice , Mice, Transgenic , Organ Size , Promoter Regions, Genetic , Sweating
8.
Mol Carcinog ; 56(1): 49-61, 2017 01.
Article in English | MEDLINE | ID: mdl-26878440

ABSTRACT

Indole-3-carbinol (I3C), an anti-cancer phytochemical derived from cruciferous vegetables, strongly inhibited proliferation and down-regulated protein levels of the melanocyte master regulator micropthalmia-associated transcription factor (MITF-M) in oncogenic BRAF-V600E expressing melanoma cells in culture as well as in vivo in tumor xenografted athymic nude mice. In contrast, wild type BRAF-expressing melanoma cells remained relatively insensitive to I3C anti-proliferative signaling. In BRAF-V600E-expressing melanoma cells, I3C treatment inhibited phosphorylation of MEK and ERK/MAPK, the down stream effectors of BRAF. The I3C anti-proliferative arrest was concomitant with the down-regulation of MITF-M transcripts and promoter activity, loss of endogenous BRN-2 binding to the MITF-M promoter, and was strongly attenuated by expression of exogenous MITF-M. Importantly, in vitro kinase assays using immunoprecipitated BRAF-V600E and wild type BRAF demonstrated that I3C selectively inhibited the enzymatic activity of the oncogenic BRAF-V600E but not of the wild type protein. In silico modeling predicted an I3C interaction site in the BRAF-V600E protomer distinct from where the clinically used BRAF-V600E inhibitor Vemurafenib binds to BRAF-V600E. Consistent with this prediction, combinations of I3C and Vemurafenib more potently inhibited melanoma cell proliferation and reduced MITF-M levels in BRAF-V600E expressing melanoma cells compared to the effects of each compound alone. Thus, our results demonstrate that oncogenic BRAF-V600E is a new cellular target of I3C that implicate this indolecarbinol compound as a potential candidate for novel single or combination therapies for melanoma. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Indoles/therapeutic use , Melanoma/drug therapy , Microphthalmia-Associated Transcription Factor/genetics , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 4/genetics , Indoles/pharmacology , Melanoma/genetics , Melanoma/pathology , Mice, Nude , Point Mutation , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Vemurafenib
9.
J Biol Chem ; 289(8): 5250-60, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24385434

ABSTRACT

Neuronal restricted progenitors (NRPs) represent a type of transitional intermediate cells that lie between multipotent neural progenitors and terminal differentiated neurons during neurogenesis. These NRPs have the ability to self-renew and differentiate into neurons, but not into glial cells, which is considered an advantage for cellular therapy of human neurodegenerative diseases. However, difficulty in the extraction of highly purified NRPs from normal nervous tissue prevents further studies and applications. In this study, we report the conversion of human fetal fibroblasts into human induced NRPs (hiNRPs) in 11 days by using just three defined factors: Sox2, c-Myc, and either Brn2 or Brn4. The hiNRPs exhibited distinct neuronal characteristics, including cell morphology, multiple neuronal marker expression, self-renewal capacity, and a genome-wide transcriptional profile. Moreover, hiNRPs were able to differentiate into various terminal neurons with functional membrane properties but not glial cells. Direct generation of hiNRPs from somatic cells will provide a new source of cells for cellular replacement therapy of human neurodegenerative diseases.


Subject(s)
Fibroblasts/cytology , Neural Stem Cells/cytology , Neurons/cytology , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Cellular Reprogramming , Gene Expression Profiling , Genome, Human/genetics , Humans , Mice , Neural Stem Cells/transplantation , Stem Cell Transplantation , Transcription Factors/metabolism
10.
Exp Dermatol ; 24(7): 481-4, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25866058

ABSTRACT

Microphthalmia-associated transcription factor (MITF) is a nodal point in melanoma transcriptional network that regulates dozens of genes with critical functions in cell differentiation, proliferation and survival. Highly variable MITF expression levels exist in tumor cell subpopulations conferring marked heterogeneity and plasticity in the tumor tissue. A model has been postulated whereby lower MITF levels favour cell invasion and suppress proliferation, whereas high levels stimulate differentiation and proliferation. Additionally, MITF is considered to be a prosurvival gene and a lineage addiction oncogene in melanoma. Herein, we review how MITF expression may affect the melanoma phenotype with consequences on the survival, invasion and metastasis of melanoma cells, and we discuss the research challenges.


Subject(s)
Melanoma/metabolism , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Apoptosis , Cell Proliferation , Cell Survival , Epithelial-Mesenchymal Transition , Gene Expression , Homeodomain Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , MAP Kinase Signaling System , Melanoma/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Models, Biological , Neoplasm Invasiveness , Nuclear Proteins/metabolism , POU Domain Factors/metabolism , Protein Stability , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , SOXB1 Transcription Factors/metabolism , Skin Neoplasms/genetics , Tumor Microenvironment , Zinc Finger Protein Gli2
11.
Pathol Int ; 64(8): 365-74, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25143124

ABSTRACT

In lung tumors, the association between carcinoids and high-grade neuroendocrine tumors (HGNETs) is controversial. To understand the phenotypic similarities/differences between lung carcinoids and HGNETs, we comparatively investigated the expression of three kinds of developing neural transcription factors (DNTFs: BRN2, TTF1 and ASCL1) and multiple endocrine neoplasia type 1 (MEN1) as well as RB1 and P53 using 18 carcinoids and 16 HGNETs. The DNTFs were expressed in 10 of the 18 carcinoids and in all the HGNETs, while normal neuroendocrine cells, which are considered the major cell origin of lung carcinoids and small cell carcinomas, did not express DNTFs. Both the DNTF(-) and DNTF(+) carcinoids contained typical and atypical carcinoids. All the DNTF(-) carcinoids examined were formed in the bronchial wall. All the MEN1(-) carcinoids examined were classified into the DNTF(-) carcinoids, while all the HGNETs expressed MEN1. This finding suggests that DNTF(-) MEN1(-) carcinoids are unlikely to be precursors of HGNETs. Although the status of RB1 and P53 between carcinoids and HGNETs were apparently different, the DNTF(+) carcinoids of two male patients and one female patient revealed morphologies resembling HGNET cells and relatively high Ki67 indices. Further investigation of DNTF expression in carcinoids might provide important clues to understand the association between carcinoids and HGNETs.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoid Tumor/genetics , Lung Neoplasms/genetics , Transcription Factors/genetics , Adult , Aged , Aged, 80 and over , Carcinoid Tumor/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Grading/methods
12.
Anticancer Res ; 44(1): 71-84, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38160013

ABSTRACT

BACKGROUND/AIM: The Brain-Specific Homeobox/POU Domain Protein 2 (BRN2) transcription factor supports melanoma progression by regulating the expression of several genes involved in cell migration and invasion. We hypothesized that a peptide designed based on the POU domain of BRN2 could block the BRN2 transcription activity and, consequently, reduce metastasis. MATERIALS AND METHODS: Cell viability was accessed by Trypan Blue exclusion dye assay and xCelligence platform. Wound-healing scratch assay and transwell invasion with matrigel membrane assay were performed to analyze cell migration and invasion. The internalization mechanism of the L13S peptide was investigated using confocal microscopy and wound-healing scratch assay. The impact of L13S on cell protein expression was analyzed through western blotting. In vivo assays were conducted to evaluate the protective effect and toxicity of L13S in a metastatic model using murine melanoma cells. RESULTS: Here, we show that the peptide named L13S can inhibit the migration and invasion of murine melanoma cells (B16F10-Nex2) as well as the migration of human melanoma cells (SK-MEL-25 and A375) by regulating the expression of proteins involved in motility. Mechanistically, we found that L13S is internalized by murine melanoma cells via macropinocytosis and binds actin filaments and nuclei. More importantly, in vivo studies indicated that the peptide was able to significantly inhibit lung metastasis in syngeneic models without off-target effects and with virtually no cytotoxicity toward normal organs. CONCLUSION: L13S peptide is a strong candidate for further development as an anticancer agent for the treatment of melanoma metastasis.


Subject(s)
Antineoplastic Agents , Melanoma , Humans , Mice , Animals , Melanoma/pathology , Antineoplastic Agents/pharmacology , Peptides/pharmacology , Peptides/therapeutic use , Cell Movement , Cell Line, Tumor , Cell Proliferation , Neoplasm Invasiveness
13.
bioRxiv ; 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38562812

ABSTRACT

Schwann cells are critical for the proper development and function of the peripheral nervous system, where they form a mutually beneficial relationship with axons. Past studies have highlighted that a pair of proteins called the prohibitins play major roles in Schwann cell biology. Prohibitins are ubiquitously expressed and versatile proteins. We have previously shown that while prohibitins play a crucial role in Schwann cell mitochondria for long-term myelin maintenance and axon health, they may also be present at the Schwann cell-axon interface during development. Here, we expand on this work, showing that drug-mediated modulation of prohibitins in vitro disrupts myelination and confirming that Schwann cell-specific ablation of prohibitin 2 (Phb2) in vivo results in early and severe defects in peripheral nerve development. Using a proteomic approach in vitro, we identify a pool of candidate PHB2 interactors that change their interaction with PHB2 depending on the presence of axonal signals. Furthermore, we show in vivo that loss of Phb2 in mouse Schwann cells causes ineffective proliferation and dysregulation of transcription factors EGR2 (KROX20), POU3F1 (OCT6) and POU3F2 (BRN2) that are necessary for proper Schwann cell maturation. Schwann cell-specific deletion of Jun, a transcription factor associated with negative regulation of myelination, confers partial rescue of the development defect seen in mice lacking Schwann cell Phb2. This work develops our understanding of Schwann cell biology, revealing that Phb2 may directly or indirectly modulate the timely expression of transcription factors necessary for proper peripheral nervous system development, and proposing candidates that may play a role in PHB2-mediated integration of axon signals in the Schwann cell.

14.
Curr Drug Targets ; 25(1): 46-61, 2024.
Article in English | MEDLINE | ID: mdl-38444255

ABSTRACT

Transcription factors play a crucial role in providing identity to each cell population. To maintain cell identity, it is essential to balance the expression of activator and inhibitor transcription factors. Cell plasticity and reprogramming offer great potential for future therapeutic applications, as they can regenerate damaged tissue. Specific niche factors can modify gene expression and differentiate or transdifferentiate the target cell to the required fate. Ongoing research is being carried out on the possibilities of transcription factors in regenerating neurons, with neural stem cells (NSCs) being considered the preferred cells for generating new neurons due to their epigenomic and transcriptome memory. NEUROD1/ASCL1, BRN2, MYTL1, and other transcription factors can induce direct reprogramming of somatic cells, such as fibroblasts, into neurons. However, the molecular biology of transcription factors in reprogramming and differentiation still needs to be fully understood.


Subject(s)
Brain Regeneration , Neural Stem Cells , Humans , Transcription Factors , Cell Differentiation , Fibroblasts
15.
bioRxiv ; 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39257809

ABSTRACT

Prostate cancer (PCa) is the leading cause death from cancer in men worldwide. Approximately 30% of castrate-resistant PCa's become refractory to therapy due to neuroendocrine differentiation (NED) that is present in <1% of androgen-sensitive tumors. First-in-class imipridone ONC201/TIC10 has shown clinical activity against midline gliomas, neuroendocrine tumors and PCa. We explored the question of whether NED promotes sensitivity to imipridones ONC201 and ONC206 by inducible overexpression of SOX2 and BRN2, well-known neuroendocrine drivers, in human PCa cell lines DU145 or LNCaP. Slight protection from ONC201 or ONC206 with SOX2 and BRN2 overexpression was observed in the inducible LNCaP cells but not in the DU145 cells. At 2 months, there was an apparent increase in CLpP expression in LNCaP SOX2-overexpressing cells but this did not confer enhanced sensitivity to ONC201. DU145 SOX2-overexpressing cells had a significantly reduced ONC201 sensitivity than DU145 control cells. The results support the idea that treatment of castrate-resistant prostate cancer by imipridones may not be significantly impacted by neuroendocrine differentiation as a therapy-resistance mechanism. The results support further testing of imipridones across subtypes of androgen-sensitive and castrate-resistant prostate cancer.

16.
Front Mol Neurosci ; 15: 921901, 2022.
Article in English | MEDLINE | ID: mdl-35935334

ABSTRACT

The cerebellar nuclear (CN) neurons are a molecularly heterogeneous population whose specification into the different cerebellar nuclei is defined by the expression of varying sets of transcription factors. Here, we present a novel molecular marker, Pou3f1, that delineates specific sets of glutamatergic CN neurons. The glutamatergic identity of Pou3f1+ cells was confirmed by: (1) the co-expression of vGluT2, a cell marker of glutamatergic neurons; (2) the lack of co-expression between Pou3f1 and GAD67, a marker of GABAergic neurons; (3) the co-expression of Atoh1, the master regulator required for the production of all cerebellar glutamatergic lineages; and (4) the absence of Pou3f1-expressing cells in the Atoh1-null cerebellum. Furthermore, the lack of Pax6 and Tbr1 expression in Pou3f1+ cells reveals that Pou3f1-expressing CN neurons specifically settle in the interposed and dentate nuclei. In addition, the Pou3f1-labeled glutamatergic CN neurons can be further classified by the expression of Brn2 and Irx3. The results of the present study align with previous findings highlighting that the survival of the interposed and dentate CN neurons is largely independent of Pax6. More importantly, the present study extends the field's collective knowledge of the molecular diversity of cerebellar nuclei.

17.
Methods Mol Biol ; 2352: 1-12, 2021.
Article in English | MEDLINE | ID: mdl-34324176

ABSTRACT

Forced expression of specific neuronal transcription factors in mouse embryonic fibroblasts (MEFs) can lead to their direct conversion into functional neurons. Direct neuronal reprogramming has become a powerful tool to characterize individual factors and molecular mechanisms involved in forced and normal neurogenesis and to generate neuronal cell types for in vitro studies. Here we provide a detailed protocol for the isolation of MEFs devoid of neural tissue and their direct reprogramming into functional neurons by overexpression of neuronal reprogramming factors (Ascl1, Brn2, and Myt1l) using lentiviral vectors. This method enables quick and efficient generation of mouse neurons in vitro for versatile functional and mechanistic characterization.


Subject(s)
Cell Separation/methods , Cellular Reprogramming Techniques , Cellular Reprogramming , Fibroblasts/cytology , Fibroblasts/metabolism , Neurons/cytology , Neurons/metabolism , Animals , Cell Culture Techniques , Cell Differentiation/genetics , Cellular Reprogramming/genetics , Cryopreservation , Genetic Vectors/biosynthesis , Genetic Vectors/genetics , Humans , Mice , Neurogenesis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transduction, Genetic
18.
Cancers (Basel) ; 13(3)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513758

ABSTRACT

In the current study, we demonstrate that integrin α3ß1 promotes invasive and metastatic traits of triple-negative breast cancer (TNBC) cells through induction of the transcription factor, Brain-2 (Brn-2). We show that RNAi-mediated suppression of α3ß1 in MDA-MB-231 cells caused reduced expression of Brn-2 mRNA and protein and reduced activity of the BRN2 gene promoter. In addition, RNAi-targeting of Brn-2 in MDA-MB-231 cells decreased invasion in vitro and lung colonization in vivo, and exogenous Brn-2 expression partially restored invasion to cells in which α3ß1 was suppressed. α3ß1 promoted phosphorylation of Akt in MDA-MB-231 cells, and treatment of these cells with a pharmacological Akt inhibitor (MK-2206) reduced both Brn-2 expression and cell invasion, indicating that α3ß1-Akt signaling contributes to Brn-2 induction. Analysis of RNAseq data from patients with invasive breast carcinoma revealed that high BRN2 expression correlates with poor survival. Moreover, high BRN2 expression positively correlates with high ITGA3 expression in basal-like breast cancer, which is consistent with our experimental findings that α3ß1 induces Brn-2 in TNBC cells. Together, our study demonstrates a pro-invasive/pro-metastatic role for Brn-2 in breast cancer cells and identifies a role for integrin α3ß1 in regulating Brn-2 expression, thereby revealing a novel mechanism of integrin-dependent breast cancer cell invasion.

19.
Cancer Cell ; 37(4): 584-598.e11, 2020 04 13.
Article in English | MEDLINE | ID: mdl-32220301

ABSTRACT

Metastatic prostate cancer is characterized by recurrent genomic copy number alterations that are presumed to contribute to resistance to hormone therapy. We identified CHD1 loss as a cause of antiandrogen resistance in an in vivo small hairpin RNA (shRNA) screen of 730 genes deleted in prostate cancer. ATAC-seq and RNA-seq analyses showed that CHD1 loss resulted in global changes in open and closed chromatin with associated transcriptomic changes. Integrative analysis of this data, together with CRISPR-based functional screening, identified four transcription factors (NR3C1, POU3F2, NR2F1, and TBX2) that contribute to antiandrogen resistance, with associated activation of non-luminal lineage programs. Thus, CHD1 loss results in chromatin dysregulation, thereby establishing a state of transcriptional plasticity that enables the emergence of antiandrogen resistance through heterogeneous mechanisms.


Subject(s)
Androgen Antagonists/pharmacology , Chromatin/genetics , DNA Helicases/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , RNA, Small Interfering/genetics , Receptors, Androgen/chemistry , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Chromatin/metabolism , DNA Helicases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , High-Throughput Screening Assays , Humans , Male , Mice , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Pigment Cell Melanoma Res ; 32(2): 280-291, 2019 03.
Article in English | MEDLINE | ID: mdl-30277012

ABSTRACT

The BRAF kinase and the MAPK pathway are targets of current melanoma therapies. However, MAPK pathway inhibition results in dynamic changes of downstream targets that can counteract inhibitor-action not only in during treatment, but also in acquired resistant tumours. One such dynamic change involves the expression of the transcription factor MITF, a crucial regulator of cell survival and proliferation in untreated as well as drug-addicted acquired resistant melanoma. Tight control over MITF expression levels is required for optimal melanoma growth, and while it is well established that the MAPK pathway regulates MITF expression, the actual mechanism is insufficiently understood. We reveal here, how BRAF through action on the transcription factors BRN2 and PAX3 executes control over the regulation of MITF expression in a manner that allows for considerable plasticity. This plasticity provides robustness to the BRAF mediated MITF regulation and explains the dynamics in MITF expression that are observed in patients in response to MAPK inhibitor therapy.


Subject(s)
Homeodomain Proteins/metabolism , Melanoma/genetics , Microphthalmia-Associated Transcription Factor/genetics , PAX3 Transcription Factor/metabolism , POU Domain Factors/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Base Sequence , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/metabolism , Phenotype , Protein Kinase Inhibitors/pharmacology , Axl Receptor Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL