63 results on '"Ivan Nalvarte"'
Search Results
2. Proof-of-Concept for Long-Term Human Endometrial Epithelial Organoids in Modeling Menstrual Cycle Responses
- Author
-
Yanyu Jiang, Arturo Reyes Palomares, Patricia Munoz, Ivan Nalvarte, Ganesh Acharya, Jose Inzunza, Mukesh Varshney, and Kenny Alexandra Rodriguez-Wallberg
- Subjects
endometrium growth ,sequential hormone treatment ,organoid culture ,proliferative phase ,secretory phase ,estrogen ,Cytology ,QH573-671 - Abstract
Endometrial disorders, such as infertility and endometriosis, significantly impact reproductive health, thus necessitating better models to study endometrial function. Current in vitro models fail to replicate the complexity of the human endometrium throughout the entire menstrual cycle. This study aimed to assess the physiological response of human endometrial organoids (hEOs) to in vitro hormonal treatments designed to mimic the hormonal fluctuations of the menstrual cycle. Endometrial biopsies from three healthy women were used to develop hEOs, which were treated over 28 days with three hormonal stimulation strategies: (1) estrogen only (E) to mimic the proliferative phase, (2) the addition of progesterone (EP) to simulate the secretory phase, and (3) the further addition of cAMP (EPC) to enhance the secretory functions of hEOs. Gene and protein expression were analyzed using qPCR, IHC, and ELISA. The hEOs exhibited proliferation, gland formation, and appropriate expression of markers such as E-cadherin and Ki67. The hormonal treatments induced significant changes in PR, HSD17B1, PAEP, SPP1, and other genes relevant to endometrial function, closely mirroring in vivo physiological responses. The prominent changes were observed in EPC-treated hEOs (week 4) with significantly high expression of uterine milk components such as glycodelin (PAEP) and osteopontin (SPP1), reflecting mid- to late-secretory phase physiology. This model successfully recapitulates human menstrual cycle dynamics and offers a promising platform for studying endometrial disorders and advancing personalized treatments in gynecology.
- Published
- 2024
- Full Text
- View/download PDF
3. Harnessing the reverse cholesterol transport pathway to favor differentiation of monocyte-derived APCs and antitumor responses
- Author
-
Laura Raccosta, Maura Marinozzi, Susan Costantini, Daniela Maggioni, Lorena Maria Ferreira, Gianfranca Corna, Paola Zordan, Angela Sorice, Diego Farinello, Silvia Bianchessi, Michela Riba, Dejan Lazarevic, Paolo Provero, Matthias Mack, Attilio Bondanza, Ivan Nalvarte, J-A Gustafsson, Valeria Ranzani, Francesco De Sanctis, Stefano Ugel, Silvère Baron, Jean-Marc A. Lobaccaro, Lorenzo Pontini, Manuela Pacciarini, Catia Traversari, Massimiliano Pagani, Vincenzo Bronte, Giovanni Sitia, Per Antonson, Andrea Brendolan, Alfredo Budillon, and Vincenzo Russo
- Subjects
Cytology ,QH573-671 - Abstract
Abstract Lipid and cholesterol metabolism play a crucial role in tumor cell behavior and in shaping the tumor microenvironment. In particular, enzymatic and non-enzymatic cholesterol metabolism, and derived metabolites control dendritic cell (DC) functions, ultimately impacting tumor antigen presentation within and outside the tumor mass, dampening tumor immunity and immunotherapeutic attempts. The mechanisms accounting for such events remain largely to be defined. Here we perturbed (oxy)sterol metabolism genetically and pharmacologically and analyzed the tumor lipidome landscape in relation to the tumor-infiltrating immune cells. We report that perturbing the lipidome of tumor microenvironment by the expression of sulfotransferase 2B1b crucial in cholesterol and oxysterol sulfate synthesis, favored intratumoral representation of monocyte-derived antigen-presenting cells, including monocyte-DCs. We also found that treating mice with a newly developed antagonist of the oxysterol receptors Liver X Receptors (LXRs), promoted intratumoral monocyte-DC differentiation, delayed tumor growth and synergized with anti-PD-1 immunotherapy and adoptive T cell therapy. Of note, looking at LXR/cholesterol gene signature in melanoma patients treated with anti-PD-1-based immunotherapy predicted diverse clinical outcomes. Indeed, patients whose tumors were poorly infiltrated by monocytes/macrophages expressing LXR target genes showed improved survival over the course of therapy. Thus, our data support a role for (oxy)sterol metabolism in shaping monocyte-to-DC differentiation, and in tumor antigen presentation critical for responsiveness to immunotherapy. The identification of a new LXR antagonist opens new treatment avenues for cancer patients.
- Published
- 2023
- Full Text
- View/download PDF
4. Astrocytes protect dopaminergic neurons against aminochrome neurotoxicity
- Author
-
Juan Segura-Aguilar, Bengt Mannervik, José Inzunza, Mukesh Varshney, Ivan Nalvarte, and Patricia Muñoz
- Subjects
aminochrome ,astrocytes ,dopamine ,dopaminergic neurons ,exosomes ,glutathione transferase m2-2 ,neuroprotection ,parkinson’s disease ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Astrocytes protect neurons by modulating neuronal function and survival. Astrocytes support neurons in several ways. They provide energy through the astrocyte-neuron lactate shuttle, protect neurons from excitotoxicity, and internalize neuronal lipid droplets to degrade fatty acids for neuronal metabolic and synaptic support, as well as by their high capacity for glutamate uptake and the conversion of glutamate to glutamine. A recent reported astrocyte system for protection of dopamine neurons against the neurotoxic products of dopamine, such as aminochrome and other o-quinones, were generated under neuromelanin synthesis by oxidizing dopamine catechol structure. Astrocytes secrete glutathione transferase M2-2 through exosomes that transport this enzyme into dopaminergic neurons to protect these neurons against aminochrome neurotoxicity. The role of this new astrocyte protective mechanism in Parkinson´s disease is discussed.
- Published
- 2022
- Full Text
- View/download PDF
5. Loss of ERβ in Aging LXRαβ Knockout Mice Leads to Colitis
- Author
-
Xiaoyu Song, Wanfu Wu, Yubing Dai, Margaret Warner, Ivan Nalvarte, Per Antonson, Mukesh Varshney, and Jan-Åke Gustafsson
- Subjects
liver X receptor ,estrogen receptor β ,colitis ,inflammatory bowel diseases ,plectin ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Liver X receptors (LXRα and LXRβ) are oxysterol-activated nuclear receptors that play key roles in cholesterol homeostasis, the central nervous system, and the immune system. We have previously reported that LXRαβ-deficient mice are more susceptible to dextran sodium sulfate (DSS)-induced colitis than their WT littermates, and that an LXR agonist protects against colitis in mice mainly via the regulation of the immune system in the gut. We now report that both LXRα and LXRβ are expressed in the colonic epithelium and that in aging LXRαβ−/− mice there is a reduction in the intensity of goblet cells, mucin (MUC2), TFF3, and estrogen receptor β (ERβ) levels. The cytoplasmic compartment of the surface epithelial cells was markedly reduced and there was a massive invasion of macrophages in the lamina propria. The expression and localization of β-catenin, α-catenin, and E-cadherin were not changed, but the shrinkage of the cytoplasm led to an appearance of an increase in staining. In the colonic epithelium there was a reduction in the expression of plectin, a hemidesmosome protein whose loss in mice leads to spontaneous colitis, ELOVL1, a fatty acid elongase protein coding gene whose overexpression is found in colorectal cancer, and non-neuronal choline acetyltransferase (ChAT) involved in the regulation of epithelial cell adhesion. We conclude that in aging LXRαβ−/− mice, the phenotype in the colon is due to loss of ERβ expression.
- Published
- 2023
- Full Text
- View/download PDF
6. Hematopoietic stem cell‐ and induced pluripotent stem cell‐derived CAR‐NK cells as reliable cell‐based therapy solutions
- Author
-
Jonathan Arias, Jingwei Yu, Mukesh Varshney, Jose Inzunza, and Ivan Nalvarte
- Subjects
CAR‐NK ,chimeric antigen receptor (CAR) ,hematopoietic stem cell (HSC) ,induced pluripotent stem (iPS) cell ,natural killer cells (NK) ,Medicine (General) ,R5-920 ,Cytology ,QH573-671 - Abstract
Abstract Hematopoietic stem cell‐ (HSC) and induced pluripotent stem (iPS) cell‐derived natural killer (NK) cells containing engineered functions, such as chimeric antigen receptors (CAR), offer great promise for the treatment of seemingly incurable oncological malignancies. Today, some of the main challenges of CAR cell‐based therapeutics are the long manufacturing time and safety of the cell sources used. Additional challenges include avoiding graft vs host disease (GVHD) and cytokine release syndrome (CRS). Here, we show compelling evidence for the use of NK cell therapeutics as a reliable off‐the‐shelf option, as they address key issues. Furthermore, we highlight how iPS cells and directed differentiation toward HSC and NK cells address industrial scalability and safety.
- Published
- 2021
- Full Text
- View/download PDF
7. Expression of Sex Hormone Receptor and Immune Response Genes in Peripheral Blood Mononuclear Cells During the Menstrual Cycle
- Author
-
Peik M. A. Brundin, Britt-Marie Landgren, Peter Fjällström, Mohamed M. Shamekh, Jan-Åke Gustafsson, Anders F. Johansson, and Ivan Nalvarte
- Subjects
menstrual cycle ,estrogen receptor ,progesterone ,sex hormone ,immune response ,estrogen ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Sex hormones are known to interact with the immune system on multiple levels but information on the types of sex hormone receptors (SHR) and their expression levels in immune cells is scarce. Estrogen, testosterone and progesterone are all considered to interact with the immune system through their respective cell receptors (ERα and ERβ including the splice variant ERβ2, AR and PGR). In this study expression levels of SHR genes in peripheral blood mononuclear cells (PBMCs) and cell subsets (CD4+ and CD8+ T-cells, CD56+ NK-cells, CD14+ monocytes and CD19+ B-cells) were analyzed using standard manual qPCR or a qPCR array (TLDA). Nine healthy individuals including men (n = 2), premenopausal (Pre-MP, n = 5) and postmenopausal (post-MP, n = 2) women were sampled for PBMCs which were separated to cell subsets using FACS. Ten Pre-MP women were longitudinally sampled for total PBMCs at different phases of the menstrual cycle. We found that ERα was most abundant and, unexpectedly, that ERβ2 was the dominant ERβ variant in several FACS sorted cell subsets. In total PBMCs, SHR (ERα, ERβ1, ERβ2, and AR) expression did not fluctuate according to the phase of the menstrual cycle and PGR was not expressed. However, several immune response genes (GATA3, IFNG, IL1B, LTA, NFKB1, PDCD1, STAT3, STAT5A, TBX21, TGFB1, TNFA) were more expressed during the ovulatory and mid-luteal phases. Sex hormone levels did not correlate significantly with gene expression of SHR or immune response genes, but sex hormone-binding globulin (SHBG), a steroid hormone transporting protein, was positively correlated to expression of ERβ1 gene. This study provides new insights in the distribution of ERs in immune cells. Furthermore, expression patterns of several immune response genes differ significantly between phases of the menstrual cycle, supporting a role for sex hormones in the immune response.
- Published
- 2021
- Full Text
- View/download PDF
8. Generation of nonviral integration-free human iPS cell line KISCOi001-A from normal human fibroblasts, under defined xeno-free and feeder-free conditions
- Author
-
Jose Inzunza, Jonathan Arias-Fuenzalida, Juan Segura-Aguilar, Ivan Nalvarte, and Mukesh Varshney
- Subjects
Biology (General) ,QH301-705.5 - Abstract
KISCOi001-A is a healthy feeder-free and fully characterized human induced pluripotent stem (iPS) cell line cultured under xeno-free and defined conditions. The cell line is generated from normal human foreskin fibroblasts with non-integrating episomal plasmid vectors encoding OCT4, SOX2, KLF4, NANOG, LIN28, nontransforming L-MYC and dominant negative p53. The generated iPS cells are transgene-free and their pluripotency is confirmed by the expression of stem cell markers and capacity to differentiate into the cells of ectoderm, endoderm and mesoderm while their identity and karyotype stability is confirmed with Genomic assays.
- Published
- 2021
- Full Text
- View/download PDF
9. Corrigendum to 'Blood hormones and torque teno virus in peripheral blood mononuclear cells' [Heliyon 6 (11) (2020) e05535]
- Author
-
Peik M.A. Brundin, Britt-Marie Landgren, Peter Fjällström, Anders F. Johansson, and Ivan Nalvarte
- Subjects
Science (General) ,Q1-390 ,Social sciences (General) ,H1-99 - Published
- 2021
- Full Text
- View/download PDF
10. Blood hormones and torque teno virus in peripheral blood mononuclear cells
- Author
-
Peik M.A. Brundin, Britt-Marie Landgren, Peter Fjällström, Anders F. Johansson, and Ivan Nalvarte
- Subjects
Infectious disease ,Immunology ,Hematology ,Immune response ,Immunodeficiency ,Viruses ,Science (General) ,Q1-390 ,Social sciences (General) ,H1-99 - Abstract
Men and women respond differently to infectious diseases. Women show less morbidity and mortality, partially due to the differences in sex hormone levels which can influence the immune response. Torque teno virus (TTV) is non-pathogenic and ubiquitously present in serum from a large proportion (up to 90%) of adult humans with virus levels correlating with the status of the host immune response. The source of TTV replication is unknown, but T-lymphocytes have been proposed. In this study we investigated the presence and levels of TTV in peripheral blood mononuclear cells (PBMCs) in premenopausal (pre-MP) women, post-menopausal (post-MP) women, and men, and determined their serum sex hormone levels. Of the examined subjects (n = 27), we found presence of TTV in PMBC from 17.6% pre-MP (n = 17), 25.0% post-MP (n = 4) and 50.0% men (n = 6). The levels of TTV/μg DNA were lower among TTV-positive men and post-MP women compared to pre-MP women. All the positive pre-MP women were either anovulatory, hypothyroid, or both. In addition, the TTV-positive pre-MP women had significantly lower progesterone levels compared to TTV-negative pre-MP women. Although our study was performed on a limited number of subjects, the data suggests that TTV in PBMC is associated with an anovulatory menstrual cycle with low progesterone levels, and possibly with male sex.
- Published
- 2020
- Full Text
- View/download PDF
11. Neuroprotection against Aminochrome Neurotoxicity: Glutathione Transferase M2-2 and DT-Diaphorase
- Author
-
Juan Segura-Aguilar, Patricia Muñoz, Jose Inzunza, Mukesh Varshney, Ivan Nalvarte, and Bengt Mannervik
- Subjects
glutathione ,glutathione transferase ,dopamine ,Parkinson’s disease ,neuron ,astrocytes ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Glutathione is an important antioxidant that plays a crucial role in the cellular protection against oxidative stress and detoxification of electrophilic mutagens, and carcinogens. Glutathione transferases are enzymes catalyzing glutathione-dependent reactions that lead to inactivation and conjugation of toxic compounds, processes followed by subsequent excretion of the detoxified products. Degeneration and loss of neuromelanin-containing dopaminergic neurons in the nigrostriatal neurons generally involves oxidative stress, neuroinflammation, alpha-synuclein aggregation to neurotoxic oligomers, mitochondrial dysfunction, protein degradation dysfunction, and endoplasmic reticulum stress. However, it is still unclear what triggers these neurodegenerative processes. It has been reported that aminochrome may elicit all of these mechanisms and, interestingly, aminochrome is formed inside neuromelanin-containing dopaminergic neurons during neuromelanin synthesis. Aminochrome is a neurotoxic ortho-quinone formed in neuromelanin synthesis. However, it seems paradoxical that the neurotoxin aminochrome is generated during neuromelanin synthesis, even though healthy seniors have these neurons intact when they die. The explanation of this paradox is the existence of protective tools against aminochrome neurotoxicity composed of the enzymes DT-diaphorase, expressed in these neurons, and glutathione transferase M2-2, expressed in astrocytes. Recently, it has been reported that dopaminergic neurons can be protected by glutathione transferase M2-2 from astrocytes, which secrete exosomes containing the protective enzyme.
- Published
- 2022
- Full Text
- View/download PDF
12. Sex Stratified Treatment of Neurological Disorders: Challenges and Perspectives
- Author
-
Ivan Nalvarte
- Subjects
sex hormone ,sex chromosome ,gender ,neurobiology ,neurodegeneration ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Despite the obvious sex differences in many of the most common neuropsychiatric and neurodegenerative disorders, males and females are still often treated the same from a clinical perspective. Why is that? The simple answer is that there is still too little known about this very complex matter. Sex hormone signaling, genetic sex, sex-biased comorbidities, and social gender perceptions all interact, making discrimination between the impacts of each of these factors hard to evaluate. Adding to the complexity is that important species differences must be taken into account when interpreting data from animal models. Clearly, to overcome this, larger efforts are needed that incorporate epidemiological, experimental, and clinical data to provide a solid scientific base for more personalized and informed clinical decisions that will benefit both men and women suffering from neurological disorders.
- Published
- 2020
- Full Text
- View/download PDF
13. Ret inhibition decreases growth and metastatic potential of estrogen receptor positive breast cancer cells
- Author
-
Albana Gattelli, Ivan Nalvarte, Anne Boulay, Tim C. Roloff, Martin Schreiber, Neil Carragher, Kenneth K. Macleod, Michaela Schlederer, Susanne Lienhard, Lukas Kenner, Maria I. Torres‐Arzayus, and Nancy E. Hynes
- Subjects
endocrine‐therapy ,Fak ,IL6 ,metastasis ,Ret ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract We show that elevated levels of Ret receptor are found in different sub‐types of human breast cancers and that high Ret correlates with decreased metastasis‐free survival. The role of Ret in ER+ breast cancer models was explored combining in vitro and in vivo approaches. Our analyses revealed that ligand‐induced Ret activation: (i) stimulates migration of breast cancer cells; (ii) rescues cells from anti‐proliferative effects of endocrine treatment and (iii) stimulates expression of cytokines in the presence of endocrine agents. Indeed, we uncovered a positive feed‐forward loop between the inflammatory cytokine IL6 and Ret that links them at the expression and the functional level. In vivo inhibition of Ret in a metastatic breast cancer model inhibits tumour outgrowth and metastatic potential. Ret inhibition blocks the feed‐forward loop by down‐regulating Ret levels, as well as decreasing activity of Fak, an integrator of IL6‐Ret signalling. Our results suggest that Ret kinase should be considered as a novel therapeutic target in subsets of breast cancer.
- Published
- 2013
- Full Text
- View/download PDF
14. Genes, Gender, Environment, and Novel Functions of Estrogen Receptor Beta in the Susceptibility to Neurodevelopmental Disorders
- Author
-
Mukesh Varshney and Ivan Nalvarte
- Subjects
estrogen ,testosterone ,dyslexia ,aromatase ,brain ,neurodevelopment ,sex-difference ,hormone ,BPA ,epigenetics ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Many neurological disorders affect men and women differently regarding prevalence, progression, and severity. It is clear that many of these disorders may originate from defective signaling during fetal or perinatal brain development, which may affect males and females differently. Such sex-specific differences may originate from chromosomal or sex-hormone specific effects. This short review will focus on the estrogen receptor beta (ERβ) signaling during perinatal brain development and put it in the context of sex-specific differences in neurodevelopmental disorders. We will discuss ERβ’s recent discovery in directing DNA de-methylation to specific sites, of which one such site may bear consequences for the susceptibility to the neurological reading disorder dyslexia. We will also discuss how dysregulations in sex-hormone signaling, like those evoked by endocrine disruptive chemicals, may affect this and other neurodevelopmental disorders in a sex-specific manner through ERβ.
- Published
- 2017
- Full Text
- View/download PDF
15. Estrogen Receptor Knockout Mice and Their Effects on Fertility
- Author
-
Ivan Nalvarte and Per Antonson
- Abstract
Estrogens play a crucial role in sexual development and fertility as well as many other physiological processes, and it is estrogen receptors that mediate the physiological responses. To study the role of the estrogen receptors in these processes, several genetic mouse models have been developed using different strategies, which also in some cases yield different results. Here, we summarize the models that have been made and their impact on fertility in relation to known cases of human estrogen receptor mutations.
- Published
- 2023
16. ERβ in Granulosa Cell Tumors and Its Clinical Potential
- Author
-
Madeleine Birgersson, Rajitha Indukuri, Per Antonson, Ivan Nalvarte, Amena Archer, and Cecilia Williams
- Subjects
Endocrinology - Abstract
Granulosa cell tumors (GCTs) are rare ovarian tumors comprising an adult and a juvenile subtype. They have a generally good prognosis, but the survival rate drastically declines in patients with late-stage or recurring tumors. Due to the rarity of GCTs, the tumor type is largely understudied and lacks a specific treatment strategy. Estrogen receptor beta (ERβ/ESR2) has been found to be highly expressed in GCTs, which could be of therapeutic importance since it can be targeted with small molecules. However, its role in GCTs is not known. In this review, we summarize the current knowledge about the action of ERβ in the ovary and discuss its prospective role in GCTs.
- Published
- 2023
17. Estrogen receptors and the aging brain
- Author
-
Ivan Nalvarte, Karin Leander, Silvia Maioli, and Per Nilsson
- Subjects
Male ,Aging ,medicine.drug_class ,Parkinson's disease ,Estrogen receptor ,Biology ,Biochemistry ,Neuroprotection ,Endocrinology ,Alzheimer Disease ,Sex differences ,medicine ,Humans ,Aging brain ,Sex hormones ,Review Articles ,Molecular Biology ,Neuroinflammation ,Brain ,Alzheimer's disease ,Ageing ,Metabolism ,Receptors, Estrogen ,Estrogen ,Synaptic plasticity ,Female ,Estrogen receptor alpha ,Neuroscience ,Hormone - Abstract
The female sex hormone estrogen has been ascribed potent neuroprotective properties. It signals by binding and activating estrogen receptors that, depending on receptor subtype and upstream or downstream effectors, can mediate gene transcription and rapid non-genomic actions. In this way, estrogen receptors in the brain participate in modulating neural differentiation, proliferation, neuroinflammation, cholesterol metabolism, synaptic plasticity, and behavior. Circulating sex hormones decrease in the course of aging, more rapidly at menopause in women, and slower in men. This review will discuss what this drop entails in terms of modulating neuroprotection and resilience in the aging brain downstream of spatiotemporal estrogen receptor alpha (ERα) and beta (ERβ) signaling, as well as in terms of the sex differences observed in Alzheimer’s disease (AD) and Parkinson’s disease (PD). In addition, controversies related to ER expression in the brain will be discussed. Understanding the spatiotemporal signaling of sex hormones in the brain can lead to more personalized prevention strategies or therapies combating neurodegenerative diseases.
- Published
- 2021
18. Hematopoietic Stem Cell- and Induced Pluripotent Stem Cell-Derived CAR-NK Cells as Reliable Cell-Based Therapy Solutions
- Author
-
Mukesh Varshney, José Inzunza, Ivan Nalvarte, Jonathan Arias, and Jingwei Yu
- Subjects
0301 basic medicine ,Medicine (General) ,Induced Pluripotent Stem Cells ,Cell ,Biology ,Concise Reviews ,Immunotherapy, Adoptive ,03 medical and health sciences ,R5-920 ,0302 clinical medicine ,Directed differentiation ,medicine ,Humans ,natural killer cells (NK) ,hematopoietic stem cell (HSC) ,Host disease ,Induced pluripotent stem cell ,chimeric antigen receptor (CAR) ,Receptors, Chimeric Antigen ,QH573-671 ,Concise Review ,Hematopoietic stem cell ,Cell Biology ,General Medicine ,Hematopoietic Stem Cells ,medicine.disease ,Chimeric antigen receptor ,Killer Cells, Natural ,induced pluripotent stem (iPS) cell ,Cytokine release syndrome ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,Cytology ,030217 neurology & neurosurgery ,CAR‐NK ,Developmental Biology ,Cell based - Abstract
Hematopoietic stem cell‐ (HSC) and induced pluripotent stem (iPS) cell‐derived natural killer (NK) cells containing engineered functions, such as chimeric antigen receptors (CAR), offer great promise for the treatment of seemingly incurable oncological malignancies. Today, some of the main challenges of CAR cell‐based therapeutics are the long manufacturing time and safety of the cell sources used. Additional challenges include avoiding graft vs host disease (GVHD) and cytokine release syndrome (CRS). Here, we show compelling evidence for the use of NK cell therapeutics as a reliable off‐the‐shelf option, as they address key issues. Furthermore, we highlight how iPS cells and directed differentiation toward HSC and NK cells address industrial scalability and safety., Main areas of improvement in the creation of reliable off‐the‐shelf chimeric antigen receptor‐natural killer (CAR‐NK)‐based therapies. Superdonor induced pluripotent stem (iPS) cell and hematopoietic stem cell (HSC) sources, defined xeno‐free differentiation protocols, CAR genes inserted at defined genetic dose in safe harbor loci, and CAR proteins with effective on‐off switch transition and endodomains.
- Published
- 2021
19. Generation of an all-exon Esr2 deleted mouse line: Effects on fertility
- Author
-
Ivan Nalvarte, Matti Poutanen, Patricia Humire, Mohamed M. Shamekh, Jan-Åke Gustafsson, Claes Ohlsson, Per Antonson, and Leticia Montanholi Apolinário
- Subjects
Male ,Ovulation ,0301 basic medicine ,Genetically modified mouse ,medicine.drug_class ,media_common.quotation_subject ,medicine.medical_treatment ,Biophysics ,Biology ,Biochemistry ,Andrology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Estrogen Receptor beta ,Enhancer ,Molecular Biology ,Gene ,Estrogen receptor beta ,media_common ,Estrous cycle ,Ovary ,Exons ,Cell Biology ,Steroid hormone ,Fertility ,030104 developmental biology ,Estrogen ,030220 oncology & carcinogenesis ,Female ,CRISPR-Cas Systems ,Gene Deletion ,hormones, hormone substitutes, and hormone antagonists - Abstract
Estrogen receptor beta (ERβ), encoded by the Esr2 gene, is one of two nuclear receptors that mediate the functions of the steroid hormone estradiol. The binding of estradiol to the receptor results in enhanced transcription of many genes that have estrogen response elements in promoter or enhancer regions. Several genetically modified mouse lines with mutations or deletions of exons in the Esr2 gene have been developed and results from analysis of these are not completely consistent, especially regarding ERβ's role in fertility. To address these controversies, we have used the CRISPR/Cas9 genome editing system to make a deletion of the entire Esr2 gene in the mouse genome and determined the effect of this mutation on fertility. We show that female Esr2 deleted mice, Esr2ΔE1-10, are subfertile at young age, with fewer litters and smaller litter size, and that they become infertile/have severely reduced fertility at around six months of age, while the male Esr2ΔE1-10 mice are fertile. Ovaries from Esr2ΔE1-10 mice are smaller than those from wild-type littermates and the morphology of the ovary displays very few corpora lutea, indicating a defect in ovulation. We also show that the estradiol levels are reduced at diestrus, the phase in the estrous cycle when levels are expected to start to increase before ovulation. Our results verify that ERβ has an important function in female reproduction, likely as a regulator of serum estradiol levels, and that its loss does not affect male reproductive function.
- Published
- 2020
20. Neuroprotection against Aminochrome Neurotoxicity: Glutathione Transferase M2-2 and DT-Diaphorase
- Author
-
Juan, Segura-Aguilar, Patricia, Muñoz, Jose, Inzunza, Mukesh, Varshney, Ivan, Nalvarte, and Bengt, Mannervik
- Abstract
Glutathione is an important antioxidant that plays a crucial role in the cellular protection against oxidative stress and detoxification of electrophilic mutagens, and carcinogens. Glutathione transferases are enzymes catalyzing glutathione-dependent reactions that lead to inactivation and conjugation of toxic compounds, processes followed by subsequent excretion of the detoxified products. Degeneration and loss of neuromelanin-containing dopaminergic neurons in the nigrostriatal neurons generally involves oxidative stress, neuroinflammation, alpha-synuclein aggregation to neurotoxic oligomers, mitochondrial dysfunction, protein degradation dysfunction, and endoplasmic reticulum stress. However, it is still unclear what triggers these neurodegenerative processes. It has been reported that aminochrome may elicit all of these mechanisms and, interestingly, aminochrome is formed inside neuromelanin-containing dopaminergic neurons during neuromelanin synthesis. Aminochrome is a neurotoxic ortho-quinone formed in neuromelanin synthesis. However, it seems paradoxical that the neurotoxin aminochrome is generated during neuromelanin synthesis, even though healthy seniors have these neurons intact when they die. The explanation of this paradox is the existence of protective tools against aminochrome neurotoxicity composed of the enzymes DT-diaphorase, expressed in these neurons, and glutathione transferase M2-2, expressed in astrocytes. Recently, it has been reported that dopaminergic neurons can be protected by glutathione transferase M2-2 from astrocytes, which secrete exosomes containing the protective enzyme.
- Published
- 2021
21. Citalopram attenuates social behavior deficits in the BTBR T+Itpr3tf/J mouse model of autism
- Author
-
Rui Xiao, Ivan Nalvarte, Yulong Cai, Xin Li, Lian Wang, and Xiaotang Fan
- Subjects
0301 basic medicine ,business.industry ,General Neuroscience ,Serotonin reuptake inhibitor ,Citalopram ,Serotonergic ,medicine.disease ,Affect (psychology) ,behavioral disciplines and activities ,Tail suspension test ,Marble burying ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Autism spectrum disorder ,mental disorders ,Medicine ,Autism ,business ,Neuroscience ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Autism spectrum disorder (ASD) is diagnosed by two core symptoms: impaired social communication and the presence of repetitive, stereotyped behaviors and/or restricted interests. Alterations in serotonergic signaling are involved in the genesis of ASD. Selective serotonin reuptake inhibitors (SSRIs) have been reported to reduce repetitive behaviors and rescue social deficits in ASD mouse models and patients. In the present study, we examined the potential of citalopram (a representative selective serotonin reuptake inhibitor) on sociability and repetitive behaviors in the BTBR T+Itpr3tf/J (BTBR) mouse model of ASD. We found that the deficits of sociability in the BTBR mice were reversed by a 20 mg/kg dose of citalopram treatment without any adverse effects on locomotor activity or anxiety level. In addition, both high (20 mg/kg) and low (10 mg/kg) doses decreased the repetitive behavior of marble burying but did not affect self-grooming behavior. Furthermore, both doses were shown to have antidepressant-like activity in both the B6 and the BTBR mice in the tail suspension test. Taken together, these findings further demonstrate that citalopram can alleviate behavioral abnormalities in the BTBR autism model and lend support to the hypothesis that SSRIs may be potential therapeutic drugs for the treatment of behavioral dysfunctions in ASD.
- Published
- 2019
22. Estrogen receptor beta and neural development
- Author
-
Ivan, Nalvarte, Mukesh, Varshney, Jose, Inzunza, and Jan-Åke, Gustafsson
- Subjects
Mice ,Estradiol ,Receptors, Estrogen ,Estrogen Receptor alpha ,Animals ,Estrogen Receptor beta ,Female - Abstract
The female sex hormone estradiol (E2, 17β-estradiol) has important functions in the developing brain. In addition to regulating sexual differentiation of the brain, E2 participates in the development of brain areas involved in functions unrelated to reproduction, such as cognition. E2 signals mainly thorough two estrogen receptors; estrogen receptor alpha (ERα) and beta (ERβ). While ERα has distinct functions for sexual imprinting of the developing brain, ERβ is considered to participate in the development of brain areas related to cognitive function. In this chapter we will focus on ERβ's role during neural development. We will discuss the contributions of sex chromosomal and sex hormonal effects in this process and place it in relation to recent data on ERβ obtained from stem cell models. Finally, we will discuss the lessons learned from mouse and stem cell models in understanding ERβ's role in neural development and how new stem cell models, by addressing the human relevance, may help to advance our progress in this field.
- Published
- 2021
23. Corrigendum to 'Blood hormones and torque teno virus in peripheral blood mononuclear cells' [Heliyon 6 (11) (2020) e05535]
- Author
-
Peter Fjällström, Britt-Marie Landgren, Anders Johansson, Ivan Nalvarte, and Peik M.A. Brundin
- Subjects
Torque teno virus ,Multidisciplinary ,lcsh:H1-99 ,Biology ,lcsh:Social sciences (General) ,lcsh:Science (General) ,Peripheral blood mononuclear cell ,Virology ,Hormone ,lcsh:Q1-390 - Published
- 2021
24. Estrogen receptor beta and neural development
- Author
-
Mukesh Varshney, José Inzunza, Ivan Nalvarte, and Jan-Åke Gustafsson
- Subjects
0301 basic medicine ,Sexual differentiation ,medicine.drug_class ,Estrogen receptor ,Biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Sex hormone-binding globulin ,Estrogen ,medicine ,biology.protein ,Stem cell ,Estrogen receptor alpha ,Neuroscience ,Neural development ,030217 neurology & neurosurgery ,Estrogen receptor beta - Abstract
The female sex hormone estradiol (E2, 17β-estradiol) has important functions in the developing brain. In addition to regulating sexual differentiation of the brain, E2 participates in the development of brain areas involved in functions unrelated to reproduction, such as cognition. E2 signals mainly thorough two estrogen receptors; estrogen receptor alpha (ERα) and beta (ERβ). While ERα has distinct functions for sexual imprinting of the developing brain, ERβ is considered to participate in the development of brain areas related to cognitive function. In this chapter we will focus on ERβ's role during neural development. We will discuss the contributions of sex chromosomal and sex hormonal effects in this process and place it in relation to recent data on ERβ obtained from stem cell models. Finally, we will discuss the lessons learned from mouse and stem cell models in understanding ERβ's role in neural development and how new stem cell models, by addressing the human relevance, may help to advance our progress in this field.
- Published
- 2021
25. Cellular Trafficking of Glutathione Transferase M2-2 Between U373MG and SHSY-S7 Cells is Mediated by Exosomes
- Author
-
Andres Hagg, Raúl Valdes, Ivan Nalvarte, Bengt Mannervik, José Inzunza, Alicia Armijo, Kjell Hultenby, Juan Segura-Aguilar, Mukesh Varshney, and Patricia Muñoz
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,General Neuroscience ,Intercellular transport ,Dopaminergic Neurons ,Dopaminergic ,Endogeny ,Toxicology ,Exosomes ,Neuroprotection ,Microvesicles ,Cell biology ,03 medical and health sciences ,Protein Transport ,030104 developmental biology ,0302 clinical medicine ,Enzyme ,chemistry ,Astrocytes ,Cell Line, Tumor ,Neurotoxin ,Humans ,Secretion ,030217 neurology & neurosurgery ,Glutathione Transferase - Abstract
The enzyme glutathione transferase M2-2, expressed in human astrocytes, increases its expression in the presence of aminochrome and catalyzes the conjugation of aminochrome, preventing its toxic effects. Secretion of the enzyme glutathione transferase M2-2 from U373MG cells, used as a cellular model for astrocytes, has been reported, and the enzyme is taken up by neuroblastoma SYSH-S7 cells and provide protection against aminochrome. The present study provides evidence that glutathione transferase M2-2 is released in exosomes from U373MG cells, thereby providing a means for intercellular transport of the enzyme. With particular relevance to Parkinson disease and other degenerative conditions, we propose a new mechanism by which astrocytes may protect dopaminergic neurons against the endogenous neurotoxin aminochrome.
- Published
- 2020
26. Loss of liver X receptor β in astrocytes leads to anxiety-like behaviors via regulating synaptic transmission in the medial prefrontal cortex in mice
- Author
-
Hongyu Zhong, Haiwei Xu, Margaret Warner, Zhongke Wang, Tianyao Liu, Xin Li, Jan-Åke Gustafsson, Lian Wang, Rui Xiao, Per Antonson, Xiaotang Fan, Jiao Zou, Ivan Nalvarte, and Chuan Wu
- Subjects
0301 basic medicine ,Male ,Dendritic spine ,Prefrontal Cortex ,Neurotransmission ,Anxiety ,Synaptic Transmission ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,Animals ,Liver X receptor ,Prefrontal cortex ,Molecular Biology ,Liver X Receptors ,Mice, Knockout ,Anxiety like ,Chemistry ,RNA ,Phenotype ,Psychiatry and Mental health ,030104 developmental biology ,nervous system ,Astrocytes ,Knockout mouse ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Astrocytes are integral components of synaptic transmission, and their dysfunction leads to neuropsychiatric disorders such as anxiety and depression. Liver X receptor β (LXRβ) is expressed in astrocytes, and LXRβ global knockout mice shows impaired synaptic formation. In order to define the role of LXRβ in astrocytes, we used a conditional Cre-loxP system to specifically remove LXRβ from astrocytes. We found that this deletion caused anxiety-like but not depressive-like behaviors in adult male mice. This behavioral phenotype could be completely reproduced by selective deletion of LXRβ in astrocytes in the medial prefrontal cortex (mPFC). Pyramidal neurons in layer V of mPFC are involved in mood behaviors. We found that there was an increased spontaneous excitatory synaptic transmission in layer V pyramidal neurons of the mPFC of these mice. This was concurrent with increased dendritic complexity, despite normal appearance and number of dendritic spines. In addition, gene ontology analysis of RNA sequencing revealed that deletion of astrocytic LXRβ led to the enrichment of the process of synaptic transmission in mPFC. Finally, we also confirmed that renormalized excitatory synaptic transmission in layer V pyramidal neurons alleviated the anxiety in mice with astrocytic LXRβ deletion in mPFC. Together, our findings reveal that astrocytic LXRβ in mPFC is critical in the regulation of synaptic transmission, and this provides a potential new target for treatment of anxiety-like behavior.
- Published
- 2020
27. Ventral prostate and mammary gland phenotype in mice with complete deletion of the ERβ gene
- Author
-
Jan-Åke Gustafsson, Per Antonson, Ivan Nalvarte, Wanfu Wu, Margaret Warner, and Leticia Montanholi
- Subjects
0301 basic medicine ,Male ,Medical Sciences ,Mammary gland ,Estrogen receptor ,Epithelium ,Mice ,0302 clinical medicine ,Clustered Regularly Interspaced Short Palindromic Repeats ,Sequence Deletion ,Mice, Knockout ,Multidisciplinary ,fibroplasia ,Prostate ,Biological Sciences ,DNA-Binding Proteins ,medicine.anatomical_structure ,Phenotype ,Receptors, Androgen ,030220 oncology & carcinogenesis ,CRISPR ,Knockout mouse ,Androgens ,Female ,Chemokines ,Signal Transduction ,Stromal cell ,Tumor suppressor gene ,Nerve Tissue Proteins ,Biology ,03 medical and health sciences ,Mammary Glands, Animal ,Progesterone receptor ,medicine ,Animals ,Estrogen Receptor beta ,RNA, Messenger ,Eye Proteins ,Inflammation ,Hyperplasia ,Tumor Suppressor Proteins ,chemokine ,Estrogen Receptor alpha ,Estrogens ,Molecular biology ,Androgen receptor ,030104 developmental biology ,Ki-67 Antigen ,Trans-Activators ,CRISPR-Cas Systems ,Stromal Cells ,Transcriptome ,Transcription Factors - Abstract
Significance The discovery of ERβ caused a new optimism for understanding and treatment of prostate cancer. However, over the past 20 y, many mistakes have been made in studies trying to define the physiological functions of ERβ. One of the bigger problems has been producing a good ERβ knockout mouse. Deletion of the DNA-binding domain of ERβ did not produce a knockout of ERβ function because most functions of ERβ do not rely on DNA binding of the receptor. We have now deleted the entire ERβ gene from the mouse genome and report that ERβ regulates growth and differentiation of the ventral prostate and mammary gland., Disagreements about the phenotype of estrogen receptor β (ERβ) knockout mouse, created by removing the DNA-binding domain of the ERβ gene or interruption of the gene with a neocassette (Oliver Smithies ERβ knockout mice [ERβOS−/−]), prompted us to create an ERβ knockout mouse by deleting the ERβ gene with the use of CRISPR/Cas9 technology. We confirmed that the ERβ gene was eliminated from the mouse genome and that no ERβ mRNA or protein was detectable in tissues of this mouse. Overall the phenotype of the ventral prostate (VP) and mammary gland (MG) in ERβcrispr−/− mice was similar to, but more severe than, that in the ERβOS−/−mice. In the VP of 6-mo-old ERβcrispr−/− mice there was epithelial hyperplasia, fibroplasia, inflammation, stromal overgrowth, and intraductal cancer-like lesions. This was accompanied by an increase in Ki67 and P63 and loss in DACH1 and PURα, two androgen receptor (AR) repressors. In the MG there was overexpression of estrogen receptor α and progesterone receptor, loss of collagen, increase in proliferation and expression of metalloproteases, and invasive epithelium. Surprisingly, by 18 mo of age, the number of hyperplastic foci was reduced, the ducts of the VP and MG became atrophic, and, in the VP, there was massive immune infiltration and massive desquamation of the luminal epithelial cells. These changes were coincident with reduced levels of androgens in males and estrogens in females. We conclude that ERβ is a tumor suppressor gene in the VP and MG where its loss increases the activity AR and ERα, respectively.
- Published
- 2020
28. Blood hormones and torque teno virus in peripheral blood mononuclear cells
- Author
-
Ivan Nalvarte, Anders Johansson, Britt-Marie Landgren, Peik M.A. Brundin, and Peter Fjällström
- Subjects
0301 basic medicine ,Torque teno virus ,Aging ,Physiology ,Infektionsmedicin ,0302 clinical medicine ,Sex hormone-binding globulin ,Medicine ,Sex hormones ,lcsh:Social sciences (General) ,media_common ,Infectious disease ,Multidisciplinary ,Hematology ,biology ,Viruses ,Endokrinologi och diabetes ,lcsh:H1-99 ,Infection ,Reproductive hormone ,Research Article ,medicine.medical_specialty ,Infectious Medicine ,medicine.drug_class ,media_common.quotation_subject ,Immunology ,Reproduktionsmedicin och gynekologi ,Endocrinology and Diabetes ,Peripheral blood mononuclear cell ,Virus ,Article ,Microbiology in the medical area ,03 medical and health sciences ,Anovulatory ,Hypothyroidism ,Internal medicine ,Obstetrics, Gynecology and Reproductive Medicine ,Mikrobiologi inom det medicinska området ,Immunodeficiency ,Immune response ,lcsh:Science (General) ,Menstrual cycle ,Steroid hormones ,business.industry ,Immunity ,Commensal viruses ,Immunology in the medical area ,Sex difference ,Estrogen ,030104 developmental biology ,Immunologi inom det medicinska området ,biology.protein ,Microbiome ,business ,030217 neurology & neurosurgery ,Hormone ,lcsh:Q1-390 - Abstract
Men and women respond differently to infectious diseases. Women show less morbidity and mortality, partially due to the differences in sex hormone levels which can influence the immune response. Torque teno virus (TTV) is non-pathogenic and ubiquitously present in serum from a large proportion (up to 90%) of adult humans with virus levels correlating with the status of the host immune response. The source of TTV replication is unknown, but T-lymphocytes have been proposed. In this study we investigated the presence and levels of TTV in peripheral blood mononuclear cells (PBMCs) in premenopausal (pre-MP) women, post-menopausal (post-MP) women, and men, and determined their serum sex hormone levels. Of the examined subjects (n = 27), we found presence of TTV in PMBC from 17.6% pre-MP (n = 17), 25.0% post-MP (n = 4) and 50.0% men (n = 6). The levels of TTV/μg DNA were lower among TTV-positive men and post-MP women compared to pre-MP women. All the positive pre-MP women were either anovulatory, hypothyroid, or both. In addition, the TTV-positive pre-MP women had significantly lower progesterone levels compared to TTV-negative pre-MP women. Although our study was performed on a limited number of subjects, the data suggests that TTV in PBMC is associated with an anovulatory menstrual cycle with low progesterone levels, and possibly with male sex., Infectious disease; Immunology; Hematology; Immune response; Immunodeficiency; Viruses; Reproductive hormone; Steroid hormones; Aging; Menstrual cycle; Estrogen; Anovulatory; Hypothyroidism; Infection; Immunity; Sex difference; Microbiome; Commensal viruses; Sex hormones.
- Published
- 2020
29. Motor Function Deficits in the Estrogen Receptor Beta Knockout Mouse: Role on Excitatory Neurotransmission and Myelination in the Motor Cortex
- Author
-
José Inzunza, Virpi Töhönen, Shintaro Katayama, Tianyao Liu, Juha Kere, Wanfu Wu, Kaarel Krjutškov, Nancy Yiu-Lin Yu, Xiaotang Fan, Ivan Nalvarte, Mukesh Varshney, Jan-Åke Gustafsson, Xin Li, STEMM - Stem Cells and Metabolism Research Program, Research Programs Unit, University of Helsinki, Juha Kere / Principal Investigator, and HUS Helsinki and Uusimaa Hospital District
- Subjects
Locomotor activity ,Male ,medicine.medical_specialty ,ER-BETA ,Endocrinology, Diabetes and Metabolism ,Gene Expression ,030209 endocrinology & metabolism ,Biology ,Neurotransmission ,Serotonergic ,Synaptic Transmission ,3124 Neurology and psychiatry ,030218 nuclear medicine & medical imaging ,DIFFERENTIAL EXPRESSION ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Myelination ,Mice ,0302 clinical medicine ,Endocrinology ,ADULT ,SYNAPTIC PLASTICITY ,Internal medicine ,medicine ,Animals ,Estrogen Receptor beta ,NEURONS ,Estrogen receptor beta ,Myelin Sheath ,Mice, Knockout ,NITRIC-OXIDE ,NONSELECTIVE ATTENTION ,Endocrine and Autonomic Systems ,3112 Neurosciences ,Motor Cortex ,Estrogen ,SINGLE-CELL TRANSCRIPTOME ,Motor coordination ,Oligodendroglia ,medicine.anatomical_structure ,3121 General medicine, internal medicine and other clinical medicine ,Knockout mouse ,HIPPOCAMPUS ,Primary motor cortex ,Locomotion ,Psychomotor Performance ,Basolateral amygdala ,Motor cortex - Abstract
Background: Male estrogen receptor beta (ERβ) knockout (BERKO) mice display anxiety and aggression linked to, among others, altered serotonergic signaling in the basolateral amygdala and dorsal raphe, impaired cortical radial glia migration, and reduced GABAergic signaling. The effects on primary motor cortex (M1 cortex) and locomotor activity as a consequence of ERβ loss have not been investigated. Objective: The aim of this study was to determine whether locomotor activity is altered as a consequence of the changes in the M1 cortex. Methods: The locomotor activity of male wild-type (WT) and BERKO mice was evaluated using the open-field and rotarod tests. Molecular changes in the M1 cortex were analyzed by RNA sequencing, electron microscopy, electrophysiology, and immunohistological techniques. In addition, we established oligodendrocyte (OL) cultures from WT and BERKO mouse embryonic stem cells to evaluate OL function. Results: Locomotor profiling revealed that BERKO mice were more active than WT mice but had impaired motor coordination. Analysis of the M1 cortex pointed out differences in synapse function and myelination. There was a reduction in GABAergic signaling resulting in imbalanced excitatory and inhibitory neurotransmission as well as a defective OL differentiation accompanied by myelin defects. The effects of ERβ loss on OL differentiation were confirmed in vitro. Conclusion: ERβ is an important regulator of GABAergic interneurons and OL differentiation, which impacts on adult M1 cortex function and may be linked to increased locomotor activity and decreased motor coordination in BERKO mice.
- Published
- 2019
30. Generation of nonviral integration-free human iPS cell line KISCOi001-A from normal human fibroblasts, under defined xeno-free and feeder-free conditions
- Author
-
Juan Segura-Aguilar, Jonathan Arias-Fuenzalida, Ivan Nalvarte, José Inzunza, and Mukesh Varshney
- Subjects
Male ,0301 basic medicine ,Homeobox protein NANOG ,Mesoderm ,Foreskin ,Induced Pluripotent Stem Cells ,Biology ,LIN28 ,Stem cell marker ,Cell Line ,Kruppel-Like Factor 4 ,03 medical and health sciences ,0302 clinical medicine ,SOX2 ,medicine ,Humans ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Cell Differentiation ,Cell Biology ,General Medicine ,Fibroblasts ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,Cell culture ,embryonic structures ,Endoderm ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
KISCOi001-A is a healthy feeder-free and fully characterized human induced pluripotent stem (iPS) cell line cultured under xeno-free and defined conditions. The cell line is generated from normal human foreskin fibroblasts with non-integrating episomal plasmid vectors encoding OCT4, SOX2, KLF4, NANOG, LIN28, nontransforming L-MYC and dominant negative p53. The generated iPS cells are transgene-free and their pluripotency is confirmed by the expression of stem cell markers and capacity to differentiate into the cells of ectoderm, endoderm and mesoderm while their identity and karyotype stability is confirmed with Genomic assays.
- Published
- 2021
31. Memo interacts with c-Src to control Estrogen Receptor alpha sub-cellular localization
- Author
-
Nancy E. Hynes, Ivan Nalvarte, Gwen MacDonald, Ingrid Lund, Anna Frei, and Jan-Åke Gustafsson
- Subjects
0301 basic medicine ,Nonheme Iron Proteins ,CSK Tyrosine-Protein Kinase ,0302 clinical medicine ,heregulin ,Cell Movement ,estrogen ,Phosphorylation ,Cellular localization ,Intracellular Signaling Peptides and Proteins ,src-Family Kinases ,Oncology ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Disease Progression ,MCF-7 Cells ,Neuregulin ,Female ,Signal transduction ,Research Paper ,Signal Transduction ,medicine.medical_specialty ,Cell signaling ,animal structures ,Antineoplastic Agents, Hormonal ,medicine.drug_class ,Neuregulin-1 ,c-Src ,Breast Neoplasms ,03 medical and health sciences ,Breast cancer ,Internal medicine ,medicine ,Humans ,Cell Nucleus ,business.industry ,Estrogen Receptor alpha ,Estrogens ,medicine.disease ,ER alpha ,030104 developmental biology ,Endocrinology ,Microscopy, Fluorescence ,Nuclear receptor ,Memo1 ,Drug Resistance, Neoplasm ,Estrogen ,Cancer research ,business ,Estrogen receptor alpha - Abstract
// Anna Frei 1,4 , Gwen MacDonald 1 , Ingrid Lund 2 , Jan-Ake Gustafsson 2,3 , Nancy E. Hynes 1,4 and Ivan Nalvarte 2 1 Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse, Basel, Switzerland 2 Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden 3 Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX, USA 4 University of Basel, Basel, Switzerland Correspondence to: Ivan Nalvarte, email: // Keywords : Memo1, heregulin, ER alpha, estrogen, c-Src Received : June 30, 2016 Accepted : July 14, 2016 Published : July 26, 2016 Abstract Understanding the complex interaction between growth factor and steroid hormone signaling pathways in breast cancer is key to identifying suitable therapeutic strategies to avoid progression and therapy resistance. The interaction between these two pathways is of paramount importance for the development of endocrine resistance. Nevertheless, the molecular mechanisms behind their crosstalk are still largely obscure. We previously reported that Memo is a small redox-active protein that controls heregulin-mediated migration of breast cancer cells. Here we report that Memo sits at the intersection between heregulin and estrogen signaling, and that Memo controls Estrogen Receptor alpha (ERα) sub-cellular localization, phosphorylation, and function downstream of heregulin and estrogen in breast cancer cells. Memo facilitates ERα and c-Src interaction, ERα Y537 phosphorylation, and has the ability to control ERα extra-nuclear localization. Thus, we identify Memo as an important key mediator between the heregulin and estrogen signaling pathways, which affects both breast cancer cell migration and proliferation.
- Published
- 2016
32. Estrogen receptor β controls MMP-19 expression in mouse ovaries during ovulation
- Author
-
Ivan Nalvarte, Jan-Ake Gustafsson, Virpi Töhönen, José Inzunza, Maria Lindeberg, and Mukesh Varshney
- Subjects
Ovulation ,0301 basic medicine ,Embryology ,Sp1 Transcription Factor ,media_common.quotation_subject ,Down-Regulation ,Estrogen receptor ,Biology ,Matrix metalloproteinase ,Chorionic Gonadotropin ,Extracellular matrix ,Andrology ,03 medical and health sciences ,Follicle ,Endocrinology ,Downregulation and upregulation ,Matrix Metalloproteinases, Secreted ,Animals ,Estrogen Receptor beta ,Humans ,Estrogen receptor beta ,media_common ,Mice, Knockout ,Ovary ,Estrogen Receptor alpha ,Obstetrics and Gynecology ,Cell Biology ,Mice, Inbred C57BL ,030104 developmental biology ,Reproductive Medicine ,MCF-7 Cells ,Female ,Estrogen receptor alpha - Abstract
Estrogen receptor beta (ERβ/ESR2) has a central role in mouse ovaries, as ERβ knockout (BERKO) mice are subfertile due to an increase in fibrosis around the maturing follicle and a decrease in blood supply. This has a consequence that these follicles rarely rupture to release the mature oocyte. Matrix metalloproteinases (MMPs) are modulators of the extracellular matrix, and the expression of one specific MMP, MMP-19, is normally increased in granulosa cells during their maturation until ovulation. In this study, we demonstrate that MMP-19 levels are downregulated in BERKO mouse ovaries. Using human MCF-7 cells that overexpress ERβ, we could identify MMP-19 to be a transcriptional target of ligand-bound activated ERβ acting on a specificity protein-1 binding site. These data provide a molecular explanation for the observed follicle rupture defect that contributes to the subfertility of female BERKO mice.
- Published
- 2016
33. Citalopram attenuates social behavior deficits in the BTBR T
- Author
-
Yulong, Cai, Lian, Wang, Ivan, Nalvarte, Rui, Xiao, Xin, Li, and Xiaotang, Fan
- Subjects
Male ,Behavior, Animal ,Autism Spectrum Disorder ,Mice, Inbred Strains ,Citalopram ,Grooming ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,Random Allocation ,Animals ,Stereotyped Behavior ,Social Behavior ,Selective Serotonin Reuptake Inhibitors - Abstract
Autism spectrum disorder (ASD) is diagnosed by two core symptoms: impaired social communication and the presence of repetitive, stereotyped behaviors and/or restricted interests. Alterations in serotonergic signaling are involved in the genesis of ASD. Selective serotonin reuptake inhibitors (SSRIs) have been reported to reduce repetitive behaviors and rescue social deficits in ASD mouse models and patients. In the present study, we examined the potential of citalopram (a representative selective serotonin reuptake inhibitor) on sociability and repetitive behaviors in the BTBR T
- Published
- 2018
34. Intrinsic and Extrinsic Factors That Influence Epigenetics
- Author
-
Joëlle Rüegg, Ivan Nalvarte, and Carlos Guerrero-Bosagna
- Subjects
Epigenetic Process ,Endogenous Factors ,Epigenetics ,Reproductive capacity ,Biology ,Neuroscience ,Function (biology) ,Organism - Abstract
The original definition of epigenetics by Waddington is already aimed at describing how factors within the developmental processes of organisms affect gene expression. Every epigenetic process described to date has two basic components: the intrinsic machinery of the cells, and the contribution of external factors. Epigenetic machineries are involved in any aspect of the body's function. Intrinsic (endogenous) and extrinsic (environmentally available) factors alter the functioning of epigenetic machineries in the developing and adult organism and, subsequently, their reproductive capacity. Many endogenous factors shown to alter epigenetic mechanisms relate to the endocrine system, having distinct roles at different ontogenetic stages. Environmental factors that are able to influence the establishment of epigenetic mechanisms, through a variety of biological pathways, include compounds such as endocrine disruptors, nutritional factors, pharmaceuticals and inorganic chemicals. In the present chapter we summarize known intrinsic and extrinsic factors that affect epigenetic regulation of gene expression, placing especial emphasis in the reproductive consequences of their actions.
- Published
- 2018
35. Fluoxetine Affects Differentiation of Midbrain Dopaminergic Neurons In Vitro
- Author
-
Ivan Nalvarte, José Inzunza, Ulf Norinder, Daniel Mucs, Joëlle Rüegg, Mukesh Varshney, Diana Lupu, and Felicia Loghin
- Subjects
0301 basic medicine ,animal structures ,Autism Spectrum Disorder ,Dopamine ,Neurogenesis ,Estrogen receptor ,Down-Regulation ,Biology ,03 medical and health sciences ,Mice ,Mesencephalon ,Tubulin ,Fluoxetine ,Animals ,Estrogen Receptor beta ,Transcription factor ,Orthodenticle homeobox 2 ,Cells, Cultured ,Pharmacology ,Regulation of gene expression ,Homeodomain Proteins ,Otx Transcription Factors ,Dopaminergic Neurons ,Dopaminergic ,Estrogen Receptor alpha ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Nestin ,Cell biology ,030104 developmental biology ,Molecular Medicine ,Homeobox ,Signal Transduction ,Transcription Factors - Abstract
Recent meta-analyses found an association between prenatal exposure to the antidepressant fluoxetine (FLX) and an increased risk of autism in children. This developmental disorder has been related to dysfunctions in the brains' rewards circuitry, which, in turn, has been linked to dysfunctions in dopaminergic (DA) signaling. The present study investigated if FLX affects processes involved in dopaminergic neuronal differentiation. Mouse neuronal precursors were differentiated into midbrain dopaminergic precursor cells (mDPCs) and concomitantly exposed to clinically relevant doses of FLX. Subsequently, dopaminergic precursors were evaluated for expression of differentiation and stemness markers using quantitative polymerase chain reaction. FLX treatment led to increases in early regional specification markers orthodenticle homeobox 2 (Otx2) and homeobox engrailed-1 and -2 (En1 and En2). On the other hand, two transcription factors essential for midbrain dopaminergic (mDA) neurogenesis, LIM homeobox transcription factor 1 α (Lmx1a) and paired-like homeodomain transcription factor 3 (Pitx3) were downregulated by FLX treatment. The stemness marker nestin (Nes) was increased, whereas the neuronal differentiation marker β3-tubulin (Tubb3) decreased. Additionally, we observed that FLX modulates the expression of several genes associated with autism spectrum disorder and downregulates the estrogen receptors (ERs) α and β Further investigations using ERβ knockout (BERKO) mDPCs showed that FLX had no or even opposite effects on several of the genes analyzed. These findings suggest that FLX affects differentiation of the dopaminergic system by increasing production of dopaminergic precursors, yet decreasing their maturation, partly via interference with the estrogen system.
- Published
- 2018
36. Estrogen receptor β, a regulator of androgen receptor signaling in the mouse ventral prostate
- Author
-
Shintaro Katayama, Elisabet Einarsdottir, Juha Kere, Kaarel Krjutškov, Ivan Nalvarte, Virpi Töhönen, Yu Bing Dai, Wen Su, Jan-Åke Gustafsson, Jose Insunza, Bo Huang, Wanfu Wu, Laure Maneix, Margaret Warner, Nancy Yiu-Lin Yu, and Per Antonson
- Subjects
Male ,0301 basic medicine ,Aging ,Nedd4 Ubiquitin Protein Ligases ,Down-Regulation ,Estrogen receptor ,Smad7 Protein ,Mice ,03 medical and health sciences ,Prostate cancer ,Gene expression ,medicine ,Animals ,Estrogen Receptor beta ,PTEN ,Tensin ,Mice, Knockout ,Orphan receptor ,Multidisciplinary ,biology ,Interleukin-6 ,PTEN Phosphohydrolase ,Prostate ,Chemokine CXCL16 ,medicine.disease ,Prostate Stem Cell Antigen ,Androgen receptor ,Clusterin ,030104 developmental biology ,Proto-Oncogene Proteins c-bcl-2 ,PNAS Plus ,Receptors, Androgen ,Androgens ,biology.protein ,Cancer research ,Keratins ,Chemokines, CXC ,Signal Transduction - Abstract
As estrogen receptor β-/- (ERβ-/-) mice age, the ventral prostate (VP) develops increased numbers of hyperplastic, fibroplastic lesions and inflammatory cells. To identify genes involved in these changes, we used RNA sequencing and immunohistochemistry to compare gene expression profiles in the VP of young (2-mo-old) and aging (18-mo-old) ERβ-/- mice and their WT littermates. We also treated young and old WT mice with an ERβ-selective agonist and evaluated protein expression. The most significant findings were that ERβ down-regulates androgen receptor (AR) signaling and up-regulates the tumor suppressor phosphatase and tensin homolog (PTEN). ERβ agonist increased expression of the AR corepressor dachshund family (DACH1/2), T-cadherin, stromal caveolin-1, and nuclear PTEN and decreased expression of RAR-related orphan receptor c, Bcl2, inducible nitric oxide synthase, and IL-6. In the ERβ-/- mouse VP, RNA sequencing revealed that the following genes were up-regulated more than fivefold: Bcl2, clusterin, the cytokines CXCL16 and -17, and a marker of basal/intermediate cells (prostate stem cell antigen) and cytokeratins 4, 5, and 17. The most down-regulated genes were the following: the antioxidant gene glutathione peroxidase 3; protease inhibitors WAP four-disulfide core domain 3 (WFDC3); the tumor-suppressive genes T-cadherin and caveolin-1; the regulator of transforming growth factor β signaling SMAD7; and the PTEN ubiquitin ligase NEDD4. The role of ERβ in opposing AR signaling, proliferation, and inflammation suggests that ERβ-selective agonists may be used to prevent progression of prostate cancer, prevent fibrosis and development of benign prostatic hyperplasia, and treat prostatitis.
- Published
- 2017
37. Genes, Gender, Environment, and Novel Functions of Estrogen Receptor Beta in the Susceptibility to Neurodevelopmental Disorders
- Author
-
Ivan Nalvarte and Mukesh Varshney
- Subjects
0301 basic medicine ,aromatase ,medicine.drug_class ,brain ,hormone ,Context (language use) ,Review ,lcsh:RC321-571 ,03 medical and health sciences ,0302 clinical medicine ,dyslexia ,medicine ,estrogen ,Endocrine system ,Epigenetics ,Aromatase ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Estrogen receptor beta ,biology ,neurodevelopment ,epigenetics ,General Neuroscience ,Dyslexia ,medicine.disease ,BPA ,sex-difference ,030104 developmental biology ,Estrogen ,testosterone ,biology.protein ,Psychology ,Neuroscience ,030217 neurology & neurosurgery ,Hormone - Abstract
Many neurological disorders affect men and women differently regarding prevalence, progression, and severity. It is clear that many of these disorders may originate from defective signaling during fetal or perinatal brain development, which may affect males and females differently. Such sex-specific differences may originate from chromosomal or sex-hormone specific effects. This short review will focus on the estrogen receptor beta (ERβ) signaling during perinatal brain development and put it in the context of sex-specific differences in neurodevelopmental disorders. We will discuss ERβ's recent discovery in directing DNA de-methylation to specific sites, of which one such site may bear consequences for the susceptibility to the neurological reading disorder dyslexia. We will also discuss how dysregulations in sex-hormone signaling, like those evoked by endocrine disruptive chemicals, may affect this and other neurodevelopmental disorders in a sex-specific manner through ERβ.
- Published
- 2017
38. Fluoxetine affects midbrain neuronal specification and neurogenesis in vitro
- Author
-
Mukesh Varshney, Joëlle Rüegg, Ivan Nalvarte, Diana Lupu, J. Insunza, and Felicia Loghin
- Subjects
Midbrain ,Fluoxetine ,Neurogenesis ,medicine ,General Medicine ,Biology ,Toxicology ,Neuroscience ,In vitro ,medicine.drug - Published
- 2018
39. Abstract PD01-06: Inhibition of the Ret receptor tyrosine kinase in combination with endocrine therapy impacts on migration and metastatic potential of estrogen receptor positive breast cancer models
- Author
-
Nancy E. Hynes, Tim Roloff, Albana Gattelli, and Ivan Nalvarte
- Subjects
Cancer Research ,medicine.medical_specialty ,biology ,Fulvestrant ,business.industry ,medicine.medical_treatment ,Estrogen receptor ,Cancer ,medicine.disease ,Metastatic breast cancer ,Endocrinology ,Cytokine ,Oncology ,Internal medicine ,Glial cell line-derived neurotrophic factor ,biology.protein ,medicine ,Cancer research ,Aromatase ,skin and connective tissue diseases ,business ,neoplasms ,Tamoxifen ,medicine.drug - Abstract
The Ret receptor tyrosine kinase is overexpressed in approximately 30% of human breast cancers, many of which are estrogen receptor-alpha positive (ER+). Using the ER+ breast cancer models MCF7 and T47D, we have shown that RET is an ER target gene and that Ret signaling enhances estrogen-driven anchorage-dependent and -independent proliferation (Boulay et al 2008). In our current work we are investigating the potential role of Ret in endocrine therapy response using in vivo and in vitro models. Ret is activated by peptides of the glial derived neurotrophic factor (GDNF) family. GDNF treatment of ER+ T47D and MCF7 breast cancer cell lines stimulates activation of multiple intracellular pathways, increases anchorage-dependent and -independent growth, as well as migration. To test if Ret activation impacts on endocrine therapy response, we treated MCF7/Aromatase (Aro)-expressing cells for seven days with different endocrine agents in the presence or absence of GDNF to activate Ret. Activation of Ret signaling rescued the proliferative block that resulted from treatment of the MCF7/Aro cells with tamoxifen, fulvestrant or letrozole. A transcriptome analysis was performed and an Ingenuity Pathway Analysis of the transcripts altered in MCF7/Aro cells treated with fulvestrant or letrozole revealed that the highest pathways altered by addition of GDNF are related to interferon signaling and inflammation. Indeed, proinflammatory RNAs, including IL6, IL8 and CXCL10 are among the top transcripts upregulated in cultures treated with fulvestrant plus GDNF, or letrozole plus GDNF. Elevated IL6 serum levels in breast cancer patients correlate with poor prognosis. We have concentrated on IL6 in the work that will be presented based on this clinical data, as well as our finding that IL6 stimulates Ret expression and activation in ER+ breast cancer models. We verified that addition of GDNF to MDCF/Aro cultures treated with fulvestrant or with letrozole led to an increase of IL6 in the conditional medium of the cultures. Moreover, addition of GDNF to fulvestrant- or tamoxifen-treated tumor cells increased migration, which could be inhibited by an IL6 blocking antibody, showing the importance of this cytokine for tumor cell motility. Additional data will be presented showing there is a Ret-IL6 feed-forward loop present in ER+ tumor cell models and that this is important for migration. In order to examine the role of Ret in vivo, in a metastatic breast cancer model, we have used the ER+/Ret+ J110 tumor cell line, generated from mammary tumors of MMTV-AIB1 transgenic females. Ret is active in J110 mammary tumors and its activity can be blocked with a Ret selective kinase inhibitor. In vivo data will be presented showing the effect of blocking Ret alone, or in combination with endocrine agents, on metastatic spread of primary tumors to the lungs. Citation Information: Cancer Res 2012;72(24 Suppl):Abstract nr PD01-06.
- Published
- 2012
40. The rs3743205 SNP is important for the regulation of the dyslexia candidate gene DYX1C1 by estrogen receptor ß and DNA methylation
- Author
-
Isabel Tapia-Páez, Joëlle Rüegg, Juha Kere, Ivan Nalvarte, Kristiina Tammimies, Gustaf Rosin, and Jan-Åke Gustafsson
- Subjects
Codon, Initiator ,Estrogen receptor ,Nerve Tissue Proteins ,Biology ,Response Elements ,Polymorphism, Single Nucleotide ,Epigenesis, Genetic ,Dyslexia ,Transcription Factors, TFII ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Cell Line, Tumor ,medicine ,Estrogen Receptor beta ,Humans ,Genetic Predisposition to Disease ,Dyslexia susceptibility 1 ,Molecular Biology ,Transcription factor ,Genetic Association Studies ,Estrogen receptor beta ,Original Research ,030304 developmental biology ,Regulation of gene expression ,Genetics ,0303 health sciences ,Estradiol ,Nuclear Proteins ,Estrogens ,General Medicine ,DNA Methylation ,medicine.disease ,Cytoskeletal Proteins ,Gene Expression Regulation ,DNA methylation ,5' Untranslated Regions ,Estrogen receptor alpha ,030217 neurology & neurosurgery ,Protein Binding - Abstract
Estrogen is involved in numerous physiological processes such as growth, differentiation, and function of the male and female reproductive tissues. In the developing brain, estrogen signaling has been linked to cognitive functions, such as learning and memory; however, the molecular mechanisms underlying this phenomenon are poorly understood. We have previously shown a link between developmental dyslexia and estrogen signaling, when we studied the functional interactions between the dyslexia candidate protein DYX1C1 and the estrogen receptors α (ERα) and β (ERβ). Here, we investigate the 17β-estradiol (E2)-dependent regulation of dyslexia susceptibility 1 candidate 1 (DYX1C1) expression. We demonstrate that ERβ, not ERα, binds to a transcriptionally active cis-regulatory region upstream of DYX1C1 transcriptional start site and that DYX1C1 expression is enhanced by E2 in a neuroblastoma cell line. This regulation is dependent on transcription factor II-I and liganded ERβ recruitment to this region. In addition, we describe that a single nucleotide polymorphism previously shown to be associated with dyslexia and located in the cis-regulatory region of DYX1C1 may alter the epigenetic and endocrine regulation of this gene. Our data provide important molecular insights into the relationship between developmental dyslexia susceptibility and estrogen signaling.
- Published
- 2012
- Full Text
- View/download PDF
41. Proteomics Analysis of the Estrogen Receptor α Receptosome
- Author
-
Thomas Schwend, Ivan Nalvarte, and Jan-Åke Gustafsson
- Subjects
Proteomics ,Proteome ,Macromolecular Substances ,medicine.drug_class ,Molecular Sequence Data ,Estrogen receptor ,Endosomes ,Biology ,Response Elements ,Biochemistry ,Cell Line ,Analytical Chemistry ,medicine ,Animals ,Estrogen Receptor beta ,Humans ,Electrophoresis, Gel, Two-Dimensional ,Molecular Biology ,Transcription factor ,Estrogen receptor beta ,Hormone response element ,Research ,Estrogen Receptor alpha ,Estrogens ,Molecular biology ,Cell biology ,Estrogen ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Estrogen receptor alpha ,hormones, hormone substitutes, and hormone antagonists - Abstract
The estrogen receptors (ERs) are ligand-dependent transcription factors that activate transcription by binding to estrogen response elements. Estrogen-mediated effects are tissue- and cell type-specific, determined by the cofactor recruitment to the ERs among other factors. To understand these differences in estrogen action, it is important to identify the various compositions of the ER complexes (ER receptosomes). In this report, we describe a fast and efficient method for the isolation of the ERalpha receptosome for proteomics analysis. Using immobilized estrogen response element on a Sepharose column in combination with two-dimensional electrophoresis and MALDI-TOF MS, significant amounts of proteins could be isolated and identified. Differences in ERalpha complex composition with the ER ligands 17beta-estradiol, 4-hydroxytamoxifen, and ICI-182,780 could also be observed. Thus, this approach provides an easy and relevant way of identifying ERalpha cofactor and transcription factor recruitment under different conditions.
- Published
- 2010
42. Spatiotemporal dynamics of the expression of estrogen receptors in the postnatal mouse brain
- Author
-
Richard Lathe, Sandra Andersson, Ivan Nalvarte, Thomas Schwend, Margaret Warner, Nobuhiro Sugiyama, Paloma Alonso-Magdalena, Jan-Åke Gustafsson, and Xianfeng Fan
- Subjects
medicine.medical_specialty ,medicine.drug_class ,Central nervous system ,Cytochrome P450 Family 7 ,Estrogen receptor ,Apoptosis ,Mice ,Cellular and Molecular Neuroscience ,Internal medicine ,In Situ Nick-End Labeling ,medicine ,Animals ,Estrogen Receptor beta ,RNA, Messenger ,Aromatase ,Molecular Biology ,Mice, Knockout ,Analysis of Variance ,Estradiol ,biology ,Dentate gyrus ,Age Factors ,Estrogen Receptor alpha ,Brain ,Gene Expression Regulation, Developmental ,Organ Size ,Mice, Inbred C57BL ,Psychiatry and Mental health ,Endocrinology ,medicine.anatomical_structure ,Animals, Newborn ,Receptors, Estrogen ,Estrogen ,Dihydrotestosterone ,Steroid Hydroxylases ,Knockout mouse ,biology.protein ,Immunostaining ,medicine.drug - Abstract
This study reports on the spatiotemporal dynamics of the expression of estrogen receptors (ERs) in the mouse central nervous system (CNS) during the early postnatal and the peripubertal period. At postnatal day 7 (P7), neurons with strong nuclear immunostaining for both ERalpha and ERbeta1 were widely distributed throughout the brain. Sucrose density gradient sedimentation followed by western blotting supported the histochemical evidence for high levels of both ERs at P7. Over the following 2 days, there was a rapid downregulation of ERs. At P9, ERalpha expression was visible only in the hypothalamic area. Decline in ERbeta1 expression was slower than that of ERalpha, and ERalpha-negative, ERbeta1-positive cells were observed in the dentate gyrus and walls of third ventricle. Between P14 and P35, ERs were undetectable except for the hypothalamic area. As before P7, the ovary does not produce estrogen but does produce 5alpha-androstane-3beta, 17beta-diol (3betaAdiol), an estrogenic metabolite of dihydrotestosterone, we examined the effects of high levels of 3betaAdiol in the postnatal period. We used CYP7B1 knockout mice which cannot hydroxylate and inactivate 3betaAdiol. The brains of these mice are abnormally large with reduced apoptosis. In the early postnatal period, there was 1-week delay in the timing of the reduction in ER expression in the brain. These data reveal that the time when ERs might be activated in the brain is limited to the first 8 postnatal days. In addition, the importance of aromatase has to be reconsidered as the alternative estrogen, 3betaAdiol, is important in neuronal function in the postnatal brain.
- Published
- 2008
43. Identification of proteins highly expressed in uterine fluid from mice with hydrometra
- Author
-
Patricia Humire, Li Xu, Ivan Nalvarte, Per Antonson, Sara H Windahl, Karin Dahlman-Wright, Claes Ohlsson, Mukesh Varshney, and Jan-Åke Gustafsson
- Subjects
medicine.medical_specialty ,medicine.drug_class ,Biophysics ,Estrogen receptor ,Gene Expression ,Estrous Cycle ,Biochemistry ,Mice ,Internal medicine ,Gene expression ,Conditional gene knockout ,medicine ,Animals ,Chitinase-3-Like Protein 1 ,RNA, Messenger ,Molecular Biology ,Serpins ,Glycoproteins ,Estrous cycle ,Mice, Knockout ,Uterine Diseases ,biology ,Estradiol ,Lactoferrin ,Uterus ,Estrogen Receptor alpha ,Cell Biology ,Complement C3 ,Rats ,Mice, Inbred C57BL ,Endocrinology ,Estrogen ,Knockout mouse ,biology.protein ,Female ,Estrogen receptor alpha ,hormones, hormone substitutes, and hormone antagonists - Abstract
Estrogen receptor alpha (ERα) is an important regulator of the estrous cycle and mice with global ERα deletion, as well as some conditional knockout mouse lines, have an interruption in the estrous cycle. In this study we observed that conditional ERα knockout mice where the Cre gene is regulated by the rat insulin promoter (RIP), RIP-Cre/ERα(KO) mice, have a 3.7-fold increase in serum 17β-estradiol levels, blocked estrous cycle, and develop a fluid-filled uterus (hydrometra). Using a proteomics approach, we identified three proteins, lactoferrin, complement C3 and chitinase 3-like protein 1 (CHI3L1), as highly expressed proteins in hydrometra fluid. The mRNA levels of the corresponding genes were more than 50-fold higher in RIP-Cre/ERα(KO) uterus compared to controls. High expression of CHI3L1 in the uterine fluid was not reflected as elevated levels in the serum. The high expression of lactoferrin, complement C3 and CHI3L1 in the uterine fluid, in association with elevated estrogen levels, prompted us to address if the expression of these genes is related to reproduction. However, gonadotropin treatment of mice reduced the uterine expression of these genes in a model of in vitro fertilization. Our findings identify lactoferrin, complement C3 and CHI3L1 as highly expressed proteins in hydrometra fluid in association with chronically elevated serum estradiol levels.
- Published
- 2015
44. Developmental exposure to bisphenol A alters expression and DNA methylation of Fkbp5, an important regulator of the stress response
- Author
-
Ivan Nalvarte, Joëlle Rüegg, Ali Alavian-Ghavanini, and Efthymia Kitraki
- Subjects
Male ,endocrine system ,medicine.medical_specialty ,Offspring ,Estrogen receptor ,Biology ,Biochemistry ,Hippocampus ,Cell Line ,Epigenesis, Genetic ,Tacrolimus Binding Proteins ,03 medical and health sciences ,0302 clinical medicine ,Glucocorticoid receptor ,Endocrinology ,Phenols ,Stress, Physiological ,Internal medicine ,medicine ,Animals ,Estrogen Receptor beta ,Epigenetics ,Benzhydryl Compounds ,Molecular Biology ,Estrogen receptor beta ,030304 developmental biology ,0303 health sciences ,urogenital system ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Methylation ,DNA Methylation ,Rats ,Endocrine disruptor ,DNA methylation ,Epithalamus ,hormones, hormone substitutes, and hormone antagonists ,030217 neurology & neurosurgery - Abstract
Bisphenol A (BPA), an abundant endocrine disruptor, affects stress-responsiveness and related behaviors in children. In rats, perinatal BPA exposure modifies stress response in pubertal offspring via unknown mechanisms. Here we examined possible epigenetic modifications in the glucocorticoid receptor gene and its regulator Fkbp5 in hypothalamus and hippocampus of exposed offspring. We found increased DNA methylation of Fkbp5 and reduced protein levels in the hippocampus of exposed male rats. Similar effects were obtained in a male hippocampal cell line when exposed to BPA during differentiation. The estrogen receptor (ER) antagonist ICI 182,780 or ERβ knock-down affected Fkbp5 expression and methylation similarly to BPA. Further, BPA's effect on Fkbp5 was abolished upon knock-down of ERβ, suggesting a role for this receptor in mediating BPA's effects on Fkbp5. These data demonstrate that developmental BPA exposure modifies Fkbp5 methylation and expression in male rats, which may be related to its impact on stress responsiveness.
- Published
- 2015
45. The expression and activity of thioredoxin reductase 1 splice variants v1 and v2 regulate the expression of genes associated with differentiation and adhesion
- Author
-
Joëlle Rüegg, Anastasios E. Damdimopoulos, Giannis Spyrou, and Ivan Nalvarte
- Subjects
Thioredoxin Reductase 1 ,Cellular differentiation ,Thioredoxin reductase ,Biophysics ,Retinoic acid ,Biology ,migration ,Biochemistry ,differentiation ,oxidative stress ,selenium ,thioredoxin reductase ,chemistry.chemical_compound ,Humans ,Protein Isoforms ,Gene silencing ,Molecular Biology ,Regulation of gene expression ,Original Paper ,Cadherin ,HEK 293 cells ,Klinisk medicin ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Cell Biology ,Original Papers ,Molecular biology ,Cell biology ,Fibronectin ,Alternative Splicing ,HEK293 Cells ,chemistry ,biology.protein ,Clinical Medicine ,Oxidation-Reduction - Abstract
Thioredoxin reductase (TrxR1) is involved in redox homoeostasis and cellular differentiation. In the present study, we demonstrate that overexpression of TrxR1 affects genes associated with differentiation and that differentiation increased TrxR1 expression. The TrxR1 splice variant TXNRD1_v2 was also studied in this context., The mammalian redox-active selenoprotein thioredoxin reductase (TrxR1) is a main player in redox homoeostasis. It transfers electrons from NADPH to a large variety of substrates, particularly to those containing redox-active cysteines. Previously, we reported that the classical form of cytosolic TrxR1 (TXNRD1_v1), when overexpressed in human embryonic kidney cells (HEK-293), prompted the cells to undergo differentiation [Nalvarte et al. (2004) J. Biol. Chem. 279, 54510–54517]. In the present study, we show that several genes associated with differentiation and adhesion are differentially expressed in HEK-293 cells stably overexpressing TXNRD1_v1 compared with cells expressing its splice variant TXNRD1_v2. Overexpression of these two splice forms resulted in distinctive effects on various aspects of cellular functions including gene regulation patterns, alteration of growth rate, migration and morphology and susceptibility to selenium-induced toxicity. Furthermore, differentiation of the neuroblastoma cell line SH-SY5Y induced by all-trans retinoic acid (ATRA) increased both TXNRD1_v1 and TXNRD1_v2 expressions along with several of the identified genes associated with differentiation and adhesion. Selenium supplementation in the SH-SY5Y cells also induced a differentiated morphology and changed expression of the adhesion protein fibronectin 1 and the differentiation marker cadherin 11, as well as different temporal expression of the studied TXNRD1 variants. These data suggest that both TXNRD1_v1 and TXNRD1_v2 have distinct roles in differentiation, possibly by altering the expression of the genes associated with differentiation, and further emphasize the importance in distinguishing each unique action of different TrxR1 splice forms, especially when studying the gene silencing or knockout of TrxR1.
- Published
- 2015
46. Erratum for the Research Article: 'Memo Is a Copper-Dependent Redox Protein with an Essential Role in Migration and Metastasis' by G. MacDonald, I. Nalvarte, T. Smirnova, M. Vecchi, N. Aceto, A. Dolemeyer, A. Frei, S. Lienhard, J. Wyckoff, D. Hess, J. Seebacher, J. J. Keusch, H. Gut, D. Salaun, G. Mazzarol, D. Disalvatore, M. Bentires-Alj, P. P. Di Fiore, A. Badache, N. E. Hynes
- Author
-
Mohamed Bentires-Alj, Nancy E. Hynes, Anna Frei, Tatiana Smirnova, Daniel Hess, Jeffrey Wyckoff, Nicola Aceto, Ali Badache, Susanne Lienhard, A. Dolemeyer, P. P. Di Fiore, Maurizio Vecchi, Jeremy J. Keusch, Heinz Gut, Ivan Nalvarte, Davide Disalvatore, Gwen MacDonald, Danièle Salaün, Jan Seebacher, and Giovanni Mazzarol
- Subjects
Chemistry ,medicine ,chemistry.chemical_element ,Cell Biology ,medicine.disease ,Molecular Biology ,Biochemistry ,Redox ,Copper ,Metastasis ,Cell biology - Abstract
The spelling of an author’s name is corrected.
- Published
- 2014
47. Marked For Life: How Environmental Factors Affect the Epigenome
- Author
-
Stefan Weis, Joëlle Rüegg, Ivan Nalvarte, and Pauliina Damdimopoulou
- Subjects
Adverse conditions ,Endocrine system ,Disease ,Epigenetics ,Epigenome ,Biology ,Affect (psychology) ,Phenotype ,Neuroscience ,Hormone ,Developmental psychology - Abstract
The susceptibility to develop non-communicable diseases, like cardiovascular disease and cancer, depends on an interplay between the genes and the environment. Extrinsic factors, such as maternal stress hormones, dietary constituents, and endocrine disruptive chemicals, are known to affect fetal development and later disease phenotypes. Many of these factors have been shown to modify the epigenetic landscape during development, with a consequent change in the adult phenotype. Whether fetal exposure to adverse conditions leads to the epigenetic changes underlying non-communicable diseases in humans is an intensively studied question. In this chapter, we discuss the possible involvement of environmental factors, in particular endocrine disruptive chemicals, in shaping human health and risk of disease through epigenetic mechanisms. We start by describing different mechanisms to code epigenetic information and then move on to discussing how early life influences can affect this coding and what kind of consequences it can have on the phenotype level. Finally, we will describe the mechanistic principles by which endocrine disruptors could affect the epigenetic machinery.
- Published
- 2011
48. Overexpression of thioredoxin reductase 1 inhibits migration of HEK-293 cells
- Author
-
Ivan Nalvarte, Jarosław Czyż, Jolanta Sroka, Agata Antosik, Zbigniew Madeja, Giannis Spyrou, and Jerker M. Olsson
- Subjects
Thioredoxin Reductase 1 ,cell migration ,Thioredoxin reductase ,thioredoxin reductase (TrxR) ,Blotting, Western ,Biology ,Transfection ,medicine.disease_cause ,Cell Line ,redox regulation ,Cell Movement ,Organotin Compounds ,medicine ,Humans ,HEK 293 cells ,Cell migration ,Cell Biology ,General Medicine ,diphenyltin ,rotein kinase C (PKC) ,Actins ,Cell biology ,Blot ,Protein Kinase C-delta ,Biochemistry ,Cell culture ,Oxidative stress - Abstract
TrxR (thioredoxin reductase), in addition to protecting against oxidative stress, plays a role in the redox regulation of intracellular signalling pathways controlling, among others, cell proliferation and apoptosis. The aim of the present study was to determine whether TrxR1 is involved in the regulation of cell migration.Stably transfected HEK-293 (human embryonic kidney) cells which overexpress cytosolic TrxR1 (HEK-TrxR15 and HEK-TrxR11 cells) were used in the present study. We found that the stimulation of cell motility induced by PKC (protein kinase C) activators, PMA and DPhT (diphenyltin), was inhibited significantly in the HEK-TrxR15 and HEK-TrxR11 cells compared with control cells. The overexpression of TrxR1 also inhibited characteristic morphological changes and reorganization of the F-actin cytoskeleton induced by PMA and DPhT. In addition, the selective activation of PKCdelta by DPhT was inhibited in cells that overexpressed cytosolic TrxR1. Furthermore, rottlerin, a selective inhibitor of PKCdelta, and PKCdelta siRNA (small interfering RNA), suppressed the morphological changes induced by DPhT in the control cells.The overexpression of TrxR1 inhibits migration of HEK-293 cells stimulated with PMA and DPhT. Moreover, our observations suggest that this effect is mediated by the inhibition of PKCdelta activation.
- Published
- 2007
49. The role of thioredoxin reductase activity in selenium-induced cytotoxicity
- Author
-
Jerker M. Olsson, Tomas Nordman, Ivan Nalvarte, Jolanta Sroka, Giannis Spyrou, Christina Nyström, Lennart Eriksson, Linda Björkhem-Bergman, Zbigniew Madeja, Mikael Björnstedt, and Anastasios E. Damdimopoulos
- Subjects
Thioredoxin-Disulfide Reductase ,Thioredoxin reductase ,chemistry.chemical_element ,Selenium in biology ,cell motility ,Biochemistry ,Cell Line ,Selenium ,Cell Movement ,oxidative stress ,Humans ,Cytotoxicity ,selenium ,glutathione peroxidase ,Pharmacology ,chemistry.chemical_classification ,biology ,Dose-Response Relationship, Drug ,Glutathione peroxidase ,thioredoxin reductase ,Enzyme Activation ,chemistry ,biology.protein ,cytotoxicity ,Selenoprotein ,Thioredoxin ,Peroxidase - Abstract
The selenoprotein thioredoxin reductase is a key enzyme in selenium metabolism, reducing selenium compounds and thereby providing selenide to synthesis of all selenoproteins. We evaluated the importance of active TrxR1 in selenium-induced cytotoxicity using transfected TrxR1 over-expressing stable Human Embryo Kidney (HEK-293) cells and modulation of activity by pretreatment with low concentration of selenite. Treatment with sodium selenite induced cytotoxity in a dose-dependent manner in both TrxR1 over-expressing and control cells. However, TrxR1 over-expressing cells, which were preincubated for 72h with 0.1 microM selenite, were significantly more resistant to selenite cytotoxicity than control cells. To demonstrate the early effects of selenite on behaviour of HEK-293 cells, we also investigated the influence of this compound on cell motility. We observed inhibition of cell motility by 50 microM selenite immediately after administration. Moreover, TrxR1 over-expressing cells preincubated with a low concentration of selenite were more resistant to the inhibitory effect of 50 microM selenite than those not preincubated. It was also observed that the TrxR over-expressing cells showed higher TrxR1 activity than control cells and the preincubation of over-expressing cells with 0.1 microM selenite induced further significant increase in the activity of TrxR1. On the other hand, we demonstrated that TrxR1 over-expressing cells showed decreased glutathione peroxidase activity compared to control cells. These data strongly suggest that TrxR1 may be a crucial enzyme responsible for cell resistance against selenium cytotoxicity.
- Published
- 2004
50. Overexpression of enzymatically active human cytosolic and mitochondrial thioredoxin reductase in HEK-293 cells. Effect on cell growth and differentiation
- Author
-
Ivan, Nalvarte, Anastasios E, Damdimopoulos, Christina, Nystöm, Tomas, Nordman, Antonio, Miranda-Vizuete, Jerker M, Olsson, Lennart, Eriksson, Mikael, Björnstedt, Elias S J, Arnér, and Giannis, Spyrou
- Subjects
Glutathione Peroxidase ,Thioredoxin Reductase 1 ,Binding Sites ,Thioredoxin-Disulfide Reductase ,Dose-Response Relationship, Drug ,Thioredoxin Reductase 2 ,Gene Expression ,Apoptosis ,Cell Differentiation ,Embryo, Mammalian ,Kidney ,Transfection ,Cell Line ,Mitochondria ,Selenocysteine ,Selenium ,Cytosol ,Humans ,Reactive Oxygen Species ,Oxidation-Reduction ,Cell Division - Abstract
The mammalian thioredoxin reductases (TrxR) are selenoproteins containing a catalytically active selenocysteine residue (Sec) and are important enzymes in cellular redox control. The cotranslational incorporation of Sec, necessary for activity, is governed by a stem-loop structure in the 3'-untranslated region of the mRNA and demands adequate selenium availability. The complicated translation machinery required for Sec incorporation is a major obstacle in isolating mammalian cell lines stably overexpressing selenoproteins. In this work we report on the development and characterization of stably transfected human embryonic kidney 293 cells that overexpress enzymatically active selenocysteine-containing cytosolic TrxR1 or mitochondrial TrxR2. We demonstrate that the overexpression of selenium-containing TrxR1 results in lower expression and activity of the endogenous selenoprotein glutathione peroxidase and that the activity of overexpressed TrxRs, rather than the protein amount, can be increased by selenium supplementation in the cell growth media. We also found that the TrxR-overexpressing cells grew slower over a wide range of selenium concentrations, which was an effect apparently not related to increased apoptosis nor to fatally altered intracellular levels of reactive oxygen species. Most surprisingly, the TrxR1- or TrxR2-overexpressing cells also induced novel expression of the epithelial markers CK18, CK-Cam5.2, and BerEP4, suggestive of a stimulation of cellular differentiation.
- Published
- 2004
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.