Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
2.
Nature ; 631(8022): 857-866, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987586

ABSTRACT

Systemic lupus erythematosus (SLE) is prototypical autoimmune disease driven by pathological T cell-B cell interactions1,2. Expansion of T follicular helper (TFH) and T peripheral helper (TPH) cells, two T cell populations that provide help to B cells, is a prominent feature of SLE3,4. Human TFH and TPH cells characteristically produce high levels of the B cell chemoattractant CXCL13 (refs. 5,6), yet regulation of T cell CXCL13 production and the relationship between CXCL13+ T cells and other T cell states remains unclear. Here, we identify an imbalance in CD4+ T cell phenotypes in patients with SLE, with expansion of PD-1+/ICOS+ CXCL13+ T cells and reduction of CD96hi IL-22+ T cells. Using CRISPR screens, we identify the aryl hydrocarbon receptor (AHR) as a potent negative regulator of CXCL13 production by human CD4+ T cells. Transcriptomic, epigenetic and functional studies demonstrate that AHR coordinates with AP-1 family member JUN to prevent CXCL13+ TPH/TFH cell differentiation and promote an IL-22+ phenotype. Type I interferon, a pathogenic driver of SLE7, opposes AHR and JUN to promote T cell production of CXCL13. These results place CXCL13+ TPH/TFH cells on a polarization axis opposite from T helper 22 (TH22) cells and reveal AHR, JUN and interferon as key regulators of these divergent T cell states.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , CD4-Positive T-Lymphocytes , Chemokine CXCL13 , Interferon Type I , Lupus Erythematosus, Systemic , Proto-Oncogene Proteins c-jun , Receptors, Aryl Hydrocarbon , Female , Humans , Male , Basic Helix-Loop-Helix Transcription Factors/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Chemokine CXCL13/metabolism , Epigenomics , Gene Expression Profiling , Interferon Type I/immunology , Interferon Type I/metabolism , Interleukin-22/immunology , Interleukin-22/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/genetics , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Aryl Hydrocarbon/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
3.
Sci Rep ; 13(1): 16357, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37773535

ABSTRACT

Disruptions in the gut epithelial barrier can lead to the development of chronic indications such as inflammatory bowel disease (IBD). Historically, barrier function has been assessed in cancer cell lines, which do not contain all human intestinal cell types, leading to poor translatability. To bridge this gap, we adapted human primary gut organoids grown as monolayers to quantify transcription factor phosphorylation, gene expression, cytokine production, and barrier function. In this work we describe and characterize a novel 96-well human gut organoid-derived monolayer system that enables quantitative assessment of candidate therapeutics. Normal human intestine differentiation patterns and barrier function were characterized and confirmed to recapitulate key aspects of in vivo biology. Next, cellular response to TNF-α (a central driver of IBD) was determined using a diverse cadre of quantitative readouts. We showed that TNF-α pathway antagonists rescued damage caused by TNF-α in a dose-dependent manner, indicating that this system is suitable for quantitative assessment of barrier modulating factors. Taken together, we have established a robust primary cell-based 96-well system capable of interrogating questions around mucosal response. This system is well suited to provide pivotal functional data to support translational target and drug discovery efforts.


Subject(s)
Inflammatory Bowel Diseases , Tumor Necrosis Factor-alpha , Humans , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Intestinal Mucosa/metabolism , Epithelial Cells/metabolism , Inflammatory Bowel Diseases/metabolism , Organoids/metabolism
4.
Arthritis Rheumatol ; 74(11): 1808-1821, 2022 11.
Article in English | MEDLINE | ID: mdl-35644031

ABSTRACT

OBJECTIVES: To investigate the immune cell profiles of patients with systemic lupus erythematosus (SLE), and to identify longitudinal changes in those profiles over time. METHODS: We employed mass cytometry with 3 different panels of 38-39 markers (an immunophenotyping panel, a T cell/monocyte panel, and a B cell panel) in cryopreserved peripheral blood mononuclear cells (PBMCs) from 9 patients with early SLE, 15 patients with established SLE, and 14 controls without autoimmune disease. We used machine learning-driven clustering, flow self-organizing maps, and dimensional reduction with t-distributed stochastic neighbor embedding to identify unique cell populations in early SLE and established SLE. We used mass cytometry data of PBMCs from 19 patients with early rheumatoid arthritis (RA) and 23 controls to compare levels of specific cell populations in early RA and SLE. For the 9 patients with early SLE, longitudinal mass cytometry analysis was applied to PBMCs at enrollment, 6 months after enrollment, and 1 year after enrollment. Serum samples were also assayed for 65 cytokines using Luminex multiplex assay, and associations between cell types and cytokines/chemokines were assessed. RESULTS: Levels of peripheral helper T cells, follicular helper T (Tfh) cells, and several Ki-67+ proliferating subsets (ICOS+Ki-67+ CD8 T cells, Ki-67+ regulatory T cells, CD19intermediate Ki-67high plasmablasts, and PU.1high Ki-67high monocytes) were increased in patients with early SLE, with more prominent alterations than were seen in patients with early RA. Longitudinal mass cytometry and multiplex serum cytokine assays of samples from patients with early SLE revealed that levels of Tfh cells and CXCL10 had decreased 1 year after enrollment. Levels of CXCL13 were positively correlated with levels of several of the expanded cell populations in early SLE. CONCLUSION: Two major helper T cell subsets and unique Ki-67+ proliferating immune cell subsets were expanded in patients in the early phase of SLE, and the immunologic features characteristic of early SLE evolved over time.


Subject(s)
Leukocytes, Mononuclear , Lupus Erythematosus, Systemic , Humans , Leukocytes, Mononuclear/metabolism , Ki-67 Antigen , Interleukins , Cytokines
5.
JCI Insight ; 4(20)2019 10 17.
Article in English | MEDLINE | ID: mdl-31536480

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by pathologic T cell-B cell interactions and autoantibody production. Defining the T cell populations that drive B cell responses in SLE may enable design of therapies that specifically target pathologic cell subsets. Here, we evaluated the phenotypes of CD4+ T cells in the circulation of 52 SLE patients drawn from multiple cohorts and identified a highly expanded PD-1hiCXCR5-CD4+ T cell population. Cytometric, transcriptomic, and functional assays demonstrated that PD-1hiCXCR5-CD4+ T cells from SLE patients are T peripheral helper (Tph) cells, a CXCR5- T cell population that stimulates B cell responses via IL-21. The frequency of Tph cells, but not T follicular helper (Tfh) cells, correlated with both clinical disease activity and the frequency of CD11c+ B cells in SLE patients. PD-1hiCD4+ T cells were found within lupus nephritis kidneys and correlated with B cell numbers in the kidney. Both IL-21 neutralization and CRISPR-mediated deletion of MAF abrogated the ability of Tph cells to induce memory B cell differentiation into plasmablasts in vitro. These findings identify Tph cells as a highly expanded T cell population in SLE and suggest a key role for Tph cells in stimulating pathologic B cell responses.


Subject(s)
B-Lymphocytes/immunology , Interleukins/metabolism , Lupus Erythematosus, Systemic/immunology , Proto-Oncogene Proteins c-maf/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Adult , Aged , CD11c Antigen/metabolism , CRISPR-Cas Systems/genetics , Case-Control Studies , Cell Communication/drug effects , Cell Communication/genetics , Cell Communication/immunology , Cell Culture Techniques , Cell Separation , Cells, Cultured , Coculture Techniques , Female , Flow Cytometry , Gene Knockout Techniques , Humans , Interleukins/antagonists & inhibitors , Lupus Erythematosus, Systemic/blood , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Male , Middle Aged , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-maf/genetics , RNA-Seq , Receptors, CXCR5/metabolism , T-Lymphocytes, Helper-Inducer/metabolism
6.
Innate Immun ; 25(2): 132-143, 2019 02.
Article in English | MEDLINE | ID: mdl-30774010

ABSTRACT

Crohn's disease (CD) is a chronic disorder of the gastrointestinal tract characterized by inflammation and intestinal epithelial injury. Loss of function mutations in the intracellular bacterial sensor NOD2 are major risk factors for the development of CD. In the absence of robust bacterial recognition by NOD2 an inflammatory cascade is initiated through alternative PRRs leading to CD. In the present study, MCC950, a specific small molecule inhibitor of NLR pyrin domain-containing protein 3 (NLRP3), abrogated dextran sodium sulfate (DSS)-induced intestinal inflammation in Nod2-/- mice. NLRP3 inflammasome formation was observed at a higher rate in NOD2-deficient small intestinal lamina propria cells after insult by DSS. NLRP3 complex formation led to an increase in IL-1ß secretion in both the small intestine and colon of Nod2ko mice. This increase in IL-1ß secretion in the intestine was attenuated by MCC950 leading to decreased disease severity in Nod2ko mice. Our work suggests that NLRP3 inflammasome activation may be a key driver of intestinal inflammation in the absence of functional NOD2. NLRP3 pathway inhibition can prevent intestinal inflammation in the absence of robust NOD2 signaling.


Subject(s)
Colitis/immunology , Crohn Disease/immunology , Gastrointestinal Microbiome/immunology , Inflammasomes/metabolism , Intestinal Mucosa/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Animals , Colitis/chemically induced , Dextran Sulfate , Disease Models, Animal , Furans/administration & dosage , Furans/pharmacology , Heterocyclic Compounds, 4 or More Rings , Humans , Indenes , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Signal Transduction , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Sulfones
7.
PLoS One ; 12(7): e0180870, 2017.
Article in English | MEDLINE | ID: mdl-28719615

ABSTRACT

While the immune system is essential for the maintenance of the homeostasis, health and survival of humans, aberrant immune responses can lead to chronic inflammatory and autoimmune disorders. Pharmacological modulation of drug targets in the immune system to ameliorate disease also carry a risk of immunosuppression that could lead to adverse outcomes. Therefore, it is important to understand the 'immune fingerprint' of novel therapeutics as they relate to current and, clinically used immunological therapies to better understand their potential therapeutic benefit as well as immunosuppressive ability that might lead to adverse events such as infection risks and cancer. Since the mechanistic investigation of pharmacological modulators in a drug discovery setting is largely compound- and mechanism-centric but not comprehensive in terms of immune system impact, we developed a human tissue based functional assay platform to evaluate the impact of pharmacological modulators on a range of innate and adaptive immune functions. Here, we demonstrate that it is possible to generate a qualitative and quantitative immune system impact of pharmacological modulators, which might help better understand and predict the benefit-risk profiles of these compounds in the treatment of immune disorders.


Subject(s)
Drug Evaluation, Preclinical/methods , Immune System/drug effects , Small Molecule Libraries/pharmacology , Chemokines/biosynthesis , Humans , Immune System/cytology , Immune System/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Phagocytes/drug effects , Phagocytes/immunology , Phagocytes/metabolism , Reactive Oxygen Species/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Toll-Like Receptors/metabolism , Transcriptome/drug effects
8.
Br J Pharmacol ; 173(21): 3080-3087, 2016 11.
Article in English | MEDLINE | ID: mdl-27417329

ABSTRACT

BACKGROUND AND PURPOSE: Asthma presents as a heterogeneous syndrome characterized by airway obstruction, inflammation and hyper-reactivity (AHR). Spleen tyrosine kinase (Syk) mediates allergen-induced mast cell degranulation, a central component of allergen-induced inflammation and AHR. However, the role of Syk in IgE-mediated constriction of human small airways remains unknown. In this study, we addressed whether selective inhibition of Syk attenuates IgE-mediated constriction and mast cell mediator release in human small airways. EXPERIMENTAL APPROACH: Human precision cut lung slices (hPCLS) ex vivo derived from non-asthmatic donors were incubated overnight with human IgE, dexamethasone, montelukast, antihistamines or a selective Syk inhibitor (SYKi). High-affinity IgE receptor (FcεRI) activation by anti-IgE cross-linking was performed, and constriction and mediator release measured. Airway constriction was normalized to that induced by maximal carbachol stimulation. Syk expression (determined by qPCR and immunoblot) was also evaluated in human primary airway smooth muscle (HASM) cells to determine whether Syk directly modulates HASM function. KEY RESULTS: While dexamethasone had little effect on FcεR-mediated contraction, montelukast or antihistamines partially attenuated the response. SYKi abolished anti-IgE-mediated contraction and suppressed the release of mast cell or basophil mediators from the IgE-treated hPCLS. In contrast, SYKi had little effect on the non-allergic contraction induced by carbachol. Syk mRNA and protein were undetectable in HASM cells. CONCLUSIONS AND IMPLICATIONS: A selective Syk inhibitor, but not corticosteroids, abolished FcεR-mediated contraction in human small airways ex vivo. The mechanism involved FcεRI receptor activation on mast cells or basophils that degranulate causing airway constriction, rather than direct actions on HASM.


Subject(s)
Immunoglobulin E/immunology , Lung/drug effects , Muscle, Smooth/drug effects , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Spleen/enzymology , Cells, Cultured , Humans , In Vitro Techniques , Lung/cytology , Lung/enzymology , Lung/immunology , Muscle Contraction/drug effects , Muscle Contraction/immunology , Muscle, Smooth/enzymology , Muscle, Smooth/immunology , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/metabolism
9.
J Steroid Biochem Mol Biol ; 163: 88-97, 2016 10.
Article in English | MEDLINE | ID: mdl-27106747

ABSTRACT

The androgen receptor (AR) is a member of the nuclear hormone receptor super family of transcription factors. Androgens play an essential role in the development, growth, and maintenance of male sex organs, as well as the musculoskeletal and central nervous systems. Yet with advancing age, androgens can drive the onset of prostate cancer, the second leading cause of cancer death in males within the United States. Androgen deprivation therapy (ADT) by pharmacologic and/or surgical castration induces apoptosis of prostate cells and subsequent shrinkage of the prostate and prostate tumors. However, ADT is associated with significant musculoskeletal and behavioral adverse effects. The unique pharmacological activity of selective androgen receptor modulator (SARM) MK-4541 recently has been reported as an AR antagonist with 5α-reductase inhibitor function. The molecule inhibits proliferation and induces apoptosis in AR positive, androgen dependent prostate cancer cells. Importantly, MK-4541 inhibited androgen-dependent prostate growth in male rats yet maintained lean body mass and bone formation following ovariectomy in female rats. In the present study, we evaluated the effects of SARM MK-4541 in the androgen-dependent Dunning R3327-G prostate carcinoma xenograft mouse model as well as on skeletal muscle mass and function, and AR-regulated behavior in mice. MK-4541 significantly inhibited the growth of R3327-G prostate tumors, exhibited anti-androgen effects on the seminal vesicles, reduced plasma testosterone concentrations in intact males, and inhibited Ki67 expression. MK-4541 treated xenografts appeared similar to xenografts in castrated mice. Importantly, we demonstrate that MK-4541 exhibited anabolic activity in androgen deficient conditions, increasing lean body mass and muscle function in adult castrated mice. Moreover, MK-4541 treatment restored general activity levels in castrated mice. Thus, MK-4541 exhibits an optimum profile as an adjuvant therapy to ADT which may provide potent anti-androgenic activity at the prostate yet protective activity on skeletal muscle and behavior in patients.


Subject(s)
Anabolic Agents/pharmacology , Androgen Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Azasteroids/pharmacology , Carbamates/pharmacology , Carcinoma/drug therapy , Muscle, Skeletal/drug effects , Prostatic Neoplasms/drug therapy , Animals , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Chemotherapy, Adjuvant/methods , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Male , Mice , Muscle, Skeletal/metabolism , Orchiectomy , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Testosterone/antagonists & inhibitors , Testosterone/metabolism , Xenograft Model Antitumor Assays
10.
Nat Methods ; 13(2): 151-7, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26689262

ABSTRACT

Here we describe the development of a human lung 'small airway-on-a-chip' containing a differentiated, mucociliary bronchiolar epithelium and an underlying microvascular endothelium that experiences fluid flow, which allows for analysis of organ-level lung pathophysiology in vitro. Exposure of the epithelium to interleukin-13 (IL-13) reconstituted the goblet cell hyperplasia, cytokine hypersecretion and decreased ciliary function of asthmatics. Small airway chips lined with epithelial cells from individuals with chronic obstructive pulmonary disease recapitulated features of the disease such as selective cytokine hypersecretion, increased neutrophil recruitment and clinical exacerbation by exposure to viral and bacterial infections. With this robust in vitro method for modeling human lung inflammatory disorders, it is possible to detect synergistic effects of lung endothelium and epithelium on cytokine secretion, identify new biomarkers of disease exacerbation and measure responses to anti-inflammatory compounds that inhibit cytokine-induced recruitment of circulating neutrophils under flow.


Subject(s)
Epithelium/drug effects , Inflammation/metabolism , Interleukin-13/pharmacology , Lab-On-A-Chip Devices , Lung Diseases/drug therapy , Lung Diseases/metabolism , Humans , Inflammation/pathology , Tissue Culture Techniques
11.
PLoS One ; 10(2): e0118286, 2015.
Article in English | MEDLINE | ID: mdl-25706956

ABSTRACT

OBJECTIVES: Human airway epithelial cells are the principal target of human rhinovirus (HRV), a common cold pathogen that triggers the majority of asthma exacerbations. The objectives of this study were 1) to evaluate an in vitro air liquid interface cultured human airway epithelial cell model for HRV infection, and 2) to identify gene expression patterns associated with asthma intrinsically and/or after HRV infection using this model. METHODS: Air-liquid interface (ALI) human airway epithelial cell cultures were prepared from 6 asthmatic and 6 non-asthmatic donors. The effects of rhinovirus RV-A16 on ALI cultures were compared. Genome-wide gene expression changes in ALI cultures following HRV infection at 24 hours post exposure were further analyzed using RNA-seq technology. Cellular gene expression and cytokine/chemokine secretion were further evaluated by qPCR and a Luminex-based protein assay, respectively. MAIN RESULTS: ALI cultures were readily infected by HRV. RNA-seq analysis of HRV infected ALI cultures identified sets of genes associated with asthma specific viral responses. These genes are related to inflammatory pathways, epithelial structure and remodeling and cilium assembly and function, including those described previously (e.g. CCL5, CXCL10 and CX3CL1, MUC5AC, CDHR3), and novel ones that were identified for the first time in this study (e.g. CCRL1). CONCLUSIONS: ALI-cultured human airway epithelial cells challenged with HRV are a useful translational model for the study of HRV-induced responses in airway epithelial cells, given that gene expression profile using this model largely recapitulates some important patterns of gene responses in patients during clinical HRV infection. Furthermore, our data emphasize that both abnormal airway epithelial structure and inflammatory signaling are two important asthma signatures, which can be further exacerbated by HRV infection.


Subject(s)
Asthma/genetics , Asthma/virology , Cell Differentiation/genetics , Epithelial Cells/virology , Picornaviridae Infections/genetics , Respiratory System/virology , Adolescent , Adult , Cells, Cultured , Chemokines/genetics , Child , Female , Gene Expression/genetics , Humans , Inflammation/genetics , Inflammation/virology , Male , Middle Aged , Picornaviridae Infections/virology , Rhinovirus , Signal Transduction/genetics
12.
J Steroid Biochem Mol Biol ; 143: 29-39, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24565564

ABSTRACT

Prostate cancer (PCa) initially responds to inhibition of androgen receptor (AR) signaling, but inevitably progresses to hormone ablation-resistant disease. Much effort is focused on optimizing this androgen deprivation strategy by improving hormone depletion and AR antagonism. However we found that bicalutamide, a clinically used antiandrogen, actually resembles a selective AR modulator (SARM), as it partially regulates 24% of endogenously 5α-dihydrotestosterone (DHT)-responsive genes in AR(+) MDA-MB-453 breast cancer cells. These data suggested that passive blocking of all AR functions is not required for PCa therapy. Hence, we adopted an active strategy that calls for the development of novel SARMs, which induce a unique gene expression profile that is intolerable to PCa cells. Therefore, we screened 3000 SARMs for the ability to arrest the androgen-independent growth of AR(+) 22Rv1 and LNCaP PCa cells but not AR(-) PC3 or DU145 cells. We identified only one such compound; the 4-aza-steroid, MK-4541, a potent and selective SARM. MK-4541 induces caspase-3 activity and cell death in both androgen-independent, AR(+) PCa cell lines but spares AR(-) cells or AR(+) non-PCa cells. This activity correlates with its promoter context- and cell-type dependent transcriptional effects. In rats, MK-4541 inhibits the trophic effects of DHT on the prostate, but not the levator ani muscle, and triggers an anabolic response in the periosteal compartment of bone. Therefore, MK-4541 has the potential to effectively manage prostatic hypertrophic diseases owing to its antitumor SARM-like mechanism, while simultaneously maintaining the anabolic benefits of natural androgens.


Subject(s)
Anabolic Agents/pharmacology , Apoptosis/drug effects , Azasteroids/pharmacology , Breast Neoplasms/pathology , Carbamates/pharmacology , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms/pathology , Receptors, Androgen/chemistry , Anabolic Agents/chemistry , Androgen Receptor Antagonists/pharmacology , Androgens/pharmacology , Animals , Azasteroids/chemistry , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Carbamates/chemistry , Cell Proliferation/drug effects , Combinatorial Chemistry Techniques , Female , Humans , Male , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
13.
J Gerontol A Biol Sci Med Sci ; 68(10): 1181-92, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23525481

ABSTRACT

Myostatin is a highly conserved member of the transforming growth factor-ß ligand family known to regulate muscle growth via activation of activin receptors. A fusion protein consisting of the extracellular ligand-binding domain of activin type IIB receptor with the Fc portion of human immunoglobulin G (ActRIIB-Fc) was used to inhibit signaling through this pathway. Here, we study the effects of this fusion protein in adult, 18-month-old, and orchidectomized mice. Significant muscle growth and enhanced muscle function were observed in adult mice treated for 3 days with ActRIIB-Fc. The ActRIIB-Fc-treated mice had enhanced fast fatigable muscle function, with only minor enhancement of fatigue-resistant fiber function. The ActRIIB-Fc-treated 18-month-old mice and orchidectomized mice showed significantly improved muscle function. Treatment with ActRIIB-Fc also increased bone mineral density and serum levels of a marker of bone formation. These observations highlight the potential of targeting ActRIIB receptor to treat age-related and hypogonadism-associated musculoskeletal degeneration.


Subject(s)
Activin Receptors, Type II/pharmacology , Aging/drug effects , Aging/physiology , Bone Density/drug effects , Muscle Contraction/drug effects , Activin Receptors, Type II/metabolism , Animals , Biomarkers/blood , Bone Density/physiology , Cell Line , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin Fc Fragments/pharmacology , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/physiology , Muscle Strength/drug effects , Muscle Strength/physiology , Myostatin/metabolism , Orchiectomy , Peptide Fragments/blood , Procollagen/blood , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Sarcopenia/drug therapy , Sarcopenia/pathology , Sarcopenia/physiopathology
14.
Biomarkers ; 17(2): 172-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22299632

ABSTRACT

Estrogen Receptor α (ERα) and Estrogen Receptor ß (ERß) are steroid nuclear receptors that transduce estrogen signaling to control diverse physiological processes linked to reproduction, bone remodeling, behavior, immune response and endocrine-related diseases. In order to differentiate between ERα and ERß mediated effects in vivo, ER subtype selective biomarkers are essential. We utilized ERα knockout (AERKO) and ERß knockout (BERKO) mouse liver RNA and genome wide profiling to identify novel ERα selective serum biomarker candidates. Results from the gene array experiments were validated using real-time RT-PCR and subsequent ELISA's to demonstrate changes in serum proteins. Here we present data that Lipopolysacharide Binding Protein (LBP) is a novel liver-derived ERα selective biomarker that can be measured in serum.


Subject(s)
Biomarkers/blood , Blood Proteins/analysis , Carrier Proteins/blood , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Membrane Glycoproteins/blood , RNA, Messenger/biosynthesis , Acute-Phase Proteins , Animals , Estradiol/administration & dosage , Estrogen Receptor alpha/deficiency , Estrogen Receptor beta/deficiency , Female , Gene Expression Profiling , Gene Expression Regulation , Liver/drug effects , Liver/metabolism , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , Rats , Uterus/drug effects , Uterus/metabolism
15.
BMC Musculoskelet Disord ; 12: 246, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22035016

ABSTRACT

BACKGROUND: Age-related sarcopenia is a disease state of loss of muscle mass and strength that affects physical function and mobility leading to falls, fractures, and disability. The need for therapies to treat age-related sarcopenia has attracted intensive preclinical research. To facilitate the discovery of these therapies, we have developed a non-invasive rat muscle functional assay system to efficiently measure muscle force and evaluate the efficacy of drug candidates. METHODS: The lower leg muscles of anesthetized rats are artificially stimulated with surface electrodes on the knee holders and the heel support, causing the lower leg muscles to push isometric pedals that are attached to force transducers. We developed a stimulation protocol to perform a fatigability test that reveals functional muscle parameters like maximal force, the rate of fatigue, fatigue-resistant force, as well as a fatigable muscle force index. The system is evaluated in a rat aging model and a rat glucocorticoid-induced muscle loss model RESULTS: The aged rats were generally weaker than adult rats and showed a greater reduction in their fatigable force when compared to their fatigue-resistant force. Glucocorticoid treated rats mostly lost fatigable force and fatigued at a higher rate, indicating reduced force from glycolytic fibers with reduced energy reserves. CONCLUSIONS: The involuntary contraction assay is a reliable system to assess muscle function in rodents and can be applied in preclinical research, including age-related sarcopenia and other myopathy.


Subject(s)
Aging/physiology , Isometric Contraction/physiology , Muscle Fatigue/physiology , Sarcopenia/physiopathology , Age Factors , Aging/drug effects , Animals , Biological Assay , Dexamethasone/pharmacology , Disease Models, Animal , Electric Stimulation , Glucocorticoids/pharmacology , Isometric Contraction/drug effects , Male , Muscle Fatigue/drug effects , Rats , Rats, Sprague-Dawley
16.
J Pharmacol Exp Ther ; 338(1): 220-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21459965

ABSTRACT

Both the physiological role of muscarinic receptors for bladder function and the therapeutic efficacy of antimuscarinic agents for overactive bladder syndrome are well documented. We investigated the effect of antimuscarinic agents with different subtype selectivity on urodynamic parameters in nonhuman primates and rodents and compared plasma levels of these agents between species. Anesthetized rhesus monkeys were transurethrally catheterized, and the bladder was infused with saline. Urodynamic parameters were measured before and after intravenous drug administration. Tolterodine (nonselective) and oxybutynin (moderately M(3)-selective) increased bladder capacity at lower doses than those required to decrease micturition pressure. However, higher doses of darifenacin (M(3)-selective) were needed to increase the bladder capacity than those needed to decrease the micturition pressure. In rats, tolterodine had no effect on the bladder capacity but decreased the micturition pressure at all of the doses administered. Oxybutynin also decreased micturition pressure and increased bladder capacity at the highest dose. Plasma levels of these drugs overlap in both species. These results suggest that, in addition to the M(3) receptor, other muscarinic receptor subtypes contribute to regulate bladder storage function in nonhuman primates, since less subtype-selective tolterodine and oxybutynin showed higher specificity to the bladder capacity effect than the effect on micturition pressure compared with M(3)-selective darifenacin. In addition, the role of muscarinic receptors in bladder storage function varies between primates and rodents. Compared with rodents, muscarinic receptors may play a more active role during the storage phase to regulate the functional bladder capacity in primates.


Subject(s)
Muscarinic Antagonists/pharmacology , Urinary Bladder/drug effects , Urinary Bladder/physiology , Animals , Benzhydryl Compounds/pharmacology , Cresols/pharmacology , Female , Macaca mulatta , Mandelic Acids/pharmacology , Phenylpropanolamine/pharmacology , Rats , Rats, Sprague-Dawley , Species Specificity , Tolterodine Tartrate
17.
J Steroid Biochem Mol Biol ; 122(5): 310-7, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20800684

ABSTRACT

We assessed the effects of subtype-selective ER agonists on monoamine levels in discrete regions of the female rat brain. Ovariectomized (ovx) rats were treated for 4 days with vehicle, 17ß-estradiol (E; 0.05mg/kg), an ERß agonist (C19; 3mg/kg) or an ERα agonist (PPT; 3mg/kg) and samples from brain regions were assessed for monoamines and metabolites. We also assessed effects of ERß modulation on baseline and fenfluramine-induced release of monoamines in hippocampus using microdialysis. In the first study, E and the ERα agonist increased norepinephrine in cortex and all three ER ligands increased it in the ventral hippocampus. Changes in levels of the noradrenergic metabolite, MHPG and the dopaminergic metabolite, DOPAC were noted in brain areas of ER ligand-treated animals. E also increased levels of 5HIAA in three brain areas. In the microdialysis study, there were no differences among groups in baseline levels of monoamines. However, E and the ERß agonist increased levels of the dopaminergic metabolite, HVA following fenfluramine. In summary, activation of the two nuclear ERs with selective agonists affects monoamine and metabolite levels in discrete brain areas, a number of which are known to play key roles in cognitive and affective function.


Subject(s)
Biogenic Monoamines/metabolism , Cerebral Cortex/drug effects , Estradiol/pharmacology , Fluorenes/pharmacology , Hippocampus/drug effects , Phenols/pharmacology , Pyrazoles/pharmacology , Receptors, Estrogen/agonists , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Cerebral Cortex/metabolism , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Female , Fenfluramine/pharmacology , Hippocampus/metabolism , Hydroxyindoleacetic Acid/metabolism , Methoxyhydroxyphenylglycol/metabolism , Norepinephrine/metabolism , Ovariectomy , Rats , Rats, Sprague-Dawley
18.
Endocrinology ; 149(7): 3306-12, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18356271

ABSTRACT

Immunocytochemical studies have shown that nuclear and extranuclear estrogen receptors (ERs) are present in several extrahypothalamic brain regions. The goal of this study was to determine the subcellular location of functional ERs, particularly extranuclear ERs, by demonstrating (125)I-estradiol binding in the rat forebrain and medullary sections prepared for light and electron microscopic autoradiography. Some sections were immunocytochemically labeled with the catecholamine-synthesizing enzyme, tyrosine hydroxylase (TH), prior to the autoradiographic procedure. By light microscopy, dense accumulations of silver grains denoting (125)I-estradiol binding were observed over cells in the ventromedial and arcuate hypothalamic nuclei, amygdala, and nucleus of the solitary tract. In sections labeled for TH, large accumulations of silver grains were admixed with TH-labeled processes in the medial nucleus of the amygdala and over TH-labeled perikarya in the medial and commissural nucleus of the solitary tract. Electron microscopic analyses were focused on the rostral ventrolateral medulla and the hippocampal CA1 region, two regions previously shown to have extranuclear ERs. In the rostral ventrolateral medulla, silver grains indicative of (125)I-estradiol binding were found within a few large terminals, affiliated with mitochondria. In the hippocampus, autoradiographic silver grains denoting (125)I-estradiol binding were associated with mitochondria in dendritic shafts or were near synaptic specializations on dendritic spines. These patterns of silver grain labeling were not seen in sections from rats that received (125)I-estradiol combined with cold estradiol. The association of (125)I-estradiol binding with pre- and postsynaptic profiles supports a functional role for nonnuclear ERs in brain.


Subject(s)
Estrogens/metabolism , Prosencephalon/metabolism , Receptors, Estrogen/metabolism , Animals , Binding Sites , Estradiol/metabolism , Female , Immunohistochemistry , Iodine Radioisotopes , Microscopy, Electron , Models, Biological , Prosencephalon/ultrastructure , Protein Binding , Rats , Rats, Sprague-Dawley
19.
Brain Res ; 1094(1): 163-78, 2006 Jun 13.
Article in English | MEDLINE | ID: mdl-16696957

ABSTRACT

Blood pressure in women increases after menopause, and sympathetic tone in female rats decreases with estrogen injections in the rostral ventrolateral medulla (RVLM) region that contains bulbospinal C1 adrenergic neurons and is involved in blood pressure control. We investigated the anatomical and physiological basis for estrogen effects in the RVLM. Neurons with alpha- or beta-subtypes of estrogen receptor (ER) immunoreactivity (-ir) overlapped in distribution with tyrosine hydroxylase (TH)-containing C1 neurons. Immunoelectron microscopy revealed that ERalpha- and ERbeta-ir had distinct cellular and subcellular distributions. ERalpha-ir was most commonly in TH-lacking profiles, many of which were axons and peptide-containing afferents that contacted TH-containing dendrites. ERalpha-ir was also in some TH-containing dendrites. ERbeta-ir was most frequently in TH-containing somata and dendrites, particularly on endoplasmic reticula, mitochondria, and plasma membranes. In whole-cell patch clamp recordings from isolated bulbospinal RVLM neurons, 17beta-estradiol dose-dependently reduced voltage-gated Ca(++) currents, especially the long-lasting (L-type) component. This inhibition was reversed by washing or prevented by adding the non-subtype-selective ER antagonist ICI182780. An ERbeta-selective agonist, but not an ERalpha-selective agonist, reproduced the Ca(++) current inhibition. The data indicate that estrogens can modulate the function of RVLM C1 bulbospinal neurons either directly, through extranuclear ERbeta, or indirectly through extranuclear ERalpha in selected afferents. Moreover, Ca(++) current inhibition may underlie the decrease in sympathetic tone evoked by local 17beta-estradiol application. These findings provide a structural and functional basis for the effects of estrogens on blood pressure control and suggest a mechanism for the modulation of cardiovascular function by estrogen in women.


Subject(s)
Efferent Pathways/metabolism , Epinephrine/metabolism , Estrogens/metabolism , Medulla Oblongata/metabolism , Spinal Cord/metabolism , Sympathetic Nervous System/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Dendrites/metabolism , Dendrites/ultrastructure , Dose-Response Relationship, Drug , Efferent Pathways/drug effects , Efferent Pathways/ultrastructure , Estradiol/metabolism , Estradiol/pharmacology , Estradiol Congeners/pharmacology , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/agonists , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Female , Hypertension/etiology , Hypertension/physiopathology , Medulla Oblongata/drug effects , Medulla Oblongata/ultrastructure , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques , Postmenopause/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/ultrastructure , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/ultrastructure , Tyrosine 3-Monooxygenase/metabolism
20.
J Comp Neurol ; 491(2): 81-95, 2005 Oct 17.
Article in English | MEDLINE | ID: mdl-16127691

ABSTRACT

Several lines of evidence indicate that estrogen affects hippocampal synaptic plasticity through rapid nongenomic mechanisms, possibly by binding to plasma membrane estrogen receptors (ERs). We have previously shown that ERalpha immunoreactivity (ir) is in select interneuron nuclei and in several extranuclear locations, including dendritic spines and axon terminals, within the rat hippocampal formation (Milner et al., [2001] J Comp Neurol 429:355). The present study sought to determine the cellular and subcellular locations of ERbeta-ir. Coronal hippocampal sections from diestrus rats were immunolabeled with antibodies to ERbeta and examined by light and electron microscopy. By light microscopy, ERbeta-ir was primarily in the perikarya and proximal dendrites of pyramidal and granule cells. ERbeta-ir was also in a few nonprincipal cells and scattered nuclei in the ventral subiculum and CA3 region. Ultrastructural analysis revealed ERbeta-ir at several extranuclear sites in all hippocampal subregions. ERbeta-ir was affiliated with cytoplasmic organelles, especially endomembranes and mitochondria, and with plasma membranes primarily of principal cell perikarya and proximal dendrites. ERbeta-ir was in dendritic spines, many arising from pyramidal and granule cell dendrites. In both dendritic shafts and spines, ERbeta-ir was near the perisynaptic zone adjacent to synapses formed by unlabeled terminals. ERbeta-ir was in preterminal axons and axon terminals, associated with clusters of small, synaptic vesicles. ERbeta-labeled terminals formed both asymmetric and symmetric synapses with dendrites. ERbeta-ir also was detected in glial profiles. The cellular and subcellular localization of ERbeta-ir was generally similar to that of ERalpha, except that ERbeta was more extensively found at extranuclear sites. These results suggest that ERbeta may serve primarily as a nongenomic transducer of estrogen actions in the hippocampal formation.


Subject(s)
Estrogen Receptor beta/metabolism , Hippocampus/ultrastructure , Neurons/ultrastructure , Animals , Axons/metabolism , Axons/ultrastructure , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Female , Hippocampus/metabolism , Immunohistochemistry , Microglia/metabolism , Microglia/ultrastructure , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , Synapses/metabolism , Synapses/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL