Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 222: 116095, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38423186

ABSTRACT

Aromatase is the rate-limiting enzyme in the biosynthesis of estrogens and a key risk factor for hormone receptor-positive breast cancer. In postmenopausal women, estrogens synthesized in adipose tissue promotes the growth of estrogen receptor positive breast cancers. Activation of peroxisome proliferator-activated receptor gamma (PPARγ) in adipose stromal cells (ASCs) leads to decreased expression of aromatase and differentiation of ASCs into adipocytes. Environmental chemicals can act as antagonists of PPARγ and disrupt its function. This study aimed to test the hypothesis that PPARγ antagonists can promote breast cancer by stimulating aromatase expression in human adipose tissue. Primary cells and explants from human adipose tissue as well as A41hWAT, C3H10T1/2, and H295R cell lines were used to investigate PPARγ antagonist-stimulated effects on adipogenesis, aromatase expression, and estrogen biosynthesis. Selected antagonists inhibited adipocyte differentiation, preventing the adipogenesis-associated downregulation of aromatase. NMR spectroscopy confirmed direct interaction between the potent antagonist DEHPA and PPARγ, inhibiting agonist binding. Short-term exposure of ASCs to PPARγ antagonists upregulated aromatase only in differentiated cells, and a similar effect could be observed in human breast adipose tissue explants. Overexpression of PPARG with or without agonist treatment reduced aromatase expression in ASCs. The data suggest that environmental PPARγ antagonists regulate aromatase expression in adipose tissue through two mechanisms. The first is indirect and involves inhibition of adipogenesis, while the second occurs more acutely.


Subject(s)
Breast Neoplasms , PPAR gamma , Female , Humans , PPAR gamma/genetics , PPAR gamma/metabolism , Aromatase/genetics , Aromatase/metabolism , Adipose Tissue/metabolism , Estrogens/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Adipogenesis
2.
Nutrients ; 15(2)2023 Jan 11.
Article in English | MEDLINE | ID: mdl-36678247

ABSTRACT

Vitamin A and D deficiencies are associated with immune modulatory effects and intestinal barrier impairment. However, the underlying mechanisms remain unclear. C57BL/6J mice were fed either a diet lacking in vitamin A (VAd), vitamin D (VDd) or a control diet (CD) for 12 weeks. Gut barrier function, antimicrobial peptide (AMP) defense and regulatory pathways were assessed. VAd mice compared to CD mice showed a reduced villus length in the ileum (p < 0.01) and decreased crypt depth in the colon (p < 0.05). In both VAd- and VDd-fed mice, ileal α-defensin 5 (p < 0.05/p < 0.0001 for VAd/VDd) and lysozyme protein levels (p < 0.001/p < 0.0001) were decreased. Moreover, mRNA expression of lysozyme (p < 0.05/p < 0.05) and total cryptdins (p < 0.001/p < 0.01) were reduced compared to controls. Furthermore, matrix metalloproteinase-7 (Mmp7) mRNA (p < 0.0001/p < 0.001) as well as components of the Wnt signaling pathway were decreased. VAd- and VDd-fed mice, compared to control mice, exhibited increased expression of pro-inflammatory markers and ß-defensins in the colon. Organoid cell culture confirmed that vitamins A and D regulate AMP expression, likely through the Jak/STAT5 signaling pathway. In conclusion, our data show that vitamin A and D regulate intestinal antimicrobial peptide defense through Wnt and STAT5 signaling pathways.


Subject(s)
Muramidase , Vitamin A , Animals , Mice , STAT5 Transcription Factor , Mice, Inbred C57BL , Vitamins , Diet , Signal Transduction , RNA, Messenger/genetics , Antimicrobial Peptides
3.
FEBS J ; 290(10): 2673-2691, 2023 05.
Article in English | MEDLINE | ID: mdl-36595342

ABSTRACT

Exploring mechanisms responsible for brown adipose tissue's (BAT) high metabolic activity is crucial to exploit its energy-dissipating ability for therapeutic purposes. Basigin (Bsg), a multifunctional highly glycosylated transmembrane protein, was recently proposed as one of the 98 critical markers allowing to distinguish 'white' and 'brown' adipocytes, yet its function in thermogenic brown adipocytes is unknown. Here, we report that Bsg is negatively associated with obesity in mice. By contrast, Bsg expression increased in the mature adipocyte fraction of BAT upon cold acclimation. Additionally, Bsg levels were highly induced during brown adipocyte maturation in vitro and were further increased upon ß-adrenergic stimulation in a HIF-1α-dependent manner. siRNA-mediated Bsg gene silencing in cultured brown adipocytes did not impact adipogenesis nor mitochondrial function. However, a significant decrease in mitochondrial respiration, lipolysis and Ucp1 transcription was observed in adipocytes lacking Bsg, when activated by norepinephrine. Furthermore, using gas chromatography/mass spectrometry-time-of-flight analysis to assess the composition of cellular metabolites, we demonstrate that brown adipocytes lacking Bsg have lower levels of intracellular lactate and acetoacetate. Bsg was additionally required to regulate intracellular AcAc and tricarboxylic acid cycle intermediate levels in NE-stimulated adipocytes. Our study highlights the critical role of Bsg in active brown adipocytes, possibly by controlling cellular metabolism.


Subject(s)
Adipocytes, Brown , Adipose Tissue, Brown , Mice , Animals , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Basigin/metabolism , Lipolysis , Obesity/metabolism , Thermogenesis/genetics , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
4.
J Physiol ; 600(4): 885-902, 2022 02.
Article in English | MEDLINE | ID: mdl-34387373

ABSTRACT

KEY POINTS: Afadin is a ubiquitously expressed scaffold protein with a recently discovered role in insulin signalling and glucose metabolism. Insulin-stimulated phosphorylation of Afadin at S1795 occurs in insulin-responsive tissues such as adipose tissue, muscle, liver, pancreas and heart. Afadin abundance and AfadinS1795 phosphorylation are dynamically regulated in metabolic tissues during diet-induced obesity progression. Genetic silencing of AfadinS1795 phosphorylation improves glucose homeostasis in the early stages of diet-induced metabolic dysregulation. AfadinS1795 phosphorylation contributes to the early development of obesity-related complications in mice. ABSTRACT: Obesity is associated with systemic insulin resistance and numerous metabolic disorders. Yet, the mechanisms underlying impaired insulin action during obesity remain to be fully elucidated. Afadin is a multifunctional scaffold protein with the ability to modulate insulin action through its phosphorylation at S1795 in adipocytes. In the present study, we report that insulin-stimulated AfadinS1795 phosphorylation is not restricted to adipose tissues, but is a common signalling event in insulin-responsive tissues including muscle, liver, pancreas and heart. Furthermore, a dynamic regulation of Afadin abundance occurred during diet-induced obesity progression, while its phosphorylation was progressively attenuated. To investigate the role of AfadinS1795 phosphorylation in the regulation of whole-body metabolic homeostasis, we generated a phospho-defective mouse model (Afadin SA) in which the Afadin phosphorylation site was silenced (S1795A) at the whole-body level using CRISPR-Cas9-mediated gene editing. Metabolic characterization of these mice under basal physiological conditions or during a high-fat diet (HFD) challenge revealed that preventing AfadinS1795 phosphorylation improved insulin sensitivity and glucose tolerance and increased liver glycogen storage in the early stage of diet-induced metabolic dysregulation, without affecting body weight. Together, our findings reveal that AfadinS1795 phosphorylation in metabolic tissues is critical during obesity progression and contributes to promote systemic insulin resistance and glucose intolerance in the early phase of diet-induced obesity.


Subject(s)
Insulin Resistance , Animals , Diet, High-Fat , Glucose/metabolism , Homeostasis , Insulin/metabolism , Insulin Resistance/physiology , Mice , Mice, Inbred C57BL , Mice, Obese , Microfilament Proteins , Phosphorylation
5.
Cells ; 10(8)2021 07 29.
Article in English | MEDLINE | ID: mdl-34440697

ABSTRACT

Pancreatic stellate cells (PSCs) are important pancreatic fibrogenic cells that interact with pancreatic cancer cells to promote the progression of pancreatic ductal adenocarcinoma (PDAC). In the tumor microenvironment (TME), several factors such as cytokines and nucleotides contribute to this interplay. Our aim was to investigate whether there is an interaction between IL-6 and nucleotide signaling, in particular, that mediated by the ATP-sensing P2X7 receptor (P2X7R). Using human cell lines of PSCs and cancer cells, as well as primary PSCs from mice, we show that ATP is released from both PSCs and cancer cells in response to mechanical and metabolic cues that may occur in the TME, and thus activate the P2X7R. Functional studies using P2X7R agonists and inhibitors show that the receptor is involved in PSC proliferation, collagen secretion and IL-6 secretion and it promotes cancer cell migration in a human PSC-cancer cell co-culture. Moreover, conditioned media from P2X7R-stimulated PSCs activated the JAK/STAT3 signaling pathway in cancer cells. The monoclonal antibody inhibiting the IL-6 receptor, Tocilizumab, inhibited this signaling. In conclusion, we show an important mechanism between PSC-cancer cell interaction involving ATP and IL-6, activating P2X7 and IL-6 receptors, respectively, both potential therapeutic targets in PDAC.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Carcinoma, Pancreatic Ductal/metabolism , Interleukin-6/metabolism , Pancreatic Stellate Cells/metabolism , Receptors, Purinergic P2X7/metabolism , STAT3 Transcription Factor/metabolism , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/physiopathology , Cell Communication , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Male , Mice , Pancreatic Stellate Cells/physiology , Signal Transduction , Tumor Microenvironment
6.
Mol Metab ; 44: 101137, 2021 02.
Article in English | MEDLINE | ID: mdl-33285300

ABSTRACT

OBJECTIVE: Increasing adaptive thermogenesis by stimulating browning in white adipose tissue is a promising method of improving metabolic health. However, the molecular mechanisms underlying this transition remain elusive. Our study examined the molecular determinants driving the differentiation of precursor cells into thermogenic adipocytes. METHODS: In this study, we conducted temporal high-resolution proteomic analysis of subcutaneous white adipose tissue (scWAT) after cold exposure in mice. This was followed by loss- and gain-of-function experiments using siRNA-mediated knockdown and CRISPRa-mediated induction of gene expression, respectively, to evaluate the function of the transcriptional regulator Y box-binding protein 1 (YBX1) during adipogenesis of brown pre-adipocytes and mesenchymal stem cells. Transcriptomic analysis of mesenchymal stem cells following induction of endogenous Ybx1 expression was conducted to elucidate transcriptomic events controlled by YBX1 during adipogenesis. RESULTS: Our proteomics analysis uncovered 509 proteins differentially regulated by cold in a time-dependent manner. Overall, 44 transcriptional regulators were acutely upregulated following cold exposure, among which included the cold-shock domain containing protein YBX1, peaking after 24 h. Cold-induced upregulation of YBX1 also occurred in brown adipose tissue, but not in visceral white adipose tissue, suggesting a role of YBX1 in thermogenesis. This role was confirmed by Ybx1 knockdown in brown and brite preadipocytes, which significantly impaired their thermogenic potential. Conversely, inducing Ybx1 expression in mesenchymal stem cells during adipogenesis promoted browning concurrent with an increased expression of thermogenic markers and enhanced mitochondrial respiration. At a molecular level, our transcriptomic analysis showed that YBX1 regulates a subset of genes, including the histone H3K9 demethylase Jmjd1c, to promote thermogenic adipocyte differentiation. CONCLUSION: Our study mapped the dynamic proteomic changes of murine scWAT during browning and identified YBX1 as a novel factor coordinating the genomic mechanisms by which preadipocytes commit to brite/beige lineage.


Subject(s)
Adipose Tissue, White/metabolism , Thermogenesis/genetics , Thermogenesis/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Adipocytes, Brown/metabolism , Adipogenesis , Adipose Tissue, Brown/metabolism , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Gene Expression Regulation , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Male , Mesenchymal Stem Cells , Mice , Mice, Inbred C57BL , Obesity/metabolism , Proteomics , Subcutaneous Fat/metabolism , Transcriptome , Up-Regulation
7.
Front Endocrinol (Lausanne) ; 11: 579785, 2020.
Article in English | MEDLINE | ID: mdl-33101212

ABSTRACT

Activation of thermogenic adipose tissue is linked to improved metabolic outcomes in mice and humans. Dissipation of energy as heat during thermogenesis relies on sufficient innervation of fat by sympathetic nerve fibers, a process recently proposed to be regulated by the adipose-specific calsyntenin3ß (Clstn3ß)-S100b axis. Here we aimed 1) to assess enrichment patterns of CLSTN3ß, S100b as well as the previously annotated neuronal CLSTN3α in perirenal brown and subcutaneous white human fat specimens, and 2) to investigate if the novel Clstn3ß is dynamically regulated by changes in environmental temperatures and nutritional stress in thermogenic adipose tissues in mice. We provide evidence for CLSTN3ß enrichment in multilocular perirenal fat located anatomically in the proximity to both the adrenal gland and sympathetic nerve bundles innervating the kidney in humans. Moreover, transcript levels of CLSTN3ß, but not S100b or CLSTN3α, positively correlate with uncoupling protein 1 (UCP1) expression in human adipose tissue. Our results further show that Clsnt3ß is preferentially expressed in brown adipocytes and is highly responsive to changes in environmental temperature and obesity state in mice. Collectively, this brief communication highlights CLSTN3ß as a hallmark of thermogenic adipose depots in mice and humans.


Subject(s)
Adipose Tissue, Brown/pathology , Calcium-Binding Proteins/metabolism , Diet, High-Fat/adverse effects , Heat-Shock Response , Membrane Proteins/metabolism , Obesity/physiopathology , Thermogenesis , Adipose Tissue, Brown/metabolism , Adult , Aged , Animals , Calcium-Binding Proteins/genetics , Female , Gene Expression Regulation , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Middle Aged , Subcutaneous Fat
8.
Adipocyte ; 9(1): 142-152, 2020 12.
Article in English | MEDLINE | ID: mdl-32249683

ABSTRACT

Insulin action initiates a series of phosphorylation events regulating cellular differentiation, growth and metabolism. We have previously discovered, in a mass spectrometry-based phosphoproteomic study, that insulin/IGF-1 signalling induces phosphorylation of retinoid x receptor alpha (RXRα) at S22 in mouse brown pre-adipocytes. Here, we show that insulin induces the phosphorylation of RXRα at S22 in both brown precursor and mature adipocytes through a pathway involving ERK, downstream of IRS-1 and -2. We also found that RXRα S22 phosphorylation is promoted by insulin and upon re-feeding in brown adipose tissue in vivo, and that insulin-stimulated S22 phosphorylation of RXRα is dampened by diet-induced obesity. We used Rxra knockout cells re-expressing wild type (WT) or S22A non-phosphorylatable forms of RXRα to further characterize the role of S22 in brown adipocytes. Knockout of Rxra in brown pre-adipocytes resulted in decreased lipid accumulation and adipogenic gene expression during differentiation, and re-expression of RxraWT alleviated these effects. However, we observed no significant difference in cells re-expressing the RxraS22A mutant as compared with the cells re-expressing RxraWT. Furthermore, comparison of gene expression during adipogenesis in the WT and S22A re-expressing cells by RNA sequencing revealed similar transcriptomic profiles. Thus, our data propose a dispensable role for RXRα S22 phosphorylation in adipogenesis and transcription in differentiating brown pre-adipocytes.


Subject(s)
Adipocytes, Brown/metabolism , Adipogenesis , Insulin/metabolism , Retinoid X Receptor alpha/metabolism , Serine/metabolism , Adipocytes, Brown/cytology , Animals , Cell Differentiation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation
9.
EMBO Rep ; 20(8): e48216, 2019 08.
Article in English | MEDLINE | ID: mdl-31264358

ABSTRACT

Insulin orchestrates metabolic homeostasis through a complex signaling network for which the precise mechanisms controlling its fine-tuning are not completely understood. Here, we report that Afadin, a scaffold protein, is phosphorylated on S1795 (S1718 in humans) in response to insulin in adipocytes, and this phosphorylation is impaired with obesity and insulin resistance. In turn, loss of Afadin enhances the response to insulin in adipose tissues via upregulation of the insulin receptor protein levels. This happens in a cell-autonomous and phosphorylation-dependent manner. Insulin-stimulated Afadin-S1795 phosphorylation modulates Afadin binding with interaction partners in adipocytes, among which HDAC6 preferentially interacts with phosphorylated Afadin and acts as a key intermediate to suppress insulin receptor protein levels. Adipose tissue-specific Afadin depletion protects against insulin resistance and improves glucose homeostasis in diet-induced obese mice, independently of adiposity. Altogether, we uncover a novel insulin-induced cellular feedback mechanism governed by the interaction of Afadin with HDAC6 to negatively control insulin action in adipocytes, which may offer new strategies to alleviate insulin resistance.


Subject(s)
Adipose Tissue/metabolism , Antigens, CD/genetics , Histone Deacetylase 6/genetics , Insulin/genetics , Microfilament Proteins/genetics , Obesity/genetics , Protein Processing, Post-Translational , Receptor, Insulin/genetics , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Antigens, CD/metabolism , Diet, High-Fat/adverse effects , Glucose/metabolism , Histone Deacetylase 6/metabolism , Homeostasis/genetics , Humans , Insulin/metabolism , Insulin/pharmacology , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Phosphorylation , Primary Cell Culture , Receptor, Insulin/metabolism
10.
An. Fac. Cienc. Méd. (Asunción) ; 52(2): 17-22, 20190700.
Article in Spanish | LILACS-Express | LILACS | ID: biblio-1006994

ABSTRACT

Introducción: El uso inapropiado de los antimicrobianos está conduciendo a la disminución de la sensibilidad antimicrobiana de microorganismos que producen enfermedades infecciosas. El uso clínico de los antimicrobianos ha sido paralelo al surgimiento de bacterias resistentes a su acción. Materiales y Métodos: Se realizó un estudio observacional, descriptivo, retrospectivo de corte transversal, con muestreo no probabilístico de casos consecutivos. La selección de antibióticos incluyó solo aquellos que tienen actividad frente a bacilos gram negativos y para la interpretación de los diámetros de los halos de inhibición se tomaron en cuenta los estándares de la Clinical and Laboratory Standards Institute (CLSI versión 2015). Resultados: La ampicilina presentó una resistencia del 100%, en cambio la ciprofloxacina presentó una sensibilidad del 73%, la amikacina del 100% y la gentamicina del 82%. La familia de los carbapenemicos presentó una sensibilidad del 100%. Dentro del grupo de los nitrofuranos se constató una sensibilidad del 96%. El trimetoprim/ sulfametoxazol, presentó una sensibilidad del 57%. Se determinó la presencia de Beta Lactamasa de Espectro Extendido (BLEE) en 13% de las cepas de Escherichia coli estudiadas. Conclusiones: La gentamicina, la nitrofurantoína y los carbapenemes presentaron una elevada sensibilidad, por lo que en presencia de cepas con BLEE serían opciones factibles acompañados siempre de la evaluación del perfil renal y clasificando la ITU como complicada o no.


Introduction: The inappropriate use of antimicrobials is leading to a decrease in the antimicrobial sensitivity of microorganisms that cause infectious diseases. The clinical use of antimicrobials has been parallel to the emergence of bacteria resistant to its action. Materials and methods: The sensitivity profile of Escherichia coli isolated from patients with urinary tract infections (UTI) concurrent to the Regional Hospital of Villarrica in the period from 2013 to 2015 was determined. Results: Ampicillin showed a resistance of 100%, ciprofloxacin showed a sensitivity of 73%. Amikacin showed a sensitivity of 100% and gentamicin 82%. The family of carbapenem showed a sensitivity of 100%. Within the group of nitrofurans a sensitivity of 96% was found. Trimethoprim / sulfamethoxazole had a sensitivity of 57%. The presence of Extended Spectrum Beta Lactamase (ESBL) was determined in 13% of the strains of Escherichia coli studied. Conclusions: Gentamicin, nitrofurantoin and carbapenem have a high sensitivity, so in the presence of strains with ESBL they would be feasible options always accompanied by the evaluation of the renal profile and classifying the UTI as complicated or not.

11.
Mol Metab ; 6(10): 1313-1320, 2017 10.
Article in English | MEDLINE | ID: mdl-29031730

ABSTRACT

OBJECTIVE: Functional investigation of novel gene/protein targets associated with adipocyte differentiation or function heavily relies on efficient and accessible tools to manipulate gene expression in adipocytes in vitro. Recent advances in gene-editing technologies such as CRISPR-Cas9 have not only eased gene editing but also greatly facilitated modulation of gene expression without altering the genome. Here, we aimed to develop and validate a competent in vitro adipocyte model of controllable functionality as well as multiplexed gene manipulation in adipocytes, using the CRISPRa "SAM" system and siRNAs to simultaneously overexpress and silence selected genes in the same cell populations. METHODS: We introduced a stable expression of dCas9-VP64 and MS2-P65, the core components of the CRIPSRa SAM system, in mesenchymal C3H/10T1/2 cells through viral delivery and used guide RNAs targeting Pparγ2, Prdm16, Zfp423, or Ucp1 to control the expression of key genes involved in adipocyte differentiation and function. We additionally co-transfected mature adipocytes with sgRNA plasmids and siRNA to simultaneously up-regulate and silence selected genes. Quantitative gene expression, oxygen consumption, fluorescence-activated cell sorting and immunocytochemistry served as validation proxies in pre- or mature adipocytes. RESULTS: CRISPRa SAM-mediated up-regulation of a key adipogenic gene, Pparγ2, was successfully achieved using selected sgRNAs targeting the Pparγ2 promoter region (i.e. up to 104 fold); this induction was long lasting and sufficient to promote adipogenesis. Furthermore, co-activation of Pparγ2 with either Prdm16 or Zfp423 transcripts drove distinct thermogenic gene expression patterns associated with increased or decreased oxygen consumption, respectively, mimicking typical characteristics of brite/beige or white cell lineages. Lastly, we demonstrated that up-regulation of endogenous genes in mature adipocytes was also easily and efficiently achieved using CRISPRa SAM, here exemplified by targeted Ucp1 overexpression (up to 4 × 103 fold), and that it was compatible with concomitant gene silencing using siRNA, allowing for bidirectional manipulation of gene expression in the same cell populations. CONCLUSIONS: We demonstrate that the CRISPRa SAM system can be easily adopted and used to efficiently manipulate gene expression in pre- and mature adipocytes in vitro. Moreover, we describe a novel methodological approach combining the activation of endogenous genes and siRNA-mediated gene silencing, thus providing a powerful tool to functionally decipher genetic factors controlling adipogenesis and adipocyte functions.


Subject(s)
Adipogenesis/genetics , CRISPR-Cas Systems/genetics , Genetic Engineering/methods , Adipocytes/cytology , Adipocytes/metabolism , Adipocytes, Brown/metabolism , Adipocytes, White/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Expression/genetics , Gene Expression Regulation/genetics , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Thermogenesis , Uncoupling Protein 1/metabolism
12.
Child Adolesc Ment Health ; 19(1): 2-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-32878365

ABSTRACT

BACKGROUND: This article proposes a conceptual model of child and parent engagement in the mental health intervention process. METHOD: A scoping review was performed of articles on predictors of engagement in mental health interventions, the effectiveness of engagement interventions, and interpersonal aspects of care. A comprehensive search of PsycINFO and PsycARTICLES was performed for literature published in English from 2000 to 2012. RESULTS: Based on the review, a motivational framework is proposed in which engagement is defined as a state comprised of a hopeful stance, conviction, and confidence, brought about when therapists optimize engagement processes of receptiveness, willingness, and self-efficacy. CONCLUSIONS: Implications concern the need to help clients understand what to expect from the therapy process, and to educate therapists about engagement strategies.

SELECTION OF CITATIONS
SEARCH DETAIL
...