Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Acta Psychiatr Scand ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38958035

ABSTRACT

OBJECTIVE: This systematic review aimed to summarise and synthesise research conducted in psychiatric mother-baby units (MBUs) in relation to patients, their families, or staff, published from 1st January 2016 to 1st May 2024. METHOD: Quantitative, qualitative, and mixed-method studies were included for review if they were published in peer-review journals in English and reported research on MBUs between January 2016 and May 2024. From the initial yield of 10,007 unique studies, 53 studies were included for review. RESULTS: MBU research was found to more frequently investigate maternal characteristics rather than the benefits of MBU treatment compared to studies conducted prior to 2016. Most studies that did investigate impact of admission showed favourable results, however few follow-up studies and studies comparing MBU outcomes to other clinical settings were undertaken. Little research has been conducted to investigate the differential impacts of MBU admission on different diagnoses and long-term (>1 year) patient outcomes. There was a dearth of research investigating partners of women in MBUs and few studies conducted on infant outcomes. CONCLUSIONS: MBUs were consistently found to improve mental health systems and mother-infant attachment in patients after admission. More research investigating patient support networks and child health, impact of diagnosis on outcomes, and studies with adequate follow-up are required.

2.
Trials ; 24(1): 747, 2023 Nov 23.
Article in English | MEDLINE | ID: mdl-37996896

ABSTRACT

BACKGROUND: Perinatal women are highly vulnerable to developing mental health issues and particularly susceptible to a recurrence of psychiatric illness. Poor mental health during the perinatal period can have long-term impacts on the physical and psychiatric health of both mother and child. A potentially useful strategy to improve women's mental health is through a mobile application teaching mindfulness, an evidence-based technique helping individuals focus on the present moment. METHODS: A mixed method, prospective randomised controlled trial. The study group comprise women aged 18 years and over, who are attending the public and private maternity clinics at Mater Mothers' Hospital. A sample of 360 prenatal women will be randomised into the intervention group (with the use of the mindfulness app) or usual care. Participants will remain in the study for 11 months and will be assessed at four timepoints for changes in postnatal depression, mother-infant bonding, and quality of life. A cost-effectiveness evaluation will also be conducted using quality-adjusted life year (QALY) calculations. A random selection of intervention participants will be invited to attend focus groups to give feedback on the mindfulness app. DISCUSSION: Previous studies have found mindfulness interventions can reduce stress, anxiety, depression, and sleep disturbances in a prenatal population. The risks of the intervention are low, but could be of significant benefit for women who are unable to attend face-to-face appointments due to geographical, financial, or time barriers; during endemic or pandemic scenarios; or due to health or mobility issues. TRIAL REGISTRATION: This study was approved by the Mater Misericordiae Human Research Ethics Committee (83,589). Australian New Zealand Clinical Trials Registry (ANZCTR) ACTRN12622001581752 ( https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=385107&isReview=true ). Registered on 22 Dec. 2022.


Subject(s)
Mental Health , Mindfulness , Adolescent , Adult , Female , Humans , Pregnancy , Australia , Cost-Benefit Analysis , Peripartum Period , Prospective Studies , Quality of Life , Randomized Controlled Trials as Topic , Infant, Newborn
3.
Immunol Cell Biol ; 101(7): 639-656, 2023 08.
Article in English | MEDLINE | ID: mdl-37191045

ABSTRACT

Graft-versus-host disease (GVHD) is a life-threatening complication following donor hematopoietic stem cell transplantation, where donor T cells damage host tissues. This study investigated the effect of tocilizumab (TOC) combined with post-transplant cyclophosphamide (PTCy) on immune cell engraftment and GVHD development in a humanized mouse model. NOD-scid-IL2Rγnull (NSG) mice were injected intraperitoneally with 2 × 107 human (h) peripheral blood mononuclear cells and cyclophosphamide (33 mg kg-1 ) or saline on days 3 and 4, then TOC or control antibody (0.5 mg mouse-1 ) twice weekly for 28 days. Mice were monitored for clinical signs of GVHD for either 28 or 70 days. Spleens and livers were assessed for human leukocyte subsets, and serum cytokines and tissue histology were analyzed. In the short-term model (day 28), liver and lung damage were reduced in PTCy + TOC compared with control mice. All groups showed similar splenic hCD45+ leukocyte engraftment (55-60%); however, PTCy + TOC mice demonstrated significantly increased (1.5-2-fold) splenic regulatory T cells. Serum human interferon gamma was significantly reduced in PTCy + TOC compared with control mice. Long-term (day 70), prolonged survival was similar in PTCy + TOC (median survival time, > 70 days) and PTCy mice (median survival time, 56 days). GVHD onset was significantly delayed in PTCy + TOC, compared with TOC or control mice. Notably, natural killer cells were reduced (77.5%) in TOC and PTCy + TOC mice. Overall, combining PTCy with TOC increases regulatory T cells and reduces clinical signs of early GVHD, but does not improve long-term survival compared with PTCy alone.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Humans , Animals , Mice , T-Lymphocytes, Regulatory/pathology , Leukocytes, Mononuclear , Mice, Inbred NOD , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Killer Cells, Natural/pathology , Mice, SCID
4.
J Psychiatr Res ; 156: 611-627, 2022 12.
Article in English | MEDLINE | ID: mdl-36372004

ABSTRACT

Vascular cognitive impairment (VCI) and depression frequently coexist in geriatric populations and reciprocally increase disease risks. We assert that a shared pre-disease state of the psycho-immune-neuroendocrine (PINE) network model mechanistically explains bidirectional associations between VCI and depression. Five pathophysiological sub-networks are identified that are shared by VCI and depression: neuroinflammation, kynurenine pathway imbalance, hypothalamic-pituitary-adrenal (HPA) axis overactivity, impaired neurotrophic support and cerebrovascular dysfunction. These do not act independently, and their complex interactions necessitate a systems biology approach to better define disease pathogenesis. The PINE network is already established in the context of non-communicable diseases (NCDs) such as depression, hypertension, atherosclerosis, coronary heart disease and type 2 diabetes mellitus. We build on previous literature to specifically explore mechanistic links between MDD and VCI in the context of PINE pathways and discuss key mechanistic commonalities linking these comorbid conditions and identify a common pre-disease state which precedes transition to VCI and MDD. We expand the model to incorporate bidirectional interactions with biological ageing. Diathesis factors for both VCI and depression feed into this network and the culmination of shared mechanisms (on an ageing substrate) lead to a critical network transition to one or both disease states. A common pre-disease state underlying VCI and depression can provide clinicians a unique opportunity for early risk assessment and intervention in disease development. Establishing the mechanistic elements and systems biology of this network can reveal early warning or predictive biomarkers together with novel therapeutic targets. Integrative studies are recommended to elucidate the dynamic networked biology of VCI and depression over time.


Subject(s)
Cognitive Dysfunction , Diabetes Mellitus, Type 2 , Humans , Aged , Cognitive Dysfunction/etiology
5.
Article in English | MEDLINE | ID: mdl-36293807

ABSTRACT

Both psychosocial and physical environmental stressors have been linked to chronic mental health and chronic medical conditions. The psycho-immune-neuroendocrine (PINE) network details metabolomic pathways which are responsive to varied stressors and link chronic medical conditions with mental disorders, such as major depressive disorder via a network of pathophysiological pathways. The primary objective of this review is to explore evidence of relationships between airborne particulate matter (PM, as a concrete example of a physical environmental stressor), the PINE network and chronic non-communicable diseases (NCDs), including mental health sequelae, with a view to supporting the assertion that physical environmental stressors (not only psychosocial stressors) disrupt the PINE network, leading to NCDs. Biological links have been established between PM exposure, key sub-networks of the PINE model and mental health sequelae, suggesting that in theory, long-term mental health impacts of PM exposure may exist, driven by the disruption of these biological networks. This disruption could trans-generationally influence health; however, long-term studies and information on chronic outcomes following acute exposure event are still lacking, limiting what is currently known beyond the acute exposure and all-cause mortality. More empirical evidence is needed, especially to link long-term mental health sequelae to PM exposure, arising from PINE pathophysiology. Relationships between physical and psychosocial stressors, and especially the concept of such stressors acting together to impact on PINE network function, leading to linked NCDs, evokes the concept of syndemics, and these are discussed in the context of the PINE network.


Subject(s)
Depressive Disorder, Major , Mental Health , Humans , Depressive Disorder, Major/psychology , Neurosecretory Systems , Syndemic , Chronic Disease , Disease Progression , Particulate Matter
6.
J Neuroimmunol ; 372: 577959, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36095861

ABSTRACT

BACKGROUND/AIMS: The psycho-immune-neuroendocrine (PINE) network is a predominantly physiological (metabolomic) model constructed from the literature, inter-linking multiple biological processes associated with major depressive disorder (MDD), thereby integrating putative mechanistic pathways for MDD into a single network. MATERIAL AND METHODS: Previously published metabolomic pathways for the PINE network based on literature searches conducted in 1991-2021 were used to construct an edge table summarizing all physiological pathways in pairs of origin nodes and target nodes. The Gephi software program was used to calculate network metrics from the edge table, including total degree and centrality measures, to ascertain key network nodes and construct a directed network graph. RESULTS: An edge table and directional network graph of physiological relationships in the PINE network is presented. The network has properties consistent with complex biological systems, with analysis yielding key network nodes comprising pro-inflammatory cytokines (TNF- α, IL6 and IL1), glucocorticoids and corticotropin releasing hormone (CRH). These may represent central structural and regulatory elements in the context of MDD. CONCLUSION: The identified hubs have a high degree of connection and are known to play roles in the progression from health to MDD. These nodes represent strategic targets for therapeutic intervention or prevention. Future work is required to build a weighted and dynamic simulation of the network PINE.


Subject(s)
Depressive Disorder, Major , Corticotropin-Releasing Hormone , Depressive Disorder, Major/drug therapy , Glucocorticoids/therapeutic use , Humans , Interleukin-6 , Patient-Specific Modeling , Systems Biology
7.
Methods Mol Biol ; 2510: 315-340, 2022.
Article in English | MEDLINE | ID: mdl-35776334

ABSTRACT

Humanized mouse models of graft-versus-host disease (GVHD), where human immune cells are injected into immune deficient mice, are well established and provide opportunities to investigate pathways involved in GVHD development. This chapter provides an overview of human immune cell isolation, injection of these cells into immune deficient mice, monitoring of mice for signs of GVHD, and assessment of human cell engraftment using flow cytometry. Further, this chapter focuses on the P2X7 signaling pathway involved in GVHD, and describes a strategy to block the P2X7 receptor and examine the effect of this on GVHD development.


Subject(s)
Graft vs Host Disease , Receptors, Purinergic P2X7 , Animals , Cell Separation , Disease Models, Animal , Flow Cytometry , Graft vs Host Disease/etiology , Mice , Receptors, Purinergic P2X7/genetics
8.
Int J Mol Sci ; 22(15)2021 Aug 03.
Article in English | MEDLINE | ID: mdl-34361109

ABSTRACT

Allogeneic haematopoietic stem cell transplantation (allo-HSCT) is a curative therapy for blood cancers and other haematological disorders. However, allo-HSCT leads to graft-versus-host disease (GVHD), a severe and often lethal immunological response, in the majority of transplant recipients. Current therapies for GVHD are limited and often reduce the effectiveness of allo-HSCT. Therefore, pro- and anti-inflammatory factors contributing to disease need to be explored in order to identify new treatment targets. Purinergic signalling plays important roles in haematopoiesis, inflammation and immunity, and recent evidence suggests that it can also affect haematopoietic stem cell transplantation and GVHD development. This review provides a detailed assessment of the emerging roles of purinergic receptors, most notably P2X7, P2Y2 and A2A receptors, and ectoenzymes, CD39 and CD73, in GVHD.


Subject(s)
Graft vs Host Disease/therapy , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/methods , Receptors, Purinergic/metabolism , Signal Transduction , Animals , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Transplantation, Homologous
9.
Immunology ; 164(2): 332-347, 2021 10.
Article in English | MEDLINE | ID: mdl-34021907

ABSTRACT

Graft-versus-host disease (GVHD) is a major complication of allogeneic haematopoietic stem cell transplantation (allo-HSCT) that develops when donor T cells in the graft become reactive against the host. Post-transplant cyclophosphamide (PTCy) is increasingly used in mismatched allo-HSCT, but how PTCy impacts donor T cells and reduces GVHD is unclear. This study aimed to determine the effect of PTCy on reactive human donor T cells and GVHD development in a preclinical humanized mouse model. Immunodeficient NOD-scid-IL2Rγnull mice were injected intraperitoneally (i.p.) with 20 × 106 human peripheral blood mononuclear cells stained with carboxyfluorescein succinimidyl ester (CFSE) (day 0). Mice were subsequently injected (i.p.) with PTCy (33 mg kg-1 ) (PTCy-mice) or saline (saline-mice) (days 3 and 4). Mice were assessed for T-cell depletion on day 6 and monitored for GVHD for up to 10 weeks. Flow cytometric analysis of livers at day 6 revealed lower proportions of reactive (CFSElow ) human (h) CD3+ T cells in PTCy-mice compared with saline-mice. Over 10 weeks, PTCy-mice showed reduced weight loss and clinical GVHD, with prolonged survival and reduced histological liver GVHD compared with saline-mice. PTCy-mice also demonstrated increased splenic hCD4+ :hCD8+ T-cell ratios and reduced splenic Tregs (hCD4+  hCD25+  hCD127lo ) compared with saline-mice. This study demonstrates that PTCy reduces GVHD in a preclinical humanized mouse model. This corresponded to depletion of reactive human donor T cells, but fewer human Tregs.


Subject(s)
Cyclophosphamide/immunology , Graft vs Host Disease/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Disease Models, Animal , Female , Hematopoietic Stem Cell Transplantation/methods , Humans , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Tissue Donors , Transplantation, Homologous/methods
10.
Clin Sci (Lond) ; 135(3): 495-513, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33463682

ABSTRACT

Graft-versus-host disease (GVHD) is a severe inflammatory response arising from allogeneic haematopoietic stem cell transplantation. Previous studies revealed that antagonism of the P2X7 receptor with Brilliant Blue G (BBG) reduced liver GVHD but did not alter clinical GVHD in a humanised mouse model. Therefore, the present study aimed to trial a modified injection regime using more frequent dosing of BBG to improve outcomes in this model of GVHD. NOD-scid IL2Rγnull (NSG) mice were injected intraperitoneally (i.p.) with 10 × 106 human peripheral blood mononuclear cells (hPBMCs) (day 0), then daily with BBG (50 mg/kg) or saline (days 0-10). BBG significantly reduced clinical score, mortality and histological GVHD compared with saline treatment (endpoint). BBG significantly increased proportions of human regulatory T cells (Tregs) and human B cells and reduced serum human interferon-γ compared with saline treatment prior to development of clinical GVHD (day 21). To confirm the therapeutic benefit of P2X7 antagonism, NSG mice were injected i.p. with 10 × 106 hPBMCs (day 0), then daily with pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS) (300 mg/kg) or saline (days 0-10). PPADS increased human Treg proportions compared with saline treatment (day 21), but potential clinical benefits were confounded by increased weight loss with this antagonist. To investigate the role of P2X7 antagonism on Treg survival, hPBMCs were cultured in reduced serum conditions to promote cell death. BBG increased proportions of Tregs (and B cells) compared with saline under these conditions. In conclusion, P2X7 antagonism reduces clinical and histological GVHD in a humanised mouse model corresponding to an increase in human Tregs.


Subject(s)
Graft vs Host Disease/drug therapy , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7/drug effects , Rosaniline Dyes/pharmacology , Adult , Animals , B-Lymphocytes , Disease Models, Animal , Female , Graft vs Host Disease/pathology , Hematopoietic Stem Cell Transplantation , Humans , Leukocytes, Mononuclear , Male , Mice, Inbred NOD , Mice, SCID , Purinergic P2X Receptor Antagonists/administration & dosage , Pyridoxal Phosphate/administration & dosage , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , Rosaniline Dyes/administration & dosage , T-Lymphocytes, Regulatory/drug effects
11.
Immunol Cell Biol ; 98(5): 397-410, 2020 05.
Article in English | MEDLINE | ID: mdl-32181525

ABSTRACT

Regulatory T cells (Tregs) protect against graft-versus-host disease (GVHD), a life-threatening complication of allogeneic hematopoietic stem cell transplantation. The ectoenzyme CD39 is important for increasing the immunosuppressive function of Tregs. The rs10748643 (A â†’ G) single-nucleotide polymorphism (SNP) in intron 1 of the human ENTPD1 gene is associated with increased proportions of CD39+ Tregs. This study aimed to determine whether the rs10748643 SNP corresponded to increased proportions of CD39+ Tregs in an Australian donor population, and whether this SNP influences clinical GVHD in a humanized mouse model. Donors were genotyped for the rs10748643 SNP by Sanger sequencing, and the proportion of CD39+ T cells in donor peripheral blood was determined by flow cytometry. Donors encoding the G allele (donorsAG/GG ) demonstrated higher proportions of CD39+ CD3+ CD4+ CD25+ CD127lo Tregs, but not CD39+ CD3+ CD8+ T cells or CD39+ CD3+ CD4+ conventional T cells, compared with donors homozygous for the A allele (donorsAA ). NOD-SCID-IL2Rγnull mice were injected with human peripheral blood mononuclear cells from either donorsAA (hCD39AA mice) or donorsAG/GG (hCD39AG/GG mice). hCD39AG/GG mice demonstrated significantly greater weight loss and GVHD clinical scores, and significantly reduced survival, compared with hCD39AA mice. hCD39AG/GG mice showed significantly higher hCD4+ :hCD8+ T-cell ratios than hCD39AA mice, but displayed similar proportions of CD3+ hCD4+ hCD25+ hCD127lo Tregs and hCD39+ Tregs. However, the proportion of human Tregs corresponded to survival in hCD39AA mice, but not in hCD39AG/GG mice. This study demonstrates that donors encoding the G allele show higher percentages of CD39+ Tregs, but cause worsened GVHD in humanized mice compared with donors homozygous for the A allele.


Subject(s)
Apyrase/genetics , Graft vs Host Disease , T-Lymphocytes, Regulatory/immunology , Animals , Australia , Humans , Leukocytes, Mononuclear , Mice , Mice, Inbred NOD , Mice, SCID , Polymorphism, Single Nucleotide
12.
Clin Sci (Lond) ; 134(2): 207-223, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31934722

ABSTRACT

BACKGROUND: Allogeneic haematopoietic stem cell transplantation (HSCT) is a curative therapy for blood cancers; but results in the development of graft-versus-host disease (GVHD) in up to 70% of recipients. During GVHD, tissue damage results in ATP release into the extracellular compartment activating P2X7 on antigen-presenting cells, leading to the release of pro-inflammatory cytokines and subsequent activation of donor T cells. Therefore, the aim of the present study was to examine murine (m) P2rx7 and human (h) P2RX7 gene expression in GVHD target organs of humanised mice, and further characterise disease impact in these organs. METHODS: NOD-scid IL2Rγnull (NSG) mice were injected with human peripheral blood mononuclear cells (hu-PBMC-NSG mice) or phosphate-buffered saline (PBS, control). Leucocytes were assessed by flow cytometry; gene expression was measured by quantitative polymerase chain reaction (qPCR), and tissue sections examined by histology. RESULTS: Compared with control mice, hu-PBMC-NSG mice had increased mP2rx7 and mP2rx4 expression in the duodenum, ileum and skin. hP2RX7 was expressed in all tissues examined. hu-PBMC-NSG mice also displayed increased mReg3g expression in the duodenum and ileum, despite limited histological gut GVHD. hu-PBMC-NSG mice showed histological evidence of GVHD in the skin, liver and lung. Compared with control mice, hu-PBMC-NSG mice displayed increased ear swelling. CONCLUSION: Combined data revealed that P2rx7 is up-regulated in gut and skin GVHD and that P2RX7 is present in target tissues of GVHD, corresponding to human leucocyte infiltration. Data also reveal increased mReg3g expression and ear swelling in hu-PBMC-NSG mice, offering new measurements of early-stage gut GVHD and skin GVHD, respectively.


Subject(s)
Gastrointestinal Tract/metabolism , Graft vs Host Disease/metabolism , Receptors, Purinergic P2X7/metabolism , Skin/metabolism , Adult , Animals , Antigens, CD/metabolism , Disease Models, Animal , Ear/pathology , Female , Graft vs Host Disease/blood , Humans , Interferon-gamma/blood , Leukocytes, Mononuclear/metabolism , Liver/pathology , Lung/pathology , Male , Mice , Pancreatitis-Associated Proteins/metabolism , Spleen/immunology , T-Lymphocytes/immunology , Up-Regulation/genetics , Young Adult
13.
Transpl Immunol ; 54: 38-46, 2019 06.
Article in English | MEDLINE | ID: mdl-30743002

ABSTRACT

Graft-versus-host disease (GVHD) is a frequent complication following allogeneic hematopoietic stem cell transplantation (HSCT) with current therapies limited to general immunosuppression. Humanized mouse models of GVHD are emerging as valuable intermediaries to allow translation of findings from allogeneic mouse models to humans to prevent and treat this disease, but such models require further characterization. In this study, humanized mice were generated by injecting immunodeficient non-obese diabetic severe combined immunodeficiency interleukin (IL)-2 receptor γ common chain null (NSG) mice with human peripheral blood mononuclear cells (hPBMCs). Clinical GVHD development was assessed using established scoring criteria (weight loss, posture, activity, fur texture and skin integrity). Differences between humanized NSG mice that developed clinical or subclinical GVHD were then compared. Both groups of mice demonstrated similar frequencies of human leukocyte engraftment. In contrast, mice that developed clinical GVHD demonstrated increased histological damage compared to mice with subclinical GVHD. Furthermore, mice with clinical GVHD exhibited increases in the splenic human CD4+:CD8+ T cell ratio, serum human interferon (IFN)-γ and intestinal human IL-17 expression compared to mice with subclinical GVHD. These cellular and molecular changes could be used as potential markers of disease progression in this preclinical model. This study also provides further insights into GVHD development which may be relevant to human HSCT recipients.


Subject(s)
Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Interferon-gamma/blood , Interleukin-17/metabolism , Spleen/immunology , T-Lymphocytes/immunology , Animals , Biomarkers , CD4-CD8 Ratio , Disease Models, Animal , Disease Progression , Graft vs Host Disease/diagnosis , Humans , Mice , Mice, Inbred NOD , Mice, SCID
SELECTION OF CITATIONS
SEARCH DETAIL
...