Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Stefanie Malan-Müller is active.

Publication


Featured researches published by Stefanie Malan-Müller.


Current Environmental Health Reports | 2016

The Microbiota, Immunoregulation, and Mental Health: Implications for Public Health

Christopher A. Lowry; David G. Smith; Philip H. Siebler; Dominic Schmidt; Christopher E. Stamper; James E. Hassell; Paula Shimene de Melo Yamashita; James H. Fox; Stefan O. Reber; Lisa A. Brenner; Andrew J. Hoisington; Teodor T. Postolache; Kerry A. Kinney; Dante Marciani; Mark Hernandez; Sian Hemmings; Stefanie Malan-Müller; Kenneth P. Wright; Rob Knight; Charles L. Raison; G. A. W. Rook

The hygiene or “Old Friends” hypothesis proposes that the epidemic of inflammatory disease in modern urban societies stems at least in part from reduced exposure to microbes that normally prime mammalian immunoregulatory circuits and suppress inappropriate inflammation. Such diseases include but are not limited to allergies and asthma; we and others have proposed that the markedly reduced exposure to these Old Friends in modern urban societies may also increase vulnerability to neurodevelopmental disorders and stress-related psychiatric disorders, such as anxiety and affective disorders, where data are emerging in support of inflammation as a risk factor. Here, we review recent advances in our understanding of the potential for Old Friends, including environmental microbial inputs, to modify risk for inflammatory disease, with a focus on neurodevelopmental and psychiatric conditions. We highlight potential mechanisms, involving bacterially derived metabolites, bacterial antigens, and helminthic antigens, through which these inputs promote immunoregulation. Though findings are encouraging, significant human subjects’ research is required to evaluate the potential impact of Old Friends, including environmental microbial inputs, on biological signatures and clinically meaningful mental health prevention and intervention outcomes.


Genes, Brain and Behavior | 2014

Understanding posttraumatic stress disorder: insights from the methylome

Stefanie Malan-Müller; Soraya Seedat; Sian Hemmings

Genome‐wide association studies (GWAS) have identified numerous disease‐associated variants; however, these variants have a minor effect on disease and explain only a small amount of the heritability of complex disorders. The search for the missing heritability has shifted attention to rare variants, copy number variants, copy neutral variants and epigenetic modifications. The central role of epigenetics, and specifically DNA methylation, in disease susceptibility and progression has become more apparent in recent years. Epigenetic mechanisms facilitate the response to environmental changes and challenges by regulating gene expression. This makes the study of DNA methylation in psychiatric disorders such as posttraumatic stress disorder (PTSD) highly salient, as the environment plays such a vital role in disease aetiology. The epigenome is dynamic and can be modulated by numerous factors, including learning and memory, which are important in the context of PTSD. Numerous studies have shown the effects of early life events, such as maternal separation and traumas during adulthood, on DNA methylation patterns and subsequent gene expression profiles. Aberrations in adaptive DNA methylation contribute to disease susceptibility when an organism is unable to effectively respond to environmental demands. Epigenetic mechanisms are also involved in higher order brain functions. Dysregulation of methylation is associated with neurodevelopmental and neurodegenerative cognitive disorders, affective disorders, addictive behaviours and altered stress responses. A thorough understanding of how the environment, methylome and transcriptome interact and influence each other in the context of fear and anxiety is integral to our understanding and treatment of stress‐related disorders such as PTSD.


PLOS ONE | 2013

Shorter Telomere Length - A Potential Susceptibility Factor for HIV-Associated Neurocognitive Impairments in South African Woman

Stefanie Malan-Müller; Sı̂an Megan Joanna Hemmings; Georgina Spies; Martin Kidd; Christine Fennema-Notestine; Soraya Seedat

The neuropathogenesis of the human immunodeficiency virus (HIV) may manifest as various neurocognitive impairments (NCI). HIV-positive individuals also have significantly shorter telomere length (TL) in peripheral blood mononuclear cells (PBMCs) and CD8+ T cells compared to HIV-negative individuals. Additionally, reduced TL has been found to be associated with chronic psychological stress. This study focused on the effects of HIV-infection and chronic stress associated with childhood trauma on telomere length, and investigated whether leukocyte TL (LTL), in particular, represents a risk factor for NCI. Eighty-three HIV-positive and 45 HIV-negative women were assessed for childhood trauma and were subjected to detailed neurocognitive testing. Blood from each participant was used to extract Deoxyribonucleic acid (DNA). Relative LTL were determined by performing real time quantitative PCR reactions as described by Cawthon et al. (2002). As expected, relative LTL in the HIV-positive individuals was significantly shorter than that of HIV-negative individuals (F = 51.56, p = <0.01). Notably, a significant positive correlation was evident between relative LTL and learning performance in the HIV-positive group. In addition, a significant negative correlation was observed between relative LTL and verbal fluency, but this association was only evident in HIV-positive individuals who had experienced trauma. Our results suggest that reduced LTL is associated with worse learning performance in HIV-positive individuals, indicating that TL could act as a susceptibility factor in increasing neurocognitive decline in HIV-infected individuals.


Schizophrenia Research | 2016

A systematic review of genetic variants associated with metabolic syndrome in patients with schizophrenia

Stefanie Malan-Müller; Sanja Kilian; Leigh van den Heuvel; Soraya Bardien; Laila Asmal; Louise Warnich; Robin Emsley; Sian Hemmings; Soraya Seedat

Metabolic syndrome (MetS) is a cluster of factors that increases the risk of cardiovascular disease (CVD), one of the leading causes of mortality in patients with schizophrenia. Incidence rates of MetS are significantly higher in patients with schizophrenia compared to the general population. Several factors contribute to this high comorbidity. This systematic review focuses on genetic factors and interrogates data from association studies of genes implicated in the development of MetS in patients with schizophrenia. We aimed to identify variants that potentially contribute to the high comorbidity between these disorders. PubMed, Web of Science and Scopus databases were accessed and a systematic review of published studies was conducted. Several genes showed strong evidence for an association with MetS in patients with schizophrenia, including the fat mass and obesity associated gene (FTO), leptin and leptin receptor genes (LEP, LEPR), methylenetetrahydrofolate reductase (MTHFR) gene and the serotonin receptor 2C gene (HTR2C). Genetic association studies in complex disorders are convoluted by the multifactorial nature of these disorders, further complicating investigations of comorbidity. Recommendations for future studies include assessment of larger samples, inclusion of healthy controls, longitudinal rather than cross-sectional study designs, detailed capturing of data on confounding variables for both disorders and verification of significant findings in other populations. In future, big genomic datasets may allow for the calculation of polygenic risk scores in risk prediction of MetS in patients with schizophrenia. This could ultimately facilitate early, precise, and patient-specific pharmacological and non-pharmacological interventions to minimise CVD associated morbidity and mortality.


Psychosomatic Medicine | 2017

The Microbiome in Posttraumatic Stress Disorder and Trauma-exposed Controls: An Exploratory Study

Sian Hemmings; Stefanie Malan-Müller; Leigh van den Heuvel; Brittany A. Demmitt; Maggie A. Stanislawski; David G. Smith; Adam D. Bohr; Christopher E. Stamper; Embriette R. Hyde; James T. Morton; Clarisse Marotz; Philip H. Siebler; Maarten Braspenning; Wim Van Criekinge; Andrew J. Hoisington; Lisa A. Brenner; Teodor T. Postolache; Matthew B. McQueen; Kenneth S. Krauter; Rob Knight; Soraya Seedat; Christopher A. Lowry

Objective Inadequate immunoregulation and elevated inflammation may be risk factors for posttraumatic stress disorder (PTSD), and microbial inputs are important determinants of immunoregulation; however, the association between the gut microbiota and PTSD is unknown. This study investigated the gut microbiome in a South African sample of PTSD-affected individuals and trauma-exposed (TE) controls to identify potential differences in microbial diversity or microbial community structure. Methods The Clinician-Administered PTSD Scale for DSM-5 was used to diagnose PTSD according to Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition criteria. Microbial DNA was extracted from stool samples obtained from 18 individuals with PTSD and 12 TE control participants. Bacterial 16S ribosomal RNA gene V3/V4 amplicons were generated and sequenced. Microbial community structure, &agr;-diversity, and &bgr;-diversity were analyzed; random forest analysis was used to identify associations between bacterial taxa and PTSD. Results There were no differences between PTSD and TE control groups in &agr;- or &bgr;-diversity measures (e.g., &agr;-diversity: Shannon index, t = 0.386, p = .70; &bgr;-diversity, on the basis of analysis of similarities: Bray-Curtis test statistic = –0.033, p = .70); however, random forest analysis highlighted three phyla as important to distinguish PTSD status: Actinobacteria, Lentisphaerae, and Verrucomicrobia. Decreased total abundance of these taxa was associated with higher Clinician-Administered PTSD Scale scores (r = –0.387, p = .035). Conclusions In this exploratory study, measures of overall microbial diversity were similar among individuals with PTSD and TE controls; however, decreased total abundance of Actinobacteria, Lentisphaerae, and Verrucomicrobia was associated with PTSD status.


Psychiatric Genetics | 2017

Neuroinflammatory genes associated with post-traumatic stress disorder: implications for comorbidity.

Lyndon J. Zass; Stephanie Hart; Soraya Seedat; Sian Hemmings; Stefanie Malan-Müller

Post-traumatic stress disorder (PTSD) is a debilitating condition that only occurs in the aftermath of traumatic event exposure and is characterized by an impaired stress response and chronic, low-grade inflammation. Dysregulation of the immune system may contribute towards central nervous system tissue damage and exacerbation of fear memories following trauma. Patients with PTSD often have comorbid psychiatric and somatic disorders that are of themselves associated with heightened inflammation. Several immune-related genes have been associated with PTSD and other co-occurring disorders. In this review, we propose that chronic inflammation, particularly neuroinflammation, is an important contributory factor towards PTSD comorbidity. Thus, novel treatments that target dysregulated inflammatory processes could provide symptomatic relief from PTSD and its comorbid disorders. This review investigates the intricate links between chronic stress, anxiety and neuroinflammation and the potential impact of increased neuroinflammation on PTSD pathology and comorbidity.


Anxiety Stress and Coping | 2016

Brain-derived neurotrophic factor Val66met polymorphism and plasma levels in road traffic accident survivors

Leigh van den Heuvel; Sharain Suliman; Stefanie Malan-Müller; Sian Hemmings; Soraya Seedat

ABSTRACT Background: Alterations in brain-derived neurotrophic factor (BDNF) expression and release may play a role in the pathogenesis of post-traumatic stress disorder (PTSD). Design: This study evaluated road traffic accident (RTA) survivors to determine whether PTSD and trauma-related factors were associated with plasma BDNF levels and BDNF Val66Met carrier status following RTA exposure. Methods: One hundred and twenty-three RTA survivors (mean age 33.2 years, SD = 10.6 years; 56.9% male) were assessed 10 (SD = 4.9) days after RTA exposure. Acute stress disorder (ASD), as assessed with the Acute Stress Disorder Scale, was present in 50 (42.0%) of the participants. Plasma BDNF levels were measured with enzyme-linked immunosorbent assay and BDNF Val66Met genotyping was performed. PTSD, as assessed with the Clinician-Administered PTSD Scale, was present in 10 (10.8%) participants at 6 months follow-up. Results: Neither BDNF Val66Met genotype nor plasma BDNF was significantly associated with the presence or severity of ASD or PTSD. Plasma BDNF levels were, however, significantly correlated with the lifetime number of trauma exposures. Conclusions: In RTA survivors, plasma BDNF levels increased with increasing number of prior trauma exposures. Plasma BDNF may, therefore, be a marker of trauma load.


Metabolic Brain Disease | 2016

Molecular mechanisms of D-cycloserine in facilitating fear extinction: insights from RNAseq

Stefanie Malan-Müller; Lorren Fairbairn; Willie M. U. Daniels; Mahjoubeh Jalali Sefid Dashti; Marc Altorfer; Martin Kidd; Soraya Seedat; Junaid Gamieldien; Sian Hemmings

D-cycloserine (DCS) has been shown to be effective in facilitating fear extinction in animal and human studies, however the precise mechanisms whereby the co-administration of DCS and behavioural fear extinction reduce fear are still unclear. This study investigated the molecular mechanisms of intrahippocampally administered D-cycloserine in facilitating fear extinction in a contextual fear conditioning animal model. Male Sprague Dawley rats (n = 120) were grouped into four experimental groups (n = 30) based on fear conditioning and intrahippocampal administration of either DCS or saline. The light/dark avoidance test was used to differentiate maladapted (MA) (anxious) from well-adapted (WA) (not anxious) subgroups. RNA extracted from the left dorsal hippocampus was used for RNA sequencing and gene expression data was compared between six fear-conditioned + saline MA (FEAR + SALINE MA) and six fear-conditioned + DCS WA (FEAR + DCS WA) animals. Of the 424 significantly downregulated and 25 significantly upregulated genes identified in the FEAR + DCS WA group compared to the FEAR + SALINE MA group, 121 downregulated and nine upregulated genes were predicted to be relevant to fear conditioning and anxiety and stress-related disorders. The majority of downregulated genes transcribed immune, proinflammatory and oxidative stress systems molecules. These molecules mediate neuroinflammation and cause neuronal damage. DCS also regulated genes involved in learning and memory processes, and genes associated with anxiety, stress-related disorders and co-occurring diseases (e.g., cardiovascular diseases, digestive system diseases and nervous system diseases). Identifying the molecular underpinnings of DCS-mediated fear extinction brings us closer to understanding the process of fear extinction.


Vitamins and Hormones Series | 2017

The Big Role of Small RNAs in Anxiety and Stress-Related Disorders

Stefanie Malan-Müller; Sian Hemmings

In the study of complex, heterogeneous disorders, such as anxiety and stress-related disorders, epigenetic factors provide an additional level of heritable complexity. MicroRNAs (miRNAs) are a class of small, noncoding RNAs that function as epigenetic modulators of gene expression by binding to target messenger RNAs (mRNAs) and subsequently blocking translation or accelerating their degradation. In light of their abundance in the central nervous system (CNS) and their involvement in synaptic plasticity and neuronal differentiation, miRNAs represent an exciting frontier to be explored in the etiology and treatment of anxiety and stress-related disorders. This chapter will present a thorough review of miRNAs, their functions, and mRNA targets in the CNS, focusing on their role in anxiety and stress-related disorders as described by studies performed in animals and human subjects.


Psychiatric Genetics | 2017

The role of micrornas in the therapeutic action of D-cycloserine in a post-traumatic stress disorder animal model: an exploratory study

Stefanie Malan-Müller; Lorren Fairbairn; Stephanie Hart; Willie M. U. Daniels; Mahjoubeh Jalali Sefid Dashti; Martin Kidd; Soraya Seedat; Junaid Gamieldien; Sian Hemmings

Objectives Post-traumatic stress disorder is characterized by impaired fear extinction and excessive anxiety. D-Cycloserine (DCS) has previously been shown to facilitate fear extinction and decrease anxiety in animal and human studies. This study utilized a contextual fear-conditioning animal model to investigate the involvement of microRNAs (miRNAs) in fear extinction and the reduction of anxiety, as mediated by the co-administration of DCS and behavioural fear extinction. Methods Fear conditioning consisted of an electric foot shock; fear extinction consisted of behavioural fear extinction co-administered with either DCS or saline. The light/dark avoidance test was used to evaluate anxiety-related behaviour subsequent to fear conditioning and was used to evaluate anxiety-related behaviour following fear conditioning and to subsequently group animals into well-adapted and maladapted subgroups. These subgroups also showed significant differences in terms of fear extinction. Small RNAs extracted from the left dorsal hippocampus were sequenced using next-generation sequencing to identify differentially expressed miRNAs associated with DCS-induced fear extinction and reduction of anxiety. In-silico prediction analyses identified mRNA targets (from data of the same animals) of the differentially expressed miRNAs. Two of the predicted mRNA–miRNA interactions were functionally investigated. Results Overall, 32 miRNAs were differentially expressed between rats that were fear conditioned, received DCS and were well adapted and rats that were fear conditioned, received saline and were maladapted. Nineteen of these miRNAs were predicted to target and regulate the expression of 63 genes differentially expressed between fear-conditioned, DCS-administered, well-adapted and fear-conditioned, saline-administered, and maladapted groups (several of which are associated with neuronal inflammation, learning and memory). Functional luciferase assays indicated that rno-mir-31a-5p may have regulated the expression of interleukin 1 receptor antagonist (Il1rn) and metallothionein 1a (Mt1a). Conclusion These differentially expressed miRNAs may be mediators of gene expression changes that facilitated decreased neuronal inflammation, optimum learning and memory and contributed towards effective fear extinction and reduction of anxiety following the co-administration of DCS and behavioural fear extinction.

Collaboration


Dive into the Stefanie Malan-Müller's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martin Kidd

Stellenbosch University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew J. Hoisington

United States Air Force Academy

View shared research outputs
Top Co-Authors

Avatar

Christopher A. Lowry

University of Colorado Boulder

View shared research outputs
Top Co-Authors

Avatar

Christopher E. Stamper

University of Colorado Boulder

View shared research outputs
Researchain Logo
Decentralizing Knowledge