Network


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

Hotspot


Dive into the research topics where Jerome Mertens is active.

Publication


Featured researches published by Jerome Mertens.


Nature Neuroscience | 2015

Modifiers of C9orf72 dipeptide repeat toxicity connect nucleocytoplasmic transport defects to FTD/ALS

Ana Jovičić; Jerome Mertens; Steven Boeynaems; Elke Bogaert; Noori Chai; Shizuka Yamada; Joseph West Paul; Shuying Sun; Joseph R Herdy; Gregor Bieri; Nicholas J. Kramer; Fred H. Gage; Ludo Van Den Bosch; Wim Robberecht; Aaron D. Gitler

C9orf72 mutations are the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Dipeptide repeat proteins (DPRs) produced by unconventional translation of the C9orf72 repeat expansions cause neurodegeneration in cell culture and in animal models. We performed two unbiased screens in Saccharomyces cerevisiae and identified potent modifiers of DPR toxicity, including karyopherins and effectors of Ran-mediated nucleocytoplasmic transport, providing insight into potential disease mechanisms and therapeutic targets.C9orf72 mutations are the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Dipeptide repeat proteins (DPRs) produced by unconventional translation of the C9orf72 repeat expansions cause neurodegeneration in cell culture and in animal models. We performed two unbiased screens in Saccharomyces cerevisiae and identified potent modifiers of DPR toxicity, uncovering karyopherins and effectors of Ran-mediated nucleocytoplasmic transport, providing insight into potential disease mechanisms and therapeutic targets.


Nature | 2011

Excitation-induced ataxin-3 aggregation in neurons from patients with Machado–Joseph disease

Philipp Koch; Peter Breuer; Michael Peitz; Johannes Jungverdorben; Jaideep Kesavan; Daniel Poppe; Jonas Doerr; Julia Ladewig; Jerome Mertens; Thomas Tüting; Per Hoffmann; Thomas Klockgether; Bernd O. Evert; Ullrich Wüllner; Oliver Brüstle

Machado–Joseph disease (MJD; also called spinocerebellar ataxia type 3) is a dominantly inherited late-onset neurodegenerative disorder caused by expansion of polyglutamine (polyQ)-encoding CAG repeats in the MJD1 gene (also known as ATXN3). Proteolytic liberation of highly aggregation-prone polyQ fragments from the protective sequence of the MJD1 gene product ataxin 3 (ATXN3) has been proposed to trigger the formation of ATXN3-containing aggregates, the neuropathological hallmark of MJD. ATXN3 fragments are detected in brain tissue of MJD patients and transgenic mice expressing mutant human ATXN3(Q71), and their amount increases with disease severity, supporting a relationship between ATXN3 processing and disease progression. The formation of early aggregation intermediates is thought to have a critical role in disease initiation, but the precise pathogenic mechanism operating in MJD has remained elusive. Here we show that l-glutamate-induced excitation of patient-specific induced pluripotent stem cell (iPSC)-derived neurons initiates Ca2+-dependent proteolysis of ATXN3 followed by the formation of SDS-insoluble aggregates. This phenotype could be abolished by calpain inhibition, confirming a key role of this protease in ATXN3 aggregation. Aggregate formation was further dependent on functional Na+ and K+ channels as well as ionotropic and voltage-gated Ca2+ channels, and was not observed in iPSCs, fibroblasts or glia, thereby providing an explanation for the neuron-specific phenotype of this disease. Our data illustrate that iPSCs enable the study of aberrant protein processing associated with late-onset neurodegenerative disorders in patient-specific neurons.


Nature Methods | 2012

Small molecules enable highly efficient neuronal conversion of human fibroblasts

Julia Ladewig; Jerome Mertens; Jaideep Kesavan; Jonas Doerr; Daniel Poppe; Finnja Glaue; Stefan Herms; Peter Wernet; Gesine Kögler; Franz-Josef Müller; Philipp Koch; Oliver Brüstle

Forced expression of proneural transcription factors has been shown to direct neuronal conversion of fibroblasts. Because neurons are postmitotic, conversion efficiencies are an important parameter for this process. We present a minimalist approach combining two-factor neuronal programming with small molecule–based inhibition of glycogen synthase kinase-3β and SMAD signaling, which converts postnatal human fibroblasts into functional neuron-like cells with yields up to >200% and neuronal purities up to >80%.


Cell Stem Cell | 2015

Directly Reprogrammed Human Neurons Retain Aging-Associated Transcriptomic Signatures and Reveal Age-Related Nucleocytoplasmic Defects

Jerome Mertens; Apuã C. M. Paquola; Manching Ku; Emily M. Hatch; Lena Böhnke; Shauheen Ladjevardi; Sean McGrath; Benjamin Campbell; Hyungjun Lee; Joseph R Herdy; J. Tiago Gonçalves; Tomohisa Toda; Yongsung Kim; Jürgen Winkler; Jun Yao; Martin W. Hetzer; Fred H. Gage

Aging is a major risk factor for many human diseases, and in vitro generation of human neurons is an attractive approach for modeling aging-related brain disorders. However, modeling aging in differentiated human neurons has proved challenging. We generated neurons from human donors across a broad range of ages, either by iPSC-based reprogramming and differentiation or by direct conversion into induced neurons (iNs). While iPSCs and derived neurons did not retain aging-associated gene signatures, iNs displayed age-specific transcriptional profiles and revealed age-associated decreases in the nuclear transport receptor RanBP17. We detected an age-dependent loss of nucleocytoplasmic compartmentalization (NCC) in donor fibroblasts and corresponding iNs and found that reduced RanBP17 impaired NCC in young cells, while iPSC rejuvenation restored NCC in aged cells. These results show that iNs retain important aging-related signatures, thus allowing modeling of the aging process in vitro, and they identify impaired NCC as an important factor in human aging.


Nature | 2015

Differential responses to lithium in hyperexcitable neurons from patients with bipolar disorder

Jerome Mertens; Qiu-Wen Wang; Yongsung Kim; Diana X. Yu; Son Pham; Bo Yang; Yi Zheng; Kenneth E. Diffenderfer; Jian Zhang; Sheila Soltani; Tameji Eames; Simon T. Schafer; Leah Boyer; Maria C. Marchetto; John I. Nurnberger; Joseph R. Calabrese; Ketil J. Oedegaard; Michael McCarthy; Peter P. Zandi; Martin Alda; Caroline M. Nievergelt; Shuangli Mi; Kristen J. Brennand; John R. Kelsoe; Fred H. Gage; Jun Yao

Bipolar disorder is a complex neuropsychiatric disorder that is characterized by intermittent episodes of mania and depression; without treatment, 15% of patients commit suicide. Hence, it has been ranked by the World Health Organization as a top disorder of morbidity and lost productivity. Previous neuropathological studies have revealed a series of alterations in the brains of patients with bipolar disorder or animal models, such as reduced glial cell number in the prefrontal cortex of patients, upregulated activities of the protein kinase A and C pathways and changes in neurotransmission. However, the roles and causation of these changes in bipolar disorder have been too complex to exactly determine the pathology of the disease. Furthermore, although some patients show remarkable improvement with lithium treatment for yet unknown reasons, others are refractory to lithium treatment. Therefore, developing an accurate and powerful biological model for bipolar disorder has been a challenge. The introduction of induced pluripotent stem-cell (iPSC) technology has provided a new approach. Here we have developed an iPSC model for human bipolar disorder and investigated the cellular phenotypes of hippocampal dentate gyrus-like neurons derived from iPSCs of patients with bipolar disorder. Guided by RNA sequencing expression profiling, we have detected mitochondrial abnormalities in young neurons from patients with bipolar disorder by using mitochondrial assays; in addition, using both patch-clamp recording and somatic Ca2+ imaging, we have observed hyperactive action-potential firing. This hyperexcitability phenotype of young neurons in bipolar disorder was selectively reversed by lithium treatment only in neurons derived from patients who also responded to lithium treatment. Therefore, hyperexcitability is one early endophenotype of bipolar disorder, and our model of iPSCs in this disease might be useful in developing new therapies and drugs aimed at its clinical treatment.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Neuronal medium that supports basic synaptic functions and activity of human neurons in vitro

Cedric Bardy; Mark van den Hurk; Tameji Eames; Cynthia Marchand; Ruben V. Hernandez; Mariko Kellogg; Mark Gorris; Ben Galet; Vanessa Palomares; Joshua Brown; Anne G. Bang; Jerome Mertens; Lena Böhnke; Leah Boyer; Suzanne Simon; Fred H. Gage

Significance Neuronal cultures are very valuable to learn about basic principles of the nervous system. In vivo, neural electrical activity is the essence of nervous system function, controlling emotion, memory, sensory modalities, and behavior. In this study, we discovered that many crucial neurophysiological properties were strongly altered in classic culture media that are widely used by the research community. To overcome this problem, we designed and tested a new tissue culture neuromedium that adequately supports in vitro neuronal activity. The improvements made in this medium reduce the gap between in vivo brain physiological conditions and neuronal models in vitro. Improving physiological conditions in vitro may lead to more successful translation from bench to clinics. Human cell reprogramming technologies offer access to live human neurons from patients and provide a new alternative for modeling neurological disorders in vitro. Neural electrical activity is the essence of nervous system function in vivo. Therefore, we examined neuronal activity in media widely used to culture neurons. We found that classic basal media, as well as serum, impair action potential generation and synaptic communication. To overcome this problem, we designed a new neuronal medium (BrainPhys basal + serum-free supplements) in which we adjusted the concentrations of inorganic salts, neuroactive amino acids, and energetic substrates. We then tested that this medium adequately supports neuronal activity and survival of human neurons in culture. Long-term exposure to this physiological medium also improved the proportion of neurons that were synaptically active. The medium was designed to culture human neurons but also proved adequate for rodent neurons. The improvement in BrainPhys basal medium to support neurophysiological activity is an important step toward reducing the gap between brain physiological conditions in vivo and neuronal models in vitro.


Nature Reviews Neuroscience | 2016

Evaluating cell reprogramming, differentiation and conversion technologies in neuroscience

Jerome Mertens; Maria C. Marchetto; Cedric Bardy; Fred H. Gage

The scarcity of live human brain cells for experimental access has for a long time limited our ability to study complex human neurological disorders and elucidate basic neuroscientific mechanisms. A decade ago, the development of methods to reprogramme somatic human cells into induced pluripotent stem cells enabled the in vitro generation of a wide range of neural cells from virtually any human individual. The growth of methods to generate more robust and defined neural cell types through reprogramming and direct conversion into induced neurons has led to the establishment of various human reprogramming-based neural disease models.


American Journal of Pathology | 2012

Presenilin-1 L166P Mutant Human Pluripotent Stem Cell–Derived Neurons Exhibit Partial Loss of γ-Secretase Activity in Endogenous Amyloid-β Generation

Philipp Koch; Irfan Y. Tamboli; Jerome Mertens; Patrick Wunderlich; Julia Ladewig; Kathrin Stüber; Hermann Esselmann; Jens Wiltfang; Oliver Brüstle; Jochen Walter

Alzheimers disease (AD) is the most frequent cause of dementia. There is compelling evidence that the proteolytic processing of the amyloid precursor protein (APP) and accumulation of amyloid-β (Aβ) peptides play critical roles in AD pathogenesis. Due to limited access to human neural tissue, pathogenetic studies have, so far, mostly focused on the heterologous overexpression of mutant human APP in non-human cells. In this study, we show that key steps in proteolytic APP processing are recapitulated in neurons generated from human embryonic and induced pluripotent stem cell-derived neural stem cells (NSC). These human NSC-derived neurons express the neuron-specific APP(695) splice variant, BACE1, and all members of the γ-secretase complex. The human NSC-derived neurons also exhibit a differentiation-dependent increase in Aβ secretion and respond to the pharmacotherapeutic modulation by anti-amyloidogenic compounds, such as γ-secretase inhibitors and nonsteroidal anti-inflammatory drugs. Being highly amenable to genetic modification, human NSCs enable the study of mechanisms caused by disease-associated mutations in human neurons. Interestingly, the AD-associated PS1(L166P) variant revealed a partial loss of γ-secretase function, resulting in the decreased production of endogenous Aβ40 and an increased Aβ42/40 ratio. The PS1(L166P) mutant is also resistant to γ-secretase modulation by nonsteroidal anti-inflammatory drugs. Pluripotent stem cell-derived neurons thus provide experimental access to key steps in AD pathogenesis and can be used to screen pharmaceutical compounds directly in a human neuronal system.


eLife | 2016

Metabolic reprogramming during neuronal differentiation from aerobic glycolysis to neuronal oxidative phosphorylation

Xinde Zheng; Leah Boyer; Mingji Jin; Jerome Mertens; Yongsung Kim; Li Ma; Michael Hamm; Fred H. Gage; Tony Hunter

How metabolism is reprogrammed during neuronal differentiation is unknown. We found that the loss of hexokinase (HK2) and lactate dehydrogenase (LDHA) expression, together with a switch in pyruvate kinase gene splicing from PKM2 to PKM1, marks the transition from aerobic glycolysis in neural progenitor cells (NPC) to neuronal oxidative phosphorylation. The protein levels of c-MYC and N-MYC, transcriptional activators of the HK2 and LDHA genes, decrease dramatically. Constitutive expression of HK2 and LDHA during differentiation leads to neuronal cell death, indicating that the shut-off aerobic glycolysis is essential for neuronal survival. The metabolic regulators PGC-1α and ERRγ increase significantly upon neuronal differentiation to sustain the transcription of metabolic and mitochondrial genes, whose levels are unchanged compared to NPCs, revealing distinct transcriptional regulation of metabolic genes in the proliferation and post-mitotic differentiation states. Mitochondrial mass increases proportionally with neuronal mass growth, indicating an unknown mechanism linking mitochondrial biogenesis to cell size. DOI: http://dx.doi.org/10.7554/eLife.13374.001


Molecular Psychiatry | 2016

Generation of functional human serotonergic neurons from fibroblasts.

Krishna C. Vadodaria; Jerome Mertens; Apuã C. M. Paquola; Cedric Bardy; X. Li; Roberto Jappelli; L. Fung; Maria C. Marchetto; M. Hamm; M.A.J. Gorris; P. Koch; Fred H. Gage

The brain’s serotonergic system centrally regulates several physiological processes and its dysfunction has been implicated in the pathophysiology of several neuropsychiatric disorders. While in the past our understanding of serotonergic neurotransmission has come mainly from mouse models, the development of pluripotent stem cell and induced fibroblast-to-neuron (iN) transdifferentiation technologies has revolutionized our ability to generate human neurons in vitro. Utilizing these techniques and a novel lentiviral reporter for serotonergic neurons, we identified and overexpressed key transcription factors to successfully generate human serotonergic neurons. We found that overexpressing the transcription factors NKX2.2, FEV, GATA2 and LMX1B in combination with ASCL1 and NGN2 directly and efficiently generated serotonergic neurons from human fibroblasts. Induced serotonergic neurons (iSNs) showed increased expression of specific serotonergic genes that are known to be expressed in raphe nuclei. iSNs displayed spontaneous action potentials, released serotonin in vitro and functionally responded to selective serotonin reuptake inhibitors (SSRIs). Here, we demonstrate the efficient generation of functional human serotonergic neurons from human fibroblasts as a novel tool for studying human serotonergic neurotransmission in health and disease.

Collaboration


Dive into the Jerome Mertens's collaboration.

Top Co-Authors

Avatar

Fred H. Gage

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maria C. Marchetto

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Yongsung Kim

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leah Boyer

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph R. Calabrese

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Kristen J. Brennand

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge