Network


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

Hotspot


Dive into the research topics where Matthew B. Harms is active.

Publication


Featured researches published by Matthew B. Harms.


Science | 2015

Exome sequencing in amyotrophic lateral sclerosis identifies risk genes and pathways

Elizabeth T. Cirulli; Brittany N. Lasseigne; Slavé Petrovski; Peter C. Sapp; Patrick A. Dion; Claire S. Leblond; Julien Couthouis; Yi Fan Lu; Quanli Wang; Brian Krueger; Zhong Ren; Jonathan Keebler; Yujun Han; Shawn Levy; Braden E. Boone; Jack R. Wimbish; Lindsay L. Waite; Angela L. Jones; John P. Carulli; Aaron G. Day-Williams; John F. Staropoli; Winnie Xin; Alessandra Chesi; Alya R. Raphael; Diane McKenna-Yasek; Janet Cady; J.M.B.Vianney de Jong; Kevin Kenna; Bradley Smith; Simon Topp

New players in Lou Gehrigs disease Amyotrophic lateral sclerosis (ALS), often referred to as “Lou Gehrigs disease,” is a progressive neurodegenerative disease that affects nerve cells in the brain and the spinal cord. Cirulli et al. sequenced the expressed genes of nearly 3000 ALS patients and compared them with those of more than 6000 controls (see the Perspective by Singleton and Traynor). They identified several proteins that were linked to disease in patients. One such protein, TBK1, is implicated in innate immunity and autophagy and may represent a therapeutic target. Science, this issue p. 1436; see also p. 1422 Analysis of the expressed genes of nearly 2900 patients with amyotrophic lateral sclerosis and about 6400 controls reveals a disease predisposition–associated gene. [Also see Perspective by Singleton and Traynor] Amyotrophic lateral sclerosis (ALS) is a devastating neurological disease with no effective treatment. We report the results of a moderate-scale sequencing study aimed at increasing the number of genes known to contribute to predisposition for ALS. We performed whole-exome sequencing of 2869 ALS patients and 6405 controls. Several known ALS genes were found to be associated, and TBK1 (the gene encoding TANK-binding kinase 1) was identified as an ALS gene. TBK1 is known to bind to and phosphorylate a number of proteins involved in innate immunity and autophagy, including optineurin (OPTN) and p62 (SQSTM1/sequestosome), both of which have also been implicated in ALS. These observations reveal a key role of the autophagic pathway in ALS and suggest specific targets for therapeutic intervention.


Science Translational Medicine | 2013

Targeting RNA foci in iPSC-derived motor neurons from ALS patients with a C9ORF72 repeat expansion

Dhruv Sareen; Jacqueline G O'Rourke; P. Meera; A. K. M. G. Muhammad; Sharday Grant; Megan Simpkinson; Shaughn Bell; Sharon Carmona; Loren Ornelas; Anais Sahabian; Tania F. Gendron; Leonard Petrucelli; Michael Baughn; John Ravits; Matthew B. Harms; Frank Rigo; C. F. Bennett; T. S. Otis; Clive N. Svendsen; Robert H. Baloh

Antisense oligonucleotides can correct disease-specific phenotypes in cultured motor neurons differentiated from iPSCs derived from ALS patients with a C9ORF72 repeat expansion. Clearing Toxic RNA in ALS Amyotrophic lateral sclerosis (ALS, or Lou Gehrig’s disease) is a uniformly fatal disease caused by the death of cells in the nervous system that control the musculature. Patients slowly become paralyzed and lose the ability to breathe, and no effective therapies currently exist. The expansion of a repeated DNA element (GGGGCC) in a gene called C9ORF72 was recently identified as the most common genetic cause of ALS. In their new study, Sareen et al. set out to understand how the expansion of the GGGGCC repeat in C9ORF72 causes cell degeneration. They took skin cells from patients with the disease and converted them into motor neurons in a culture dish, the cells that die in ALS patients. They found that large pieces of RNA containing the expanded GGGGCC repeat built up in neurons from ALS patients and disrupted the function of these cells. Furthermore, they observed that oligonucleotides complementary to the C9ORF72 RNA transcript sequence (“antisense oligonucleotides”) suppressed the formation of these RNA foci. These findings support the idea that the buildup of “toxic” RNA containing the GGGGCC repeat contributes to the death of motor neurons in ALS, and suggest that antisense oligonucleotides targeting this transcript may be a strategy for treating ALS patients with the C9ORF72 repeat expansion. Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative condition characterized by loss of motor neurons in the brain and spinal cord. Expansions of a hexanucleotide repeat (GGGGCC) in the noncoding region of the C9ORF72 gene are the most common cause of the familial form of ALS (C9-ALS), as well as frontotemporal lobar degeneration and other neurological diseases. How the repeat expansion causes disease remains unclear, with both loss of function (haploinsufficiency) and gain of function (either toxic RNA or protein products) proposed. We report a cellular model of C9-ALS with motor neurons differentiated from induced pluripotent stem cells (iPSCs) derived from ALS patients carrying the C9ORF72 repeat expansion. No significant loss of C9ORF72 expression was observed, and knockdown of the transcript was not toxic to cultured human motor neurons. Transcription of the repeat was increased, leading to accumulation of GGGGCC repeat–containing RNA foci selectively in C9-ALS iPSC-derived motor neurons. Repeat-containing RNA foci colocalized with hnRNPA1 and Pur-α, suggesting that they may be able to alter RNA metabolism. C9-ALS motor neurons showed altered expression of genes involved in membrane excitability including DPP6, and demonstrated a diminished capacity to fire continuous spikes upon depolarization compared to control motor neurons. Antisense oligonucleotides targeting the C9ORF72 transcript suppressed RNA foci formation and reversed gene expression alterations in C9-ALS motor neurons. These data show that patient-derived motor neurons can be used to delineate pathogenic events in ALS.


Proceedings of the National Academy of Sciences of the United States of America | 2013

RAN proteins and RNA foci from antisense transcripts in C9ORF72 ALS and frontotemporal dementia

Tao Zu; Yuanjing Liu; Monica Bañez-Coronel; Tammy Reid; Olga Pletnikova; Jada Lewis; Timothy M. Miller; Matthew B. Harms; Annet E. Falchook; S. H. Subramony; Lyle W. Ostrow; Jeffrey D. Rothstein; Juan C. Troncoso; Laura P.W. Ranum

Significance A GGGGCC expansion mutation located in intron 1 of chromosome 9 ORF 72 (C9ORF72) was recently described as a common cause of familial amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD). We show that this single mutation results in the accumulation of sense and antisense RNA foci plus six expansion proteins expressed by repeat-associated non-ATG (RAN) translation. RNAs accumulate in nuclear foci and the RAN proteins form cytoplasmic aggregates in neurons that often cluster in affected brain regions. These results indicate that bidirectional transcription and RAN translation are fundamental pathologic features of C9ORF72 ALS/FTD. Additionally these data have broad implications that change our understanding of how microsatellite expansion mutations are expressed in patient cells and how they cause disease. The finding that a GGGGCC (G4C2) hexanucleotide repeat expansion in the chromosome 9 ORF 72 (C9ORF72) gene is a common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) links ALS/FTD to a large group of unstable microsatellite diseases. Previously, we showed that microsatellite expansion mutations can be bidirectionally transcribed and that these mutations express unexpected proteins by a unique mechanism, repeat-associated non-ATG (RAN) translation. In this study, we show that C9ORF72 antisense transcripts are elevated in the brains of C9ORF72 expansion-positive [C9(+)] patients, and antisense GGCCCC (G2C4) repeat-expansion RNAs accumulate in nuclear foci in brain. Additionally, sense and antisense foci accumulate in blood and are potential biomarkers of the disease. Furthermore, we show that RAN translation occurs from both sense and antisense expansion transcripts, resulting in the expression of six RAN proteins (antisense: Pro-Arg, Pro-Ala, Gly-Pro; and sense: Gly-Ala, Gly-Arg, Gly-Pro). These proteins accumulate in cytoplasmic aggregates in affected brain regions, including the frontal and motor cortex, hippocampus, and spinal cord neurons, with some brain regions showing dramatic RAN protein accumulation and clustering. The finding that unique antisense G2C4 RNA foci and three unique antisense RAN proteins accumulate in patient tissues indicates that bidirectional transcription of expanded alleles is a fundamental pathologic feature of C9ORF72 ALS/FTD. Additionally, these findings suggest the need to test therapeutic strategies that target both sense and antisense RNAs and RAN proteins in C9ORF72 ALS/FTD, and to more broadly consider the role of antisense expression and RAN translation across microsatellite expansion diseases.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Targeted degradation of sense and antisense C9orf72 RNA foci as therapy for ALS and frontotemporal degeneration.

Clotilde Lagier-Tourenne; Michael Baughn; Frank Rigo; Shuying Sun; Patrick Liu; Hairi Li; Jie Jiang; Andrew T. Watt; Seung Chun; Melanie Katz; Jinsong Qiu; Ying Sun; Shuo-Chien Ling; Qiang Zhu; Magdalini Polymenidou; Kevin Drenner; Jonathan W. Artates; Melissa McAlonis-Downes; Sebastian Markmiller; Kasey R. Hutt; Donald P. Pizzo; Janet Cady; Matthew B. Harms; Robert H. Baloh; Scott R. VandenBerg; Gene W. Yeo; Xiang-Dong Fu; C. Frank Bennett; Don W. Cleveland; John Ravits

Significance The most frequent genetic cause of ALS and frontotemporal degeneration is a hexanucleotide expansion in a noncoding region of the C9orf72 gene. Similar to other repeat expansion diseases, we characterize the hallmark feature of repeat expansion RNA-mediated toxicity: nuclear RNA foci. Remarkably, two distinct sets of foci are found, one containing RNAs transcribed in the sense direction and the other containing antisense RNAs. Antisense oligonucleotides (ASOs) are developed that selectively target sense strand repeat-containing RNAs and reduce sense-oriented foci without affecting overall C9orf72 expression. Importantly, reducing C9orf72 expression does not cause behavioral or pathological changes in mice and induces only a few genome-wide mRNA alterations. These findings establish ASO-mediated degradation of repeat-containing RNAs as a significant therapeutic approach. Expanded hexanucleotide repeats in the chromosome 9 open reading frame 72 (C9orf72) gene are the most common genetic cause of ALS and frontotemporal degeneration (FTD). Here, we identify nuclear RNA foci containing the hexanucleotide expansion (GGGGCC) in patient cells, including white blood cells, fibroblasts, glia, and multiple neuronal cell types (spinal motor, cortical, hippocampal, and cerebellar neurons). RNA foci are not present in sporadic ALS, familial ALS/FTD caused by other mutations (SOD1, TDP-43, or tau), Parkinson disease, or nonneurological controls. Antisense oligonucleotides (ASOs) are identified that reduce GGGGCC-containing nuclear foci without altering overall C9orf72 RNA levels. By contrast, siRNAs fail to reduce nuclear RNA foci despite marked reduction in overall C9orf72 RNAs. Sustained ASO-mediated lowering of C9orf72 RNAs throughout the CNS of mice is demonstrated to be well tolerated, producing no behavioral or pathological features characteristic of ALS/FTD and only limited RNA expression alterations. Genome-wide RNA profiling identifies an RNA signature in fibroblasts from patients with C9orf72 expansion. ASOs targeting sense strand repeat-containing RNAs do not correct this signature, a failure that may be explained, at least in part, by discovery of abundant RNA foci with C9orf72 repeats transcribed in the antisense (GGCCCC) direction, which are not affected by sense strand-targeting ASOs. Taken together, these findings support a therapeutic approach by ASO administration to reduce hexanucleotide repeat-containing RNAs and raise the potential importance of targeting expanded RNAs transcribed in both directions.


JAMA Neurology | 2014

TREM2 Variant p.R47H as a Risk Factor for Sporadic Amyotrophic Lateral Sclerosis

Janet Cady; Erica D. Koval; Bruno A. Benitez; Craig M. Zaidman; Jennifer Jockel-Balsarotti; Peggy Allred; Robert H Baloh; John Ravits; Ericka Simpson; Stanley H. Appel; Alan Pestronk; Alison Goate; Timothy M. Miller; Carlos Cruchaga; Matthew B. Harms

IMPORTANCE Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease in which microglia play a significant and active role. Recently, a rare missense variant (p.R47H) in the microglial activating gene TREM2 was found to increase the risk of several neurodegenerative diseases, including Alzheimer disease. Whether the p.R47H variant is a risk factor for ALS is not known. OBJECTIVES To determine whether p.R47H (rs75932628) in TREM2 is a risk factor for ALS and assess whether TREM2 expression is dysregulated in disease. DESIGN, SETTING, AND PARTICIPANTS Samples of DNA from 923 individuals with sporadic ALS and 1854 healthy control individuals self-reported as non-Hispanic white were collected from ALS clinics in the United States and genotyped for the p.R47H variant in TREM2. Clinical data were obtained on ALS participants for genotype/phenotype correlations. Expression of TREM2 was measured by quantitative polymerase chain reaction and compared in spinal cord samples from 18 autopsied patients with ALS and 12 neurologically healthy controls, as well as from wild-type and transgenic SOD1G93A mice. MAIN OUTCOMES AND MEASURES Minor allele frequency of rs75932628 and relative expression of TREM2. RESULTS The TREM2 variant p.R47H was more common in patients with ALS than in the controls and is therefore a significant risk factor for ALS (odds ratio, 2.40; 95% CI, 1.29-4.15; P = 4.1×10-3). Furthermore, TREM2 expression was increased in spinal cord samples from ALS patients and SOD1G93A mice (P = 2.8×10-4 and P = 2.8×10-9, respectively), confirming dysregulated TREM2 in disease. Expression of TREM2 in the human spinal cord was negatively correlated with survival (P = .04) but not with other phenotypic aspects of disease. CONCLUSIONS AND RELEVANCE This study demonstrates that the TREM2 p.R47H variant is a potent risk factor for sporadic ALS. To our knowledge, these findings identify the first genetic influence on neuroinflammation in ALS and highlight the TREM2 signaling pathway as a therapeutic target in ALS and other neurodegenerative diseases.


Neurology | 2012

Mutations in the tail domain of DYNC1H1 cause dominant spinal muscular atrophy

Matthew B. Harms; Kassandra M. Ori-McKenney; M. Scoto; E.P. Tuck; Shaughn Bell; Duanduan Ma; S. Masi; Peggy Allred; M. T. Al-Lozi; Mary M. Reilly; Lindsey J. Miller; Agnes Jani-Acsadi; Alan Pestronk; Michael E. Shy; Francesco Muntoni; Richard B. Vallee; Robert H. Baloh

Objective: To identify the gene responsible for 14q32-linked dominant spinal muscular atrophy with lower extremity predominance (SMA-LED, OMIM 158600). Methods: Target exon capture and next generation sequencing was used to analyze the 73 genes in the 14q32 linkage interval in 3 SMA-LED family members. Candidate gene sequencing in additional dominant SMA families used PCR and pooled target capture methods. Patient fibroblasts were biochemically analyzed. Results: Regional exome sequencing of all candidate genes in the 14q32 interval in the original SMA-LED family identified only one missense mutation that segregated with disease state—a mutation in the tail domain of DYNC1H1 (I584L). Sequencing of DYNC1H1 in 32 additional probands with lower extremity predominant SMA found 2 additional heterozygous tail domain mutations (K671E and Y970C), confirming that multiple different mutations in the same domain can cause a similar phenotype. Biochemical analysis of dynein purified from patient-derived fibroblasts demonstrated that the I584L mutation dominantly disrupted dynein complex stability and function. Conclusions: We demonstrate that mutations in the tail domain of the heavy chain of cytoplasmic dynein (DYNC1H1) cause spinal muscular atrophy and provide experimental evidence that a human DYNC1H1 mutation disrupts dynein complex assembly and function. DYNC1H1 mutations were recently found in a family with Charcot-Marie-Tooth disease (type 2O) and in a child with mental retardation. Both of these phenotypes show partial overlap with the spinal muscular atrophy patients described here, indicating that dynein dysfunction is associated with a range of phenotypes in humans involving neuronal development and maintenance.


Neuron | 2015

C9orf72 BAC Transgenic Mice Display Typical Pathologic Features of ALS/FTD

Jacqueline G O'Rourke; Laurent Bogdanik; A. K. M. G. Muhammad; Tania F. Gendron; Kevin Kim; Andrew Austin; Janet Cady; Elaine Y. Liu; Jonah Zarrow; Sharday Grant; Ritchie Ho; Shaughn Bell; Sharon Carmona; Megan Simpkinson; Deepti Lall; Kathryn Wu; Lillian M. Daughrity; Dennis W. Dickson; Matthew B. Harms; Leonard Petrucelli; Edward B. Lee; Cathleen Lutz; Robert H. Baloh

Noncoding expansions of a hexanucleotide repeat (GGGGCC) in the C9orf72 gene are the most common cause of familial amyotrophic lateral sclerosis and frontotemporal dementia. Here we report transgenic mice carrying a bacterial artificial chromosome (BAC) containing the full human C9orf72 gene with either a normal allele (15 repeats) or disease-associated expansion (∼100-1,000 repeats; C9-BACexp). C9-BACexp mice displayed pathologic features seen in C9orf72 expansion patients, including widespread RNA foci and repeat-associated non-ATG (RAN) translated dipeptides, which were suppressed by antisense oligonucleotides targeting human C9orf72. Nucleolin distribution was altered, supporting that either C9orf72 transcripts or RAN dipeptides promote nucleolar dysfunction. Despite early and widespread production of RNA foci and RAN dipeptides in C9-BACexp mice, behavioral abnormalities and neurodegeneration were not observed even at advanced ages, supporting the hypothesis that RNA foci and RAN dipeptides occur presymptomatically and are not sufficient to drive neurodegeneration in mice at levels seen in patients.


Annals of Neurology | 2012

Exome sequencing reveals DNAJB6 mutations in dominantly-inherited myopathy

Matthew B. Harms; R. Brian Sommerville; Peggy Allred; Shaughn Bell; Duanduan Ma; Paul R. Cooper; Glenn Lopate; Alan Pestronk; Conrad C. Weihl; Robert H. Baloh

To identify the causative gene in an autosomal dominant limb‐girdle muscular dystrophy (LGMD) with skeletal muscle vacuoles.


JAMA Neurology | 2013

C9orf72 hexanucleotide repeat expansions in clinical Alzheimer disease.

Matthew B. Harms; Bruno A. Benitez; Nigel J. Cairns; Breanna Cooper; Paul R. Cooper; Kevin Mayo; David Carrell; Kelley Faber; Jennifer Williamson; Bird Td; Ramon Diaz-Arrastia; Tatiana Foroud; Bradley F. Boeve; Neill R. Graff-Radford; Richard Mayeux; Sumitra Chakraverty; Alison Goate; Carlos Cruchaga

IMPORTANCE Hexanucleotide repeat expansions in the chromosome 9 open reading frame 72 (C9orf72) gene underlie a significant fraction of frontotemporal dementia and amyotrophic lateral sclerosis. OBJECTIVE To investigate the frequency of C9orf72 repeat expansions in clinically diagnosed late-onset Alzheimer disease (AD). DESIGN, SETTING, AND PATIENTS This case-control study genotyped the C9orf72 repeat expansion in 872 unrelated familial AD cases and 888 control subjects recruited as part of the National Institute on Aging Late-Onset Alzheimer Disease Family Study cohort, a multisite collaboration studying 1000 families with 2 or more individuals clinically diagnosed as having late-onset AD. MAIN OUTCOMES AND MEASURES We determined the presence or absence of the C9orf72 repeat expansion by repeat-primed polymerase chain reaction, the length of the longest nonexpanded allele, segregation of the genotype with disease, and clinical features of repeat expansion carriers. RESULTS Three families showed large C9orf72 hexanucleotide repeat expansions. Two additional families carried more than 30 repeats. Segregation with disease could be demonstrated in 3 families. One affected expansion carrier had neuropathology compatible with AD. In the National Institute on Aging Late-Onset Alzheimer Disease Family Study series, the C9orf72 repeat expansions constituted the second most common pathogenic mutation, just behind the PSEN1 A79V mutation, highlighting the heterogeneity of clinical presentations associated with repeat expansions. CONCLUSIONS AND RELEVANCE C9orf72 repeat expansions explain a small proportion of patients with a clinical presentation indistinguishable from AD, and they highlight the necessity of screening frontotemporal dementia genes in clinical AD cases with strong family history.


Neurobiology of Aging | 2013

Lack of C9ORF72 coding mutations supports a gain of function for repeat expansions in amyotrophic lateral sclerosis

Matthew B. Harms; Janet Cady; Craig M. Zaidman; Paul R. Cooper; Taha Bali; Peggy Allred; Carlos Cruchaga; Michael Baughn; Ryan T. Libby; Alan Pestronk; Alison Goate; John Ravits; Robert H. Baloh

Hexanucleotide repeat expansions in C9ORF72 are a common cause of familial and apparently sporadic amyotrophic lateral sclerosis (ALS) and frontal temporal dementia (FTD). The mechanism by which expansions cause neurodegeneration is unknown, but current evidence supports both loss-of-function and gain-of-function mechanisms. We used pooled next-generation sequencing of the C9ORF72 gene in 389 ALS patients to look for traditional loss-of-function mutations. Although rare variants were identified, none were likely to be pathogenic, suggesting that mutations other than the repeat expansion are not a common cause of ALS, and providing supportive evidence for a gain-of-function mechanism. We also show by repeat-primed PCR genotyping that the C9ORF72 expansion frequency varies by geographical region within the United States, with an unexpectedly high frequency in the Mid-West. Finally we also show evidence of somatic instability of the expansion size by Southern blot, with the largest expansions occurring in brain tissue.

Collaboration


Dive into the Matthew B. Harms's collaboration.

Top Co-Authors

Avatar

Robert H. Baloh

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alan Pestronk

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Peggy Allred

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Timothy M. Miller

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Janet Cady

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Alison Goate

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

John Ravits

University of California

View shared research outputs
Top Co-Authors

Avatar

Carlos Cruchaga

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Conrad C. Weihl

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Shaughn Bell

Cedars-Sinai Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge