Network


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

Hotspot


Dive into the research topics where Anita P. Merriam is active.

Publication


Featured researches published by Anita P. Merriam.


Journal of Bone and Mineral Research | 1999

A quadripotential mesenchymal progenitor cell isolated from the marrow of an adult mouse.

James E. Dennis; Anita P. Merriam; Amad Awadallah; Jung U. Yoo; Brian Johnstone; Arnold I. Caplan

Adult marrow contains mesenchymal progenitor cells (MPCs) that have multiple differentiation potentials. A conditionally immortalized MPC clone, BMC9, has been identified that exhibits four mesenchymal cell phenotypes: chondrocyte, adipocyte, stromal (support osteoclast formation), and osteoblast. The BMC9 clone, control brain fibroblasts and another marrow‐derived clone, BMC10, were isolated from a transgenic mouse (H‐2Kb‐tsA58) containing a gene for conditional immortality. To test for chondrogenic potential, cells were cultured in defined medium containing 10 ng/ml transforming growth factor β and 10−7 M dexamethasone in 15‐ml polypropylene tubes (“aggregate cultures”). Adipogenic potential was quantitated by flow cytometry of Nile Red–stained cells cultured for 1 and 2 weeks in medium containing isobutyl methylxanthine, indomethacin, insulin, and dexamethasone. Support of osteoclast formation was measured by quantitating multinucleated tartrate‐resistant acid phosphatase–positive cells in spleen cell cocultures of test clones (immortomouse clones and positive control ST2 cells) cultured in the presence of 10−7 M vitamin D3 and 150 mM ascorbate‐2‐phosphate. In vivo osteogenic potential was assayed by histologic examination of bone formation in subcutaneous implants, into athymic mouse hosts, of a composite of cells combined with porous calcium phosphate ceramics. The bone marrow–derived clone BMC9 has the potential to express each of the four mesenchymal characteristics tested, while brain fibroblasts, tested under identical conditions, did not exhibit any of these four mesenchymal characteristics. BMC10 cells exhibited osteogenic and chondrogenic phenotypes, but showed only minimal expression of adipocytic or osteoclast‐supportive phenotypes. Clone BMC9 is, minimally, a quadripotential MPC isolated from the marrow of an adult mouse that can differentiate into cartilage and adipose, support osteoclast formation, and form bone. The BMC9 clone is an example of an adult‐derived multipotential progenitor cell that is situated early in the mesenchymal lineage.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Extraocular muscle is defined by a fundamentally distinct gene expression profile.

John D. Porter; Sangeeta Khanna; Henry J. Kaminski; J. S. Rao; Anita P. Merriam; Chelliah R. Richmonds; Patrick Leahy; J. Li; Francisco H. Andrade

Skeletal muscle fibers are defined by patterned covariation of key traits that determine contractile and metabolic characteristics. Although the functional properties of most skeletal muscles result from their proportional content of a few conserved muscle fiber types, some, typically craniofacial, muscles exhibit fiber types that appear to lie outside the common phenotypic range. We analyzed gene expression profiles of three putative muscle classes, limb, masticatory, and extraocular muscle (EOM), in adult mice by high-density oligonucleotide arrays. Pairwise comparisons using conservative acceptance criteria identified expression differences in 287 genes between EOM and limb and/or masticatory muscles. Use of significance analysis of microarrays methodology identified up to 400 genes as having an EOM-specific expression pattern. Genes differentially expressed in EOM reflect key aspects of muscle biology, including transcriptional regulation, sarcomeric organization, excitation-contraction coupling, intermediary metabolism, and immune response. These patterned differences in gene expression define EOM as a distinct muscle class and may explain the unique response of these muscles in neuromuscular diseases.


Neuromuscular Disorders | 2003

Persistent over-expression of specific CC class chemokines correlates with macrophage and T-cell recruitment in mdx skeletal muscle

John D. Porter; Wei Guo; Anita P. Merriam; Sangeeta Khanna; Georgiana Cheng; Xiaohua Zhou; Francisco H. Andrade; Chellah Richmonds; Henry J. Kaminski

Prior studies and the efficacy of immunotherapies provide evidence that inflammation is mechanistic in pathogenesis of Duchenne muscular dystrophy. To identify putative pro-inflammatory mechanisms, we evaluated chemokine gene/protein expression patterns in skeletal muscle of mdx mice. By DNA microarray, reverse transcription-polymerase chain reaction, quantitative polymerase chain reaction, and immunoblotting, convergent evidence established the induction of six distinct CC class chemokine ligands in adult MDX: CCL2/MCP-1, CCL5/RANTES, CCL6/mu C10, CCL7/MCP-3, CCL8/MCP-2, and CCL9/MIP-1gamma. CCL receptors, CCR2, CCR1, and CCR5, also showed increased expression in mdx muscle. CCL2 and CCL6 were localized to both monocular cells and muscle fibers, suggesting that dystrophic muscle may contribute toward chemotaxis. Temporal patterns of CCL2 and CCL6 showed early induction and maintained expression in mdx limb muscle. These data raise the possibility that chemokine signaling pathways coordinate a spatially and temporally discrete immune response that may contribute toward muscular dystrophy. The chemokine pro-inflammatory pathways described here in mdx may represent new targets for treatment of Duchenne muscular dystrophy.


The FASEB Journal | 2003

Constitutive properties, not molecular adaptations, mediate extraocular muscle sparing in dystrophic mdx mice

John D. Porter; Anita P. Merriam; Sangeeta Khanna; Francisco H. Andrade; Chelliah R. Richmonds; Patrick Leahy; Georgiana Cheng; Xiaohua Zhou; Linda L. Kusner; Marvin E. Adams; Michael Willem; Ulrike Mayer; Henry J. Kaminski

Extraocular muscle (EOM) is spared in Duchenne muscular dystrophy. Here, we tested putative EOM sparing mechanisms predicted from existing dystrophinopathy models. Data show that mdx mouse EOM contains dystrophin‐glycoprotein complex (DGC)‐competent and DGC‐deficient myofibers distributed in a fiber type‐specific pattern. Up‐regulation of a dystrophin homologue, utrophin, mediates selective DGC retention. Counter to the DGC mechanical hypothesis, an intact DGC is not a precondition for EOM sarcolemmal integrity, and active adaptation at the level of calcium homeostasis is not mechanistic in protection. A partial, fiber type‐specific retention of antiischemic nitric oxide to vascular smooth muscle signaling is not a factor in EOM sparing, because mice deficient in dystrophin and α‐syntrophin, which localizes neuronal nitric oxide synthase to the sarcolemma, have normal EOMs. Moreover, an alternative transmembrane protein, α7β1 integrin, does not appear to substitute for the DGC in EOM. Finally, genomewide expression profiling showed that EOM does not actively adapt to dystrophinopathy but identified candidate genes for the constitutive protection of mdx EOM. Taken together, data emphasize the conditional nature of dystrophinopathy and the potential importance of nonmechanical DGC roles and support the hypothesis that broad, constitutive structural cell signaling, and/or biochemical differences between EOM and other skeletal muscles are determinants of differential disease responsiveness.


Neuromuscular Disorders | 2006

Temporal and spatial mRNA expression patterns of TGF-β1, 2, 3 and TβRI, II, III in skeletal muscles of mdx mice

Lan Zhou; John D. Porter; Georgiana Cheng; Bendi Gong; Denise A. Hatala; Anita P. Merriam; Xiaohua Zhou; Jill A. Rafael; Henry J. Kaminski

Abstract To address potential regulatory roles of TGF-β1 in muscle inflammation and fibrosis associated with dystrophin deficiency, we performed quantitative RT-PCR and in situ hybridization to characterize the temporal and spatial mRNA expression patterns of TGF-β1 and other TGF-β subfamily members, TGF-β2 and TGF-β3, as well as their receptors, in quadriceps and diaphragm muscles of mdx mice. TGF-β1 mRNA was markedly upregulated in the endomysial inflammatory cells and regenerating fibers of mdx quadriceps and diaphragm, with the mRNA levels correlated with the degree of endomysial inflammation. Upregulation of TGF-β2, β3, and their receptors was also appreciated but to a much lesser degree. While high levels of TGF-β1 mRNA remained in the aging mdx quadriceps but not the diaphragm, progressive fibrosis only occurred in the diaphragm. Our data support a regulatory role for TGF-β1 in muscle inflammation in mdx mice. It also suggests different susceptibility of quadriceps and diaphragm muscles to fibrosis induced by TGF-β1 signaling pathway.


Neuromuscular Disorders | 2001

Extraocular muscle is spared despite the absence of an intact sarcoglycan complex in γ- or δ-sarcoglycan-deficient mice

John D. Porter; Anita P. Merriam; Andrew A. Hack; Francisco H. Andrade; Elizabeth M. McNally

Abstract Models of the dystrophin–glycoprotein complex do not reconcile the novel sparing of extraocular muscle in muscular dystrophy. Extraocular muscle sparing in Duchenne muscular dystrophy implies the existence of adaptive properties in these muscles that may extend protection to other neuromuscular diseases. We studied the extraocular muscle morphology and dystrophin–glycoprotein complex organization in murine targeted deletion of the γ-sarcoglycan ( gsg −/− ) and δ-sarcoglycan ( dsg −/− ) genes, two models of autosomal recessive limb girdle muscular dystrophy. In contrast to limb and diaphragm, the principal extraocular muscles were intact in gsg −/− and dsg −/− mice. However, central nucleated, presumptive regenerative, fibers were seen in the accessory extraocular muscles (retractor bulbi, levator palpebrae superioris) of both strains. Skeletal muscles of gsg −/− mice exhibited in vivo Evans Blue dye permeability, while the principal extraocular muscles did not. Disruption of γ-sarcoglycan produced secondary displacement of α- and β-sarcoglycans in the extraocular muscles. The intensity of immunofluorescence for dystrophin and α- and β-dystroglycan also appeared to be slightly reduced. Utrophin localization was unchanged. The finding that sarcoglycan disruption was insufficient to elicit alterations in extraocular muscle suggests that loss of mechanical stability and increased sarcolemmal permeability are not inevitable consequences of mutations that disrupt the dystrophin–glycoprotein complex organization and must be accounted for in models of muscular dystrophy.


The FASEB Journal | 2003

Comprehensive expression profiling by muscle tissue class and identification of the molecular niche of extraocular muscle

Sangeeta Khanna; Anita P. Merriam; Bendi Gong; Patrick Leahy; John D. Porter

Muscle tissue is an elegant model for biologic integration of structure with function and is frequently affected by a variety of inherited diseases. Traditional muscle classes‐‐skeletal, cardiac, and smooth‐‐share basic aspects of contractile and energetics mechanisms but also have distinctive role‐specific adaptations. We used large‐scale oligonucleotide microarrays to broaden knowledge of the adaptive expression patterns underlying muscle tissue differences and to identify transcript subsets that are most likely to represent candidate disease genes. Using stringent analysis criteria, we found ≥95 transcripts, which were preferentially expressed by each muscle class and were validated by inclusion of known muscle class‐specific and inherited disease‐related genes. Differentially expressed transcripts not previously identified as class‐specific extend understanding of muscle class transcriptomes and may represent novel muscle‐specific disease genes. We also analyzed the expression profile of extraocular muscle, which is divergent from other skeletal muscles, in the broader context of all major muscle classes. Data show that the extraocular muscle phenotype results from the combination of tissue‐specific transcripts, novel expression levels of skeletal muscle transcripts, and partial sharing of gene expression patterns with cardiac and smooth muscle. These, and additional proteomic data, establish that extraocular muscle does not constitute a distinctive muscle class but that it does occupy a novel niche within the skeletal muscle class.


Neurogenetics | 2006

Analysis of gene expression differences between utrophin/dystrophin-deficient vs mdx skeletal muscles reveals a specific upregulation of slow muscle genes in limb muscles

Patrick Ericson Baker; Jessica A. Kearney; Bendi Gong; Anita P. Merriam; Donald E. Kuhn; John D. Porter; Jill A. Rafael-Fortney

Dystrophin deficiency leads to the progressive muscle wasting disease Duchenne muscular dystrophy (DMD). Dystrophin-deficient mdx mice are characterized by skeletal muscle weakness and degeneration but they appear outwardly normal in contrast to DMD patients. Mice lacking both dystrophin and the dystrophin homolog utrophin [double knockout (dko)] have muscle degeneration similar to mdx mice, but they display clinical features similar to DMD patients. Dko limb muscles also lack postsynaptic membrane folding and display fiber-type abnormalities including an abundance of phenotypically oxidative muscle fibers. Extraocular muscles, which are spared in mdx mice, show a significant pathology in dko mice. In this study, microarray analysis was used to characterize gene expression differences between mdx and dko tibialis anterior and extraocular skeletal muscles in an effort to understand the phenotypic differences between these two dystrophic mouse models. Analysis of gene expression differences showed that upregulation of slow muscle genes specifically characterizes dko limb muscle and suggests that upregulation of these genes may directly account for the more severe phenotype of dko mice. To investigate whether any upregulation of slow genes is retained in vitro, independent of postsynaptic membrane abnormalities, we derived mdx and dko primary myogenic cultures and analyzed the expression of Myh7 and Myl2. Real-time reverse transcriptase-polymerase chain reaction analysis demonstrates that transcription of these slow genes is also upregulated in dko vs mdx myotubes. This data suggests that at least part of the fiber-type abnormality is due directly to the combined absence of utrophin and dystrophin and is not an indirect effect of the postsynaptic membrane abnormalities.


The Journal of Experimental Biology | 2003

Postnatal suppression of myomesin, muscle creatine kinase and the M-line in rat extraocular muscle

John D. Porter; Anita P. Merriam; Bendi Gong; Sriram Kasturi; Xiaohua Zhou; Kurt F. Hauser; Francisco H. Andrade; Georgiana Cheng

SUMMARY The M-line and its associated creatine kinase (CK) M-isoform (CK-M) are ubiquitous features of skeletal and cardiac muscle. The M-line maintains myosin myofilaments in register, links the contractile apparatus to the cytoskeleton for external force transfer and localizes CK-based energy storage and transfer to the site of highest ATP demand. We establish here that the muscle group responsible for movements of the eye, extraocular muscle (EOM), is divergent from other striated muscles in lacking both an M-line and its associated CK-M. Although an M-line forms during myogenesis, both in vivo and in vitro, it is actively repressed after birth. Transcripts of the major M-line structural proteins, myomesin 1 and myomesin 2, follow the same pattern of postnatal downregulation, while the embryonic heart-specific EH-myomesin 1 transcript is expressed early and retained in adult eye muscle. By immunocytochemistry, myomesin protein is absent from adult EOM sarcomeres. M-line suppression does not occur in organotypic co-culture with oculomotor motoneurons, suggesting that the mechanism for suppression may lie in muscle group-specific activation or workload patterns experienced only in vivo. The M-line is, however, still lost in dark-reared rats, despite the developmental delay this paradigm produces in the visuomotor system and EOMs. EOM was low in all CK isoform transcripts except for the sarcomeric mitochondrial (Ckmt2) isoform. Total CK enzyme activity of EOM was one-third that of hindlimb muscle. These findings are singularly unique among fast-twitch skeletal muscles. Since EOM exhibits isoform diversity for other sarcomeric proteins, the M-line/CK-M divergence probably represents a key physiological adaptation for the unique energetics and functional demands placed on this muscle group in voluntary and reflexive eye movements.


Annals of the New York Academy of Sciences | 2002

Extraocular Muscle Gene Expression and Function after Dark Rearing

Francisco H. Andrade; Anita P. Merriam; John D. Porter

The development of the visual system includes a postnatal time window, the “critical period” during which normal sensory input is needed to establish the adult phenotype. The final effectors of the ocular motor system, the extraocular muscles (EOMs), depend also on postnatal sensory experience for the induction of the adult myosin expression pattern and calcium handling systems. The mechanisms regulating this period of postnatal EOM development, and the functional properties dependent on it, remain incompletely understood. For this study, we tested the hypothesis that absence of visual stimulation during the postnatal critical period would alter gene expression patterns and the contractile function of rat EOMs.

Collaboration


Dive into the Anita P. Merriam's collaboration.

Top Co-Authors

Avatar

John D. Porter

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Sangeeta Khanna

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Bendi Gong

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Patrick Leahy

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Henry J. Kaminski

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Wei Guo

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Xiaohua Zhou

University Hospitals of Cleveland

View shared research outputs
Top Co-Authors

Avatar

Chelliah R. Richmonds

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge