Network


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

Hotspot


Dive into the research topics where Clay E. Pandorf is active.

Publication


Featured researches published by Clay E. Pandorf.


Frontiers in Physiology | 2013

Alterations in muscle mass and contractile phenotype in response to unloading models: role of transcriptional/pretranslational mechanisms.

Kenneth M. Baldwin; Fadia Haddad; Clay E. Pandorf; Roland R. Roy; V. Reggie Edgerton

Skeletal muscle is the largest organ system in mammalian organisms providing postural control and movement patterns of varying intensity. Through evolution, skeletal muscle fibers have evolved into three phenotype clusters defined as a motor unit which consists of all muscle fibers innervated by a single motoneuron linking varying numbers of fibers of similar phenotype. This fundamental organization of the motor unit reflects the fact that there is a remarkable interdependence of gene regulation between the motoneurons and the muscle mainly via activity-dependent mechanisms. These fiber types can be classified via the primary type of myosin heavy chain (MHC) gene expressed in the motor unit. Four MHC gene encoded proteins have been identified in striated muscle: slow type I MHC and three fast MHC types, IIa, IIx, and IIb. These MHCs dictate the intrinsic contraction speed of the myofiber with the type I generating the slowest and IIb the fastest contractile speed. Over the last ~35 years, a large body of knowledge suggests that altered loading state cause both fiber atrophy/wasting and a slow to fast shift in the contractile phenotype in the target muscle(s). Hence, this review will examine findings from three different animal models of unloading: (1) space flight (SF), i.e., microgravity; (2) hindlimb suspension (HS), a procedure that chronically eliminates weight bearing of the lower limbs; and (3) spinal cord isolation (SI), a surgical procedure that eliminates neural activation of the motoneurons and associated muscles while maintaining neurotrophic motoneuron-muscle connectivity. The collective findings demonstrate: (1) all three models show a similar pattern of fiber atrophy with differences mainly in the magnitude and kinetics of alteration; (2) transcriptional/pretranslational processes play a major role in both the atrophy process and phenotype shifts; and (3) signaling pathways impacting these alterations appear to be similar in each of the models investigated.


American Journal of Physiology-cell Physiology | 2009

Differential epigenetic modifications of histones at the myosin heavy chain genes in fast and slow skeletal muscle fibers and in response to muscle unloading

Clay E. Pandorf; Fadia Haddad; Carola Wright; Paul W. Bodell; Kenneth M. Baldwin

Recent advances in chromatin biology have enhanced our understanding of gene regulation. It is now widely appreciated that gene regulation is dependent upon post-translational modifications to the histones which package genes in the nucleus of cells. Active genes are known to be associated with acetylation of histones (H3ac) and trimethylation of lysine 4 in histone H3 (H3K4me3). Using chromatin immunoprecipitation (ChIP), we examined histone modifications at the myosin heavy chain (MHC) genes expressed in fast vs. slow fiber-type skeletal muscle, and in a model of muscle unloading, which results in a shift to fast MHC gene expression in slow muscles. Both H3ac and H3K4me3 varied directly with the transcriptional activity of the MHC genes in fast fiber-type plantaris and slow fiber-type soleus. During MHC transitions with muscle unloading, histone H3 at the type I MHC becomes de-acetylated in correspondence with down-regulation of that gene, while upregulation of the fast type IIx and IIb MHCs occurs in conjunction with enhanced H3ac in those MHCs. Enrichment of H3K4me3 is also increased at the type IIx and IIb MHCs when these genes are induced with muscle unloading. Downregulation of IIa MHC, however, was not associated with corresponding loss of H3ac or H3K4me3. These observations demonstrate the feasibility of using the ChIP assay to understand the native chromatin environment in adult skeletal muscle, and also suggest that the transcriptional state of types I, IIx and IIb MHC genes are sensitive to histone modifications both in different muscle fiber-types and in response to altered loading states.


Journal of Biological Chemistry | 2006

Dynamics of Myosin Heavy Chain Gene Regulation in Slow Skeletal Muscle ROLE OF NATURAL ANTISENSE RNA

Clay E. Pandorf; Fadia Haddad; Roland R. Roy; Anqi X. Qin; V. Reggie Edgerton; Kenneth M. Baldwin

The evolutionarily conserved order of the skeletal muscle myosin heavy chain (MHC) genes and their close tandem proximity on the same chromosome are intriguing and may be important for their coordinated regulation. We investigated type II MHC gene regulation in slow-type muscle fibers undergoing a slow to fast MHC transformation in response to inactivity, 7 days after spinal cord isolation (SI) in rats. We examined the transcriptional products of both the sense and antisense strands across the IIa-IIx-IIb MHC gene locus. A strand-specific reverse transcription (RT)-PCR approach was utilized to study the expression of the mRNA, the primary transcript (pre-mRNA), the antisense RNA overlapping the MHC genes, and both the intergenic sense and antisense RNAs. Results showed that the mRNA and pre-mRNA of each MHC had a similar response to SI, suggesting regulation of these genes at the transcriptional level. In addition, we detected previously unknown antisense strand transcription that produced natural antisense transcripts (NATs). RT-PCR mapping of the RNA products revealed that the antisense activity resulted in the formation of three major products: aII, xII, and bII NATs (antisense products of the IIa, IIx, and IIb genes, respectively). The aII NAT begins in the IIa-IIx intergenic region in close proximity to the IIx promoter, extends across the 27-kb IIa MHC gene, and continues to the IIa MHC gene promoter. The expression of the aII NAT was significantly up-regulated in muscles after SI, was negatively correlated with IIa MHC gene expression, and was positively correlated with IIx MHC gene expression. The exact role of the aII NAT is not clear; however, it is consistent with the inhibition of IIa MHC gene transcription. In conclusion, NATs may mediate cross-talk between adjacent genes, which may be essential to the coordinated regulation of the skeletal muscle MHC genes during dynamic phenotype shifts.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2009

Calcineurin plays a modulatory role in loading-induced regulation of type I myosin heavy chain gene expression in slow skeletal muscle.

Clay E. Pandorf; Weihua H. Jiang; Anqi X. Qin; Paul W. Bodell; Kenneth M. Baldwin; Fadia Haddad

The role of calcineurin (Cn) in skeletal muscle fiber-type expression has been a subject of great interest because of reports indicating that it controls the slow muscle phenotype. To delineate the role of Cn in phenotype remodeling, particularly its role in driving expression of the type I myosin heavy chain (MHC) gene, we used a novel strategy whereby a profound transition from fast to slow fiber type is induced and examined in the absence and presence of cyclosporin A (CsA), a Cn inhibitor. To induce the fast-to-slow transition, we first subjected rats to 7 days of hindlimb suspension (HS) + thyroid hormone [triiodothyronine (T(3))] to suppress nearly all expression of type I MHC mRNA in the soleus muscle. HS + T(3) was then withdrawn, and rats resumed normal ambulation and thyroid state, during which vehicle or CsA (30 mg x kg(-1) x day(-1)) was administered for 7 or 14 days. The findings demonstrate that, despite significant inhibition of Cn, pre-mRNA, mRNA, and protein abundance of type I MHC increased markedly during reloading relative to HS + T(3) (P < 0.05). Type I MHC expression was, however, attenuated by CsA compared with vehicle treatment. In addition, type IIa and IIx MHC pre-mRNA, mRNA, and relative protein levels were increased in Cn-treated compared with vehicle-treated rats. These findings indicate that Cn has a modulatory role in MHC transcription, rather than a role as a primary regulator of slow MHC gene expression.


American Journal of Physiology-regulatory Integrative and Comparative Physiology | 2012

Regulation of an antisense RNA with the transition of neonatal to IIb myosin heavy chain during postnatal development and hypothyroidism in rat skeletal muscle

Clay E. Pandorf; Weihua Jiang; Anqi X. Qin; Paul W. Bodell; Kenneth M. Baldwin; Fadia Haddad

Postnatal development of fast skeletal muscle is characterized by a transition in expression of myosin heavy chain (MHC) isoforms, from primarily neonatal MHC at birth to primarily IIb MHC in adults, in a tightly coordinated manner. These isoforms are encoded by distinct genes, which are separated by ∼17 kb on rat chromosome 10. The neonatal-to-IIb MHC transition is inhibited by a hypothyroid state. We examined RNA products [mRNA, pre-mRNA, and natural antisense transcript (NAT)] of developmental and adult-expressed MHC genes (embryonic, neonatal, I, IIa, IIx, and IIb) at 2, 10, 20, and 40 days after birth in normal and thyroid-deficient rat neonates treated with propylthiouracil. We found that a long noncoding antisense-oriented RNA transcript, termed bII NAT, is transcribed from a site within the IIb-Neo intergenic region and across most of the IIb MHC gene. NATs have previously been shown to mediate transcriptional repression of sense-oriented counterparts. The bII NAT is transcriptionally regulated during postnatal development and in response to hypothyroidism. Evidence for a regulatory mechanism is suggested by an inverse relationship between IIb MHC and bII NAT in normal and hypothyroid-treated muscle. Neonatal MHC transcription is coordinately expressed with bII NAT. A comparative phylogenetic analysis also suggests that bII NAT-mediated regulation has been a conserved trait of placental mammals for most of the eutherian evolutionary history. The evidence in support of the regulatory model implicates long noncoding antisense RNA as a mechanism to coordinate the transition between neonatal and IIb MHC during postnatal development.


American Journal of Physiology-cell Physiology | 2007

IIx myosin heavy chain promoter regulation cannot be characterized in vivo by direct gene transfer.

Clay E. Pandorf; Fadia Haddad; Anqi X. Qin; Kenneth M. Baldwin


Archive | 2006

Striated Muscle Plasticity: Regulation of the Myosin Heavy Chain Genes

Fadia Haddad; Clay E. Pandorf; Julia M. Giger; Kenneth M. Baldwin


Archive | 2015

suspension on skeletal myosin heavy chain expression Interaction of hyperthyroidism and hindlimb

Anqi Qin; M. Zeng; S. A. McCue; Kenneth M. Baldwin; Julia M. Giger; Paul W. Bodell; Ming Zeng; Fadia Haddad; Clay E. Pandorf; Weihua Jiang; Anqi X. Qin; M. Coenen-Schimke; K. Sreekumaran Nair; Stéphane Walrand; Kevin R. Short; Lydia A. Heemstra; Colleen M. Novak; James A. Levine


Archive | 2015

hindlimb development. II. Expression of MHC isoforms Effects of spaceflight and thyroid deficiency on rat

Kenneth M. Baldwin; F. Haddad; S. A. McCue; P. W. Bodell; L. Qin; Anqi Qin; Stefano Schiaffino; Carlo Reggiani; Clay E. Pandorf; Weihua Jiang; Anqi X. Qin; Paul W. Bodell; Phil Wyrick; M. Dean Palmer; Ronald G. Haller; Benjamin D. Levine; Felix Krainski; Jeffrey L. Hastings; Katja Heinicke; Nadine Romain; Eric Pacini; Peter G. Snell


The FASEB Journal | 2012

The presence and regulation of antisense long non-coding RNA with altered myosin expression in exercising human muscle

Clay E. Pandorf; Fadia Haddad; Tomasz Owerkowicz; Kenneth M. Baldwin; Vincent J. Caiozzo; Gregory R. Adams

Collaboration


Dive into the Clay E. Pandorf's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Fadia Haddad

University of California

View shared research outputs
Top Co-Authors

Avatar

Anqi X. Qin

University of California

View shared research outputs
Top Co-Authors

Avatar

Paul W. Bodell

University of California

View shared research outputs
Top Co-Authors

Avatar

Roland R. Roy

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julia M. Giger

University of California

View shared research outputs
Top Co-Authors

Avatar

S. A. McCue

University of California

View shared research outputs
Top Co-Authors

Avatar

Benjamin D. Levine

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Carola Wright

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge