Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Development ; 128(13): 2537-44, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11493570

ABSTRACT

During embryonic development, and before functional innervation, a highly stereotypic pattern of slow- and fast-contracting primary muscle fibers is established within individual muscles of the limbs, from distinct populations of myoblasts. A difference between the fiber-type pattern found within chicken and quail pectoral muscles was exploited to investigate the contributions of somite-derived myogenic precursors and lateral plate-derived mesenchymal stroma to the establishment of muscle fiber-type patterns. Chimeric chicken/quail embryos were constructed by reciprocal transplantation of somites or lateral plate mesoderm at stages prior to muscle formation. Muscle fibers derived from quail myogenic precursors that had migrated into chicken stroma showed a quail pattern of mixed fast- and slow-contracting muscle fibers. Conversely, chicken myogenic precursors that had migrated into quail stroma showed a chicken pattern of nearly exclusive fast muscle fiber formation. These results demonstrate in vivo an intrinsic commitment to fiber-type on the part of the myoblast, independent of extrinsic signals it receives from the mesenchymal stroma in which it differentiates.


Subject(s)
Mesoderm/physiology , Muscle Fibers, Fast-Twitch/cytology , Muscle Fibers, Slow-Twitch/cytology , Signal Transduction , Stem Cells/cytology , Animals , Cell Differentiation , Chick Embryo , Muscles/cytology , Muscles/embryology , Quail
2.
Breast J ; 7(2): 131-7, 2001.
Article in English | MEDLINE | ID: mdl-11328324

ABSTRACT

A retrospective analysis of the treatment of locally advanced breast cancer (LABC) was undertaken at Stanford Medical Center to assess the outcome of patients who did not undergo surgical removal of their tumors. Between 1981 and 1998, 64 patients with locally advanced breast cancer were treated with induction chemotherapy, radiation with or without breast surgery, and additional chemotherapy. Sixty-two (97%) patients received cyclophosphamide, doxorubicin, and 5-fluorouracil (CAF) induction chemotherapy. Induction chemotherapy was followed by local radiotherapy in 59 (92%) patients. Based on the clinical response to chemotherapy and patient preference, 44 (69%) patients received no local breast surgery. Radiotherapy was followed by an additional, non-doxorubicin-containing chemotherapy in all patients. The mean age of patients was 49 years. Of the 65 locally advanced breast cancers in 64 patients, 26 (41%) were stage IIIA, 35 (55%) were stage IIIB, and 4 (6%) were stage IV (supraclavicular lymph nodes only). Response to induction chemotherapy was seen in 59 patients (92%), with 29 (45%) achieving a complete clinical response and 30 (47%) a partial clinical response. With a mean follow-up of 51 months (range 7-187 months), 43 patients (67.2%) have no evidence of recurrent disease. Eight (12.5%) have recurred locally, and 21 (32.8%) have recurred with distant metastasis. Actuarial 5-year survival is 75%, disease-free survival is 58%, and local control rate is 87.5%. These data indicate that the routine inclusion of breast surgery in a combined modality treatment program for LABC does not appear necessary for the majority of patients who experience a response to induction chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/radiotherapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/mortality , Breast Neoplasms/surgery , Carcinoma, Ductal, Breast/mortality , Carcinoma, Ductal, Breast/surgery , Cyclophosphamide/administration & dosage , Disease-Free Survival , Doxorubicin/administration & dosage , Drug Administration Schedule , Female , Fluorouracil/administration & dosage , Humans , Mastectomy/statistics & numerical data , Middle Aged , Radiotherapy, Adjuvant , Retrospective Studies , Survival Analysis , Treatment Outcome , Unnecessary Procedures
3.
J Biol Chem ; 276(31): 28835-41, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11382777

ABSTRACT

The slow myosin heavy chain 3 gene (slow MyHC3) is restricted in its expression to the atrial chambers of the heart. Understanding its regulation provides a basis for determination of the mechanisms controlling chamber-specific gene expression in heart development. The observed chamber distribution results from repression of slow MyHC3 gene expression in the ventricles. A binding site, the vitamin D response element (VDRE), for a heterodimer of vitamin D receptor (VDR) and retinoic X receptor alpha (RXR alpha) within the slow MyHC3 promoter mediates chamber-specific expression of the gene. Irx4, an Iroquois family homeobox gene whose expression is restricted to the ventricular chambers at all stages of development, inhibits AMHC1, the chick homolog of quail slow MyHC3, gene expression within developing ventricles. Repression of the slow MyHC3 gene in ventricular cardiomyocytes by Irx4 requires the VDRE. Unlike VDR and RXR alpha, Irx4 does not bind directly to the VDRE. Instead two-hybrid and co-immunoprecipitation assays show that Irx4 interacts with the RXR alpha component of the VDR/RXR alpha heterodimer and that the amino terminus of the Irx4 protein is required for its inhibitory action. These observations indicate that the mechanism of atrial chamber-specific expression requires the formation of an inhibitory protein complex composed of VDR, RXR alpha, and Irx4 that binds at the VDRE inhibiting slow MyHC3 expression in the ventricles.


Subject(s)
Atrial Myosins , Avian Proteins , Gene Expression Regulation/physiology , Homeodomain Proteins/metabolism , Myocardium/metabolism , Myosin Heavy Chains/genetics , Promoter Regions, Genetic , Receptors, Calcitriol/metabolism , Receptors, Retinoic Acid/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Cells, Cultured , Chick Embryo , Cloning, Molecular , DNA-Binding Proteins/metabolism , Gene Deletion , Heart Atria/embryology , Heart Atria/metabolism , Heart Ventricles/embryology , Heart Ventricles/metabolism , Homeodomain Proteins/genetics , Myosins/genetics , Polymerase Chain Reaction , Protein Biosynthesis , Quail , Receptors, Calcitriol/genetics , Receptors, Retinoic Acid/genetics , Retinoid X Receptors , Transcription Factors/genetics
5.
Dev Dyn ; 219(3): 304-21, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11066088

ABSTRACT

Much of our understanding of early vertebrate embryogenesis derives from experimental work done with the chick embryo. Studies of the avian somite have played a key role in elucidating the developmental history of this important structure, the source of most muscle and bone in the organism. Here we review the development of the avian somite including morphological and molecular data on the origin of paraxial mesoderm, maturation of the segmental plate, specification and formation of somite compartments, and somite cell differentiation into cartilage and skeletal muscle.


Subject(s)
Somites/cytology , Animals , Body Patterning , Cartilage/embryology , Cell Differentiation , Cell Division , Chick Embryo , Gene Expression Regulation, Developmental , In Situ Hybridization , Mesoderm/cytology , Microscopy, Electron, Scanning , Molecular Biology , Muscle, Skeletal/embryology , Signal Transduction , Somites/metabolism , Terminology as Topic
6.
Anat Embryol (Berl) ; 200(3): 239-52, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10463340

ABSTRACT

Primary skeletal muscle fibers first form in the segmented portions of paraxial mesoderm called somites. Although the neural tube and notochord are recognized as crucial in patterning myogenic cell lineages during avian and mammalian somitic myogenesis, the source, identities, and actions of the signals governing this process remain controversial. It has been shown that signals emanating from the ventral neural tube and/or notochord alone or Shh alone serve to activate MyoD expression in somites. However, beyond a role in initiating MyoD expression, little is known about the effects of Shh on primary muscle fiber formation in somites of higher vertebrates. The studies reported here investigate how the ventral neural tube promotes myogenesis and compare the effects of the ventral neural tube with those of purified Shh protein on fiber formation in somites. We show that purified Shh protein mimics actions of the ventral neural tube on somites including initiation of muscle fiber formation, enhancement of numbers of primary muscle fibers, and particularly, the formation of primary fibers that express slow myosin. There is a marked increase in slow myosin expression in fibers in response to Shh as somites mature. The effects of ventral neural tube on fiber formation can be blocked by disrupting the Shh signaling pathway by increasing the activity of somitic cyclic AMP-dependent protein kinase A. Furthermore, it was demonstrated that apoptosis is a dominant fate of somite cells, but not somitic muscle fibers, when cultured in the absence of the neural tube, and that application of Shh protein to somites reduced apoptosis. The block to apoptosis by Shh is a manifestation of the maturity of the somite with a progressive increase in the block as somites are displaced rostrally from somite III forward. We conclude that purified Shh protein in mimicking the effects of the ventral neural tube on segmented mesoderm can exert pleiotropic effects during primary myogenesis, including: control of the proliferative expansion of myogenic progenitor cells, antagonism of cell death pathways within the precursors to muscle fibers, and during the crucial process of primary myogenesis, can exert an effect on diversification of muscle fiber types.


Subject(s)
Apoptosis , Embryonic Induction/physiology , Muscle Fibers, Slow-Twitch/cytology , Muscle, Skeletal/embryology , Proteins/pharmacology , Trans-Activators , Animals , Chick Embryo , Hedgehog Proteins , Mitogens/pharmacology , Somites , Spinal Cord/embryology
7.
Development ; 126(12): 2677-87, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10331979

ABSTRACT

To study the specification of inflow structures in the heart we generated transgenic animals harboring the human alkaline phosphatase (HAP) gene driven by the proximal 840 bp of a quail SMyHC3 promoter. In transgenic mice, the SMyHC3-HAP reporter was expressed in posterior heart precursors at 8.25 dpc, in sinus venosa and in the atrium at 8.5 and 9.0 dpc, and in the atria from 10.5 dpc onwards. SMyHC3-HAP transgene expression overlapped synthesis and endogenous response to retinoic acid (RA) in the heart, as determined by antibodies directed against a key RA synthetic enzyme and by staining of RAREhsplacZ transgenic animals. A single pulse of all-trans RA administered to pregnant mice at 7.5, but not after 8.5, dpc induced cardiac dismorphology, ranging from complete absence of outflow tract and ventricles to hearts with reduced ventricles expressing both SMyHC3-HAP and ventricular markers. Blockade of RA synthesis with disulfiram inhibited RA-induced transcription and produced hearts lacking the atrial chamber. This study defines a novel marker for atrial-restricted transcription in the developing mouse heart. It also suggests that atrial-specific gene expression is controlled by localized synthesis of RA, and that exclusion of RA from ventricular precursors is essential for correct specification of the ventricles.


Subject(s)
Alkaline Phosphatase/genetics , Heart Defects, Congenital/chemically induced , Heart/embryology , Tretinoin/metabolism , Alkaline Phosphatase/drug effects , Animals , Disulfiram/pharmacology , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Genetic Markers , Heart Atria/embryology , Heart Atria/metabolism , Heart Defects, Congenital/genetics , Heart Ventricles/embryology , Heart Ventricles/metabolism , Humans , Mice , Mice, Transgenic , Pregnancy , Promoter Regions, Genetic , Response Elements , Signal Transduction , Transcription, Genetic , Transgenes , Tretinoin/pharmacology
8.
Mol Cell Biol ; 18(10): 6023-34, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9742119

ABSTRACT

We have used the slow myosin heavy chain (MyHC) 3 gene to study the molecular mechanisms that control atrial chamber-specific gene expression. Initially, slow MyHC 3 is uniformly expressed throughout the tubular heart of the quail embryo. As cardiac development proceeds, an anterior-posterior gradient of slow MyHC 3 expression develops, culminating in atrial chamber-restricted expression of this gene following chamberization. Two cis elements within the slow MyHC 3 gene promoter, a GATA-binding motif and a vitamin D receptor (VDR)-like binding motif, control chamber-specific expression. The GATA element of the slow MyHC 3 is sufficient for expression of a heterologous reporter gene in both atrial and ventricular cardiomyocytes, and expression of GATA-4, but not Nkx2-5 or myocyte enhancer factor 2C, activates reporter gene expression in fibroblasts. Equivalent levels of GATA-binding activity were found in extracts of atrial and ventricular cardiomyocytes from embryonic chamberized hearts. These observations suggest that GATA factors positively regulate slow MyHC 3 gene expression throughout the tubular heart and subsequently in the atria. In contrast, an inhibitory activity, operating through the VDR-like element, increased in ventricular cardiomyocytes during the transition of the heart from a tubular to a chambered structure. Overexpression of the VDR, acting via the VDR-like element, duplicates the inhibitory activity in ventricular but not in atrial cardiomyocytes. These data suggest that atrial chamber-specific expression of the slow MyHC 3 gene is achieved through the VDR-like inhibitory element in ventricular cardiomyocytes at the time distinct atrial and ventricular chambers form.


Subject(s)
Gene Expression Regulation, Developmental , Heart Atria/metabolism , Myosin Heavy Chains/genetics , Receptors, Calcitriol/metabolism , Animals , Base Sequence , Binding Sites , Cells, Cultured , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , GATA4 Transcription Factor , Heart/embryology , Heart Atria/cytology , Heart Ventricles/metabolism , Molecular Sequence Data , Morphogenesis , Myosin Heavy Chains/metabolism , Promoter Regions, Genetic , Quail , Receptors, Retinoic Acid/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics
9.
Dev Biol ; 188(1): 167-80, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9245520

ABSTRACT

Skeletal muscle fibers express members of the myosin heavy chain (MyHC) gene family in a fiber-type-specific manner. In avian skeletal muscle it is the expression of the slow MyHC isoforms that most clearly distinguishes slow- from fast-contracting fiber types. Two hypotheses have been proposed to explain fiber-type-specific expression of distinct MyHC genes during development-an intrinsic mechanism based on the formation of different myogenic lineage(s) and an extrinsic, innervation-dependent mechanism. We developed a cell culture model system in which both mechanisms were evaluated during fetal muscle development. Myoblasts isolated from prospective fast (pectoralis major) or slow (medial adductor) fetal chick muscles formed muscle fibers in cell culture, none of which expressed slow MyHC genes. By contrast, when muscle fibers formed from myoblasts derived from the slow muscle were cocultured with neural tube, the muscle fibers expressed a slow MyHC gene, while muscle fibers formed from myoblasts of fast muscle origin continued to express only fast MyHC. Motor endplates formed on the fibers derived from myoblasts of both fast and slow muscle origin in cocultures, and slow MyHC gene expression did not occur when neuromuscular transmission or depolarization was blocked. We have cloned the slow MyHC gene that is expressed in response to innervation and identified it as the slow MyHC 2 gene, the predominant adult slow isoform. cDNAs encoding portions of the three slow myosin heavy chain genes (MyHC1, slow MyHC 2, and slow MyHC 3) were isolated. Only slow MyHC 2 mRNA was demonstrated to be abundant in the cocultures of neural tube and muscle fibers derived from myoblasts of slow muscle origin. Thus, expression of the slow MyHC 2 gene in this in vitro system indicates that formation of slow muscle fiber types is dependent on both myoblast lineage (intrinsic mechanisms) and innervation (extrinsic mechanisms), and suggests neither mechanism alone is sufficient to explain formation of muscle fibers of different types during fetal development.


Subject(s)
Gene Expression Regulation, Developmental , Muscle Fibers, Fast-Twitch/cytology , Muscle Fibers, Slow-Twitch/cytology , Myosin Heavy Chains/genetics , Neurons/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Lineage , Cells, Cultured , Chick Embryo , Cloning, Molecular , Coculture Techniques , DNA, Complementary , Molecular Sequence Data , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Myosin Heavy Chains/immunology , Neuromuscular Blocking Agents/pharmacology , Receptors, Cholinergic/analysis , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Spinal Cord/cytology , Synaptic Transmission , Tetrodotoxin/pharmacology
11.
J Biol Chem ; 271(33): 19836-45, 1996 Aug 16.
Article in English | MEDLINE | ID: mdl-8702693

ABSTRACT

The quail slow myosin heavy chain 3 (slow MyHC 3) gene is expressed in the developing heart and in slow muscles of the developing limb. It is first expressed in the pulsatile cardiac tube in the embryo, and as the heart chamberizes its expression becomes restricted to the atria. To identify regulatory elements responsible for atrial-specific expression, the 5' upstream region of slow MyHC 3 gene was investigated. An atrial regulatory domain (ARD1) between -840 and -680 acts as an atrial cell-specific enhancer in primary cardiocyte cultures. ARD1 also specifies atrial-specific expression in vivo when the ARD1/heterologous promoter was introduced into developing chick embryos by a replication-competent retroviral vector. ARD1 is the first atrial cell-specific enhancer to be identified. Fine deletion and mutation analysis within ARD1 defined a 40-base pair vitamin D3 receptor-like element that controls atrial cell-specific expression of the slow MyHC 3 gene by inhibiting its expression in ventricular cardiocytes.


Subject(s)
Heart Atria/embryology , Myosin Heavy Chains/metabolism , Animals , Base Sequence , Cells, Cultured , Chick Embryo , Coturnix , DNA Primers/chemistry , Enhancer Elements, Genetic , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Developmental , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Messenger/genetics , Sequence Deletion
12.
J Biol Chem ; 271(29): 17047-56, 1996 Jul 19.
Article in English | MEDLINE | ID: mdl-8663323

ABSTRACT

We have isolated and begun characterization of the quail slow myosin heavy chain (MyHC) 3 gene, the first reported avian slow MyHC gene. Expression of slow MyHC 3 in skeletal muscle is restricted to the embryonic period of development, when the fiber pattern of future fast and slow muscle is established. In embryonic hindlimb development, slow MyHC 3 gene expression coincides with slow muscle fiber formation as distinguished by slow MyHC-specific antibody staining. In addition to expression in embryonic appendicular muscle, slow MyHC 3 is expressed continuously in the atria. Transfection of slow MyHC 3 promoter-reporter constructs into embryonic myoblasts that form slow MyHC-expressing fibers identified two regions regulating expression of this gene in skeletal muscle. The proximal promoter, containing potential muscle-specific regulatory motifs, permits expression of a reporter gene in embryonic slow muscle fibers, while a distal element, located greater than 2600 base pairs upstream, further enhances expression 3-fold. The slow muscle fiber-restricted expression of slow MyHC 3 during embryonic development, and expression of slow MyHC 3 promoter-reporter constructs in embryonic muscle fibers in vitro, makes this gene a useful marker to study the mechanism establishing the slow fiber lineage in the embryo.


Subject(s)
Embryo, Nonmammalian/physiology , Gene Expression Regulation, Developmental , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Slow-Twitch/physiology , Muscle, Skeletal/embryology , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Amino Acid Sequence , Animals , Base Sequence , Chick Embryo , Chickens , Cloning, Molecular , Exons , Genomic Library , Heart/embryology , In Situ Hybridization , Molecular Sequence Data , Muscle, Skeletal/metabolism , Myocardium/metabolism , Oligodeoxyribonucleotides , Promoter Regions, Genetic , Quail , RNA, Messenger/biosynthesis , Rats , Sequence Homology, Amino Acid , Transcription, Genetic
13.
Cancer ; 76(2): 259-67, 1995 Jul 15.
Article in English | MEDLINE | ID: mdl-8625101

ABSTRACT

BACKGROUND: The impact of the surgical margin status on long-term local control rates for breast cancer in women treated with lumpectomy and radiation therapy is unclear. METHODS: The records of 289 women with 303 invasive breast cancers who were treated with lumpectomy and radiation therapy from 1972 to 1992 were reviewed. The surgical margin was classified as positive (transecting the inked margin), close (less than or equal to 2 mm from the margin), negative, or indeterminate, based on the initial biopsy findings and reexcision specimens, as appropriate. Various clinical and pathologic factors were analyzed as potential prognostic factors for local recurrence in addition to the margin status, including T classification, N classification, age, histologic features, and use of adjuvant therapy. The mean follow-up was 6.25 years. RESULTS: The actuarial probability of freedom from local recurrence for the entire group of patients at 5 and 10 years was 94% and 87%, respectively. The actuarial probability of local control at 10 years was 98% for those patients with negative surgical margins versus 82% for all others (P = 0.007). The local control rate at 10 years was 97% for patients who underwent reexcision and 84% for those who did not. Reexcision appears to convey a local control benefit for those patients with close, indeterminate, or positive initial margins, when negative final margins are attained (P = 0.0001). Final margin status was the most significant determinant of local recurrence rates in univariate analysis. By multivariate analysis, the final margin status and use of adjuvant chemotherapy were significant prognostic factors. CONCLUSIONS: The attainment of negative surgical margins, initially or at the time of reexcision, is the most significant predictor of local control after breast-conserving treatment with lumpectomy and radiation therapy.


Subject(s)
Breast Neoplasms/radiotherapy , Breast Neoplasms/surgery , Carcinoma/surgery , Neoplasm Recurrence, Local/prevention & control , Adult , Aged , Breast Neoplasms/pathology , Carcinoma/pathology , Carcinoma/radiotherapy , Combined Modality Therapy , Female , Humans , Mastectomy, Segmental , Middle Aged , Multivariate Analysis , Neoplasm Metastasis , Survival Analysis
14.
Dev Biol ; 169(1): 96-108, 1995 May.
Article in English | MEDLINE | ID: mdl-7750661

ABSTRACT

Specification of myogenesis in early chicken somites and segmental plate was studied using transfilter explant cultures to determine if myogenic specification by axial structures is mediated by cell-cell contact. Formation of muscle fibers that express myosin heavy chain was assessed in somites transfilter from neural tube and notochord. Either the neural tube or the notochord from early chick embryos (ED 2) induces myogenesis in unspecified somites (Hamburger and Hamilton (HH) stages 11-14) when these tissues are separated by a 0.2- or a 0.05-micron pore filter. The ventral neural tube is found to be a strong inducer of myogenesis, while the dorsal neural tube is found to have low inducing activity. The reduced myogenic inducing activity of the dorsal neural tube is associated with an activity that inhibits myogenic differentiation in specified somites, somites containing cells already committed to myogenesis. Only 6-8 hr of transfilter exposure to neural tube is required to initiate somite myogenesis that is sustained in the absence of neural tube. Other tissues such as specified somites, heart, or ectoderm from the same aged embryo do not induce myogenesis in unspecified somites. Removal of the prospective floor plate of the caudal neural plate at HH stage 8 or 9 does not eliminate the inducing activity of the neurectoderm on unspecified somites (HH stage 11-14). Recombination of somites with neural tube or spinal cord from progressively older embryos (ED 4-20) showed myogenesis inducing activity at all ages, though the activity waned as development proceeded. Paraxial mesoderm need not segment into somites to respond to the inducing activity of the neural tube. We conclude that induction of myogenesis in somites does not require cell-cell contact between either the neural tube or the notochord and therefore induction is mediated by diffusible factor(s); that the inducing activity is localized principally to the notochord and ventral half of the neural tube, but not necessarily to the floor plate; that the myogenic response is specific to neural tube and notochord; and that the dorsal neural tube weakly induces myogenesis and contains an activity that inhibits myogenic cells from differentiating.


Subject(s)
Embryonic Induction , Muscles/embryology , Animals , Chick Embryo , Immunohistochemistry , In Vitro Techniques , Mesoderm , Models, Biological , Nervous System/embryology , Notochord/embryology , Time Factors
15.
Exp Cell Res ; 216(2): 431-42, 1995 Feb.
Article in English | MEDLINE | ID: mdl-7843288

ABSTRACT

Myoblasts from embryonic, fetal, and adult quail and chick muscles were transplanted into limb buds of chick embryos to determine if myoblasts can form muscle fibers in heterochronic limbs and to define the conditions that affect the ability of transplanted cells to populate newly developing limb musculature. Myoblasts from each developmental stage were either freshly isolated and transplanted or were cultured prior to transplantation into limb buds of 4- to 5-day (ED4-5) chick embryos. Transplanted myoblasts, regardless of the age of the donor from which they were derived, formed muscle fibers within embryonic limb muscles. Transplanted cloned myoblasts formed muscle fibers, although there was little evidence that the number of transplanted myoblasts significantly increased following transplantation or that they migrated any distance from the site of injection. The fibers that formed from transplanted clonal myoblasts often did not persist in the host limb muscles until ED10. Diminished fiber formation from myoblasts transplanted into host limbs was observed whether myoblasts were cloned or cultured at high density. However, when freshly isolated myoblasts were transplanted, the fibers they formed were numerous, widely dispersed within the limb musculature, and persisted in the muscles until at least ED10. These results indicate that transplanted myoblasts of embryonic, fetal, and adult origin are capable of forming fibers during early limb muscle formation. They also indicate that even in an embryonic chick limb where proliferation of endogenous myoblasts and muscle fiber formation is rapidly progressing, myoblasts that are cultured in vitro do not substantially contribute to long-term muscle fiber formation after they are transplanted into developing limbs. However, when the same myoblasts are freshly isolated and transplanted without prior cell culture, substantial numbers of fibers form and persist after transplantation into developing limbs. Thus, these studies demonstrate that the extent to which transplanted myoblasts fuse to form fibers which persist in host musculature depends upon whether donor myoblasts are freshly isolated or maintained in vitro prior to injection.


Subject(s)
Cell Transplantation , Extremities/embryology , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/cytology , Alcohol Dehydrogenase/analysis , Alcohol Dehydrogenase/genetics , Animals , Cell Division/drug effects , Cell Fusion , Cells, Cultured , Chick Embryo , Chickens , Clone Cells , Coturnix/embryology , Extremities/physiology , Fibroblast Growth Factor 2/pharmacology , Genes, Reporter/genetics , Muscle, Skeletal/chemistry , Myosins/analysis , Transforming Growth Factor beta/pharmacology
18.
Development ; 120(6): 1443-52, 1994 Jun.
Article in English | MEDLINE | ID: mdl-8050355

ABSTRACT

Specification of the myogenic phenotype in somites was examined in the early chick embryo using organotypic explant cultures stained with monoclonal antibodies to myosin heavy chain. It was found that myogenic specification (formation of muscle fibers in explants of somites or segmental plates cultured alone) does not occur until Hamburger and Hamilton stage 11 (12-14 somites). At this stage, only the somites in the rostral half of the embryo are myogenically specified. By Hamburger and Hamilton stage 12 (15-17 somites), the three most caudal somites were not specified to be myogenic while most or all of the more rostral somites are specified to myogenesis. Somites from older embryos (stage 13-15, 18-26 somites) showed the same pattern of myogenic specification--all but the three most caudal somites were specified. We investigated the effects of the axial structures, the notochord and neural tube, on myogenic specification. Both the notochord and neural tube were able to induce myogenesis in unspecified somites. In contrast, the neural tube, but not the notochord, was able to induce myogenesis in explants of segmental plate, a structure which is not myogenic when cultured alone. When explants of specified somites were stained with antibodies to slow or fast MyHC, it was found that myofiber diversity (fast and fast slow fibers) was established very early in development (as early as Hamburger and Hamilton stage 11). We also found fiber diversity in explants of unspecified somites (the three most caudal somites from stage 11 to 15) when they were recombined with notochord or neural tube. We conclude that myogenic specification in the embryo results in diverse fiber types and is an inductive process which is mediated by factors produced by the neural tube and notochord.


Subject(s)
Embryonic Induction/physiology , Mesoderm/physiology , Muscles/embryology , Nervous System/embryology , Notochord/physiology , Animals , Cell Differentiation/physiology , Cells, Cultured , Chick Embryo , Immunohistochemistry , Mesoderm/cytology , Morphogenesis/physiology , Phenotype
19.
Nature ; 362(6416): 165-7, 1993 Mar 11.
Article in English | MEDLINE | ID: mdl-8383807

ABSTRACT

In the limb bud of the 5-day-old avian embryo, when primary muscle fibre formation is beginning and before specific muscles appear, differences in the expression of fast and slow myosin heavy chain genes can be detected among primary fibres of the premuscle masses. Myoblasts that form colonies of fibres of specific types can be isolated from these limb buds. To assess the role of myoblast commitment in specifying fibre types during embryonic development, we cloned myoblasts of specific types from embryonic and adult muscles, transfected them with a reporter gene, and transferred them into developing limb buds. After transfer, cloned myoblasts formed fibres in the limb with the same patterns of myosin heavy chain gene expression as the fibres they formed in cell culture. These results demonstrate that initial skeletal muscle fibre type diversity during avian limb development can originate, in part, from the commitment of distinct myoblast types to the formation of specific fibre types.


Subject(s)
Alcohol Dehydrogenase/genetics , Muscles/cytology , Muscles/transplantation , Myosins/analysis , Myosins/genetics , Alcohol Dehydrogenase/analysis , Animals , Antibodies , Antibodies, Monoclonal , Avian Sarcoma Viruses/genetics , Chick Embryo , Clone Cells , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Fetal Tissue Transplantation , Immunohistochemistry , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL