Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
FASEB J ; 38(9): e23634, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38679876

ABSTRACT

Insulin-like growth factor-I (IGF-I) facilitates mitotic and anabolic actions in all tissues. In skeletal muscle, IGF-I can promote growth and resolution of damage by promoting satellite cell proliferation and differentiation, suppressing inflammation, and enhancing fiber formation. While the most well-characterized form of IGF-I is the mature protein, alternative splicing and post-translational modification complexity lead to several additional forms of IGF-I. Previous studies showed muscle efficiently stores glycosylated pro-IGF-I. However, non-glycosylated forms display more efficient IGF-I receptor activation in vitro, suggesting that the removal of the glycosylated C terminus is a necessary step to enable increased activity. We employed CRISPR-Cas9 gene editing to ablate IGF-I glycosylation sites (2ND) or its cleavage site (3RA) in mice to determine the necessity of glycosylation or cleavage for IGF-I function in postnatal growth and during muscle regeneration. 3RA mice had the highest circulating and muscle IGF-I content, whereas 2ND mice had the lowest levels compared to wild-type mice. After weaning, 4-week-old 2ND mice exhibited higher body and skeletal muscle mass than other strains. However, by 16 weeks of age, muscle and body size differences disappeared. Even though 3RA mice had more IGF-I stored in muscle in homeostatic conditions, regeneration was delayed after cardiotoxin-induced injury, with prolonged necrosis most evident at 5 days post injury (dpi). In contrast, 2ND displayed improved regeneration with reduced necrosis, and greater fiber size and muscle mass at 11 and 21 dpi. Overall, these results demonstrate that while IGF-I glycosylation may be important for storage, cleavage is needed to enable IGF-I to be used for efficient activity in postnatal growth and following acute injury.


Subject(s)
Insulin-Like Growth Factor I , Muscle, Skeletal , Regeneration , Animals , Glycosylation , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/genetics , Muscle, Skeletal/metabolism , Mice , Regeneration/physiology , Mice, Inbred C57BL , Male , Female
2.
Sci Rep ; 13(1): 18916, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37919321

ABSTRACT

Proinsulin Like Growth Factor I (prolGF-I) and myostatin (Mstn) regulate muscle regeneration and mass when intravenously delivered. We tested if chloroplast bioencapsulated forms of these proteins may serve as a non-invasive means of drug delivery through the digestive system. We created tobacco (Nicotiana tabacum) plants carrying GFP-Fc1, proIGF-I-Fc1, and Mstn-Fc1 fusion genes, in which fusion with the immunoglobulin G Fc domain improved both protein stability and absorption in the small intestine. No transplastomic plants were obtained with the Mstn-Fc1 gene, suggesting that the protein is toxic to plant cells. proIGF-I-Fc1 protein levels were too low to enable in vivo testing. However, GFP-Fc1 accumulated at a high level, enabling evaluation of chloroplast-made Fc fusion proteins for oral delivery. Tobacco leaves were lyophilized for testing in a mouse system. We report that the orally administered GFP-Fc1 fusion protein (5.45 µg/g GFP-Fc1) has been taken up by the intestinal epithelium cells, evidenced by confocal microscopy. GFP-Fc1 subsequently entered the circulation where it was detected by ELISA. Data reported here confirm that chloroplast expression and oral administration of lyophilized leaves is a potential delivery system of therapeutic proteins fused with Fc1, with the advantage that the proteins may be stored at room temperature.


Subject(s)
Chloroplasts , Immunoglobulin G , Mice , Animals , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Chloroplasts/genetics , Chloroplasts/metabolism , Nicotiana/genetics , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism
3.
Res Sq ; 2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37546919

ABSTRACT

Proinsulin Like Growth Factor (prolGF1) and myostatin (Mstn) regulate muscle regeneration when intravenously delivered. We set out to test if chloroplast bioencapsulated forms of these proteins may serve as a non-invasive means of drug delivery through the digestive system. We created tobacco (Nicotiana tabacum) plants carrying GFP-Fc1, proIGF-I-Fc1, and Mstn-Fc1 fusion genes, in which fusion with the immunoglobulin G Fc domain improved both protein stability and absorption in the small intestine. No transplastomic plants were obtained with the Mstn-Fc1 gene, suggesting that the protein is toxic to plant cells. proIGF-I-Fc1 protein levels were too law to enable in vivo testing. However, GFP-Fc1 accumulated at a high level, enabling evaluation of chloroplast-made Fc fusion proteins for oral delivery. Tobacco leaves were lyophilized for testing in a mouse system. We report that the orally administered GFP-Fc fusion protein (5.45 µg/g GFP-Fc) has been taken up by the intestinal epithelium cells, evidenced by confocal microscopy. GFP-Fc subsequently entered the circulation where it was detected by ELISA. Data reported here confirm that chloroplast expression and oral administration of lyophilized leaves is a potential delivery system of therapeutic proteins fused with Fc, with the advantage that the proteins may be stored at room temperature.

4.
J Appl Physiol (1985) ; 131(3): 881-894, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34292789

ABSTRACT

Muscle atrophy occurs as a result of prolonged periods of reduced mechanical stimulation associated with injury or disease. The growth hormone/insulin-like growth factor-1 (GH/IGF-1) axis and load sensing pathways can both aid in recovery from disuse through their shared downstream signaling, but their relative contributions to these processes are not fully understood. The goal of this study was to determine whether reduced muscle IGF-1 altered the response to disuse and reloading. Adult male mice with inducible muscle-specific IGF-1 deletion (MID) induced 1 wk before suspension and age-matched controls (CON) were subjected to hindlimb suspension and reloading. Analysis of muscle force, morphology, gene expression, signaling, and tissue weights was performed in nonsuspended (NS) mice, and those suspended for 7 days or reloaded following suspension for 3, 7, and 14 days. MID mice displayed diminished IGF-1 protein levels and muscle atrophy before suspension. Muscles from suspended CON mice displayed a similar extent of atrophy and depletion of IGF-1, yet combined loss of load and IGF-1 was not additive with respect to muscle mass. In contrast, soleus force generation capacity was diminished to the greatest extent when both suspension and IGF-1 deletion occurred. Recovery of mass, force, and gene expression patterns following suspension were similar in CON and MID mice, even though IGF-1 levels increased only in muscles from CON mice. Diminished strength in disuse atrophy is exacerbated with the loss of muscle IGF-1 production, whereas recovery of mass and strength upon reloading can occur even IGF-1 is low.NEW & NOTEWORTHY A mouse model with skeletal muscle-specific inducible deletion of Igf1 was used to address the importance of this growth factor for the consequences of disuse atrophy. Rapid and equivalent loss of IGF-I and mass occurred with deletion or disuse. Decrements in strength were most severe with combined loss of load and IGF-1. Return of mass and strength upon reloading was independent of IGF-1.


Subject(s)
Insulin-Like Growth Factor I , Muscular Disorders, Atrophic , Animals , Hindlimb Suspension , Male , Mice , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Disorders, Atrophic/pathology
5.
Cell Physiol Biochem ; 54(3): 333-353, 2020 Apr 11.
Article in English | MEDLINE | ID: mdl-32275813

ABSTRACT

BACKGROUND/AIMS: Cell migration and extracellular matrix remodeling underlie normal mammalian development and growth as well as pathologic tumor invasion. Skeletal muscle is no exception, where satellite cell migration replenishes nuclear content in damaged tissue and extracellular matrix reforms during regeneration. A key set of enzymes that regulate these processes are matrix metalloproteinases (MMP)s. The collagenase MMP-13 is transiently upregulated during muscle regeneration, but its contribution to damage resolution is unknown. The purpose of this work was to examine the importance of MMP-13 in muscle regeneration and growth in vivo and to delineate a satellite cell specific role for this collagenase. METHODS: Mice with total and satellite cell specific Mmp13 deletion were utilized to determine the importance of MMP-13 for postnatal growth, regeneration after acute injury, and in chronic injury from a genetic cross with dystrophic (mdx) mice. We also evaluated insulin-like growth factor 1 (IGF-1) mediated hypertrophy in the presence and absence of MMP-13. We employed live-cell imaging and 3D migration measurements on primary myoblasts obtained from these animals. Outcome measures included muscle morphology and function. RESULTS: Under basal conditions, Mmp13-/- mice did not exhibit histological or functional deficits in muscle. However, following acute injury, regeneration was impaired at 11 and 14 days post injury. Muscle hypertrophy caused by increased IGF-1 was blunted with minimal satellite cell incorporation in the absence of MMP-13. Mmp13-/- primary myoblasts displayed reduced migratory capacity in 2D and 3D, while maintaining normal proliferation and differentiation. Satellite cell specific deletion of MMP-13 recapitulated the effects of global MMP-13 ablation on muscle regeneration, growth and myoblast movement. CONCLUSION: These results show that satellite cells provide an essential autocrine source of MMP-13, which not only regulates their migration, but also supports postnatal growth and resolution of acute damage.


Subject(s)
Cell Movement/genetics , Matrix Metalloproteinase 13/metabolism , Muscle, Skeletal/enzymology , Regeneration/genetics , Satellite Cells, Skeletal Muscle/enzymology , Animals , Cell Movement/physiology , Extracellular Matrix/enzymology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Male , Matrix Metalloproteinase 13/genetics , Mice , Mice, Inbred mdx , Mice, Knockout , Muscle, Skeletal/injuries , Muscle, Skeletal/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Regeneration/physiology
6.
FASEB J ; 33(1): 181-194, 2019 01.
Article in English | MEDLINE | ID: mdl-29932867

ABSTRACT

Insulin-like growth factors (IGFs) are essential for local skeletal muscle growth and organismal physiology, but these actions are entwined with glucose homeostasis through convergence with insulin signaling. The objective of this work was to determine whether the effects of IGF-I on growth and metabolism could be separated. We generated muscle-specific IGF-I-deficient (MID) mice that afford inducible deletion of Igf1 at any age. After Igf1 deletion at birth or in young adult mice, evaluations of muscle physiology and glucose homeostasis were performed up to 16 wk of age. MID mice generated at birth had lower muscle and circulating IGF-I, decreased muscle and body mass, and impaired muscle force production. Eight-wk-old male MID had heightened insulin levels with trends of elevated fasting glucose. This phenotype progressed to impaired glucose handling and increased fat deposition without significant muscle mass loss at 16 wk of age. The same phenotype emerged in 16-wk-old MID mice induced at 12 wk of age, compounded with heightened muscle fatigability and exercise intolerance. We assert that muscle IGF-I independently modulates anabolism and metabolism in an age-dependent manner, thus positioning muscle IGF-I maintenance to be critical for both muscle growth and metabolic homeostasis.-Vassilakos, G., Lei, H., Yang, Y., Puglise, J., Matheny, M., Durzynska, J., Ozery, M., Bennett, K., Spradlin, R., Bonanno, H., Park, S., Ahima, R. S., Barton, E. R. Deletion of muscle IGF-I transiently impairs growth and progressively disrupts glucose homeostasis in male mice.


Subject(s)
Body Weight , Exercise Tolerance , Glucose/metabolism , Homeostasis , Insulin-Like Growth Factor I/physiology , Muscle, Skeletal/pathology , Animals , Female , Male , Mice , Mice, Knockout , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal , SKP Cullin F-Box Protein Ligases/metabolism , Signal Transduction
7.
FASEB J ; 29(7): 2769-79, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25795455

ABSTRACT

Muscle loading is important for maintaining muscle mass; when load is removed, atrophy is inevitable. However, in clinical situations such as critical care myopathy, masticatory muscles do not lose mass. Thus, their properties may be harnessed to preserve mass. We compared masticatory and appendicular muscles responses to microgravity, using mice aboard the space shuttle Space Transportation System-135. Age- and sex-matched controls remained on the ground. After 13 days of space flight, 1 masseter (MA) and tibialis anterior (TA) were frozen rapidly for biochemical and functional measurements, and the contralateral MA was processed for morphologic measurements. Flight TA muscles exhibited 20 ± 3% decreased muscle mass, 2-fold decreased phosphorylated (P)-Akt, and 4- to 12-fold increased atrogene expression. In contrast, MAs had no significant change in mass but a 3-fold increase in P-focal adhesion kinase, 1.5-fold increase in P-Akt, and 50-90% lower atrogene expression compared with limb muscles, which were unaltered in microgravity. Myofibril force measurements revealed that microgravity caused a 3-fold decrease in specific force and maximal shortening velocity in TA muscles. It is surprising that myofibril-specific force from both control and flight MAs were similar to flight TA muscles, yet power was compromised by 40% following flight. Continued loading in microgravity prevents atrophy, but masticatory muscles have a different set point that mimics disuse atrophy in the appendicular muscle.


Subject(s)
Masticatory Muscles/pathology , Space Flight , Weightlessness/adverse effects , Animals , Biomechanical Phenomena , Female , Gene Expression , Mastication/physiology , Masticatory Muscles/physiopathology , Mice , Mice, Inbred C57BL , Muscle Contraction/physiology , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Myofibrils/pathology , Myofibrils/physiology , SKP Cullin F-Box Protein Ligases/genetics , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Weight-Bearing/physiology
8.
Am J Physiol Cell Physiol ; 305(5): C529-38, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23761625

ABSTRACT

Efficient skeletal muscle repair and regeneration require coordinated remodeling of the extracellular matrix (ECM). Previous reports have indicated that matrix metalloproteinases (MMPs) play the pivotal role in ECM remodeling during muscle regeneration. The goal of the current study was to determine if the interstitial collagenase MMP-13 was involved in the muscle repair process. Using intramuscular cardiotoxin injections to induce acute muscle injury, we found that MMP-13 expression and activity transiently increased during the regeneration process. In addition, in muscles from mdx mice, which exhibit chronic injury, MMP-13 expression and protein levels were elevated. In differentiating C2C12 cells, a murine myoblast cell line, Mmp13 expression was most pronounced after myoblast fusion and during myotube formation. Using pharmacological inhibition of MMP-13 to test whether MMP-13 activity is necessary for the proliferation, differentiation, migration, and fusion of C2C12 cells, we found a dramatic blockade of myoblast migration, as well as a delay in differentiation. In contrast, C2C12 cells with stable overexpression of MMP-13 showed enhanced migration, without affecting myoblast maturation. Taken together, these results support a primary role for MMP-13 in myoblast migration that leads to secondary effects on differentiation.


Subject(s)
Cell Movement/physiology , Matrix Metalloproteinase 13/metabolism , Muscle, Skeletal/enzymology , Myoblasts/enzymology , Regeneration/physiology , Animals , Cell Differentiation , Cells, Cultured , Cobra Cardiotoxin Proteins/toxicity , Gene Expression , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/enzymology , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Myoblasts/cytology , Myoblasts/drug effects
9.
FASEB J ; 26(9): 3691-702, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22649033

ABSTRACT

Insulin-like growth factors (IGFs) are critical for development and growth of skeletal muscles, but because several tissues produce IGFs, it is not clear which source is necessary or sufficient for muscle growth. Because it is critical for production of both IGF-I and IGF-II, we ablated glucose-regulated protein 94 (GRP94) in murine striated muscle to test the necessity of local IGFs for normal muscle growth. These mice exhibited smaller skeletal muscles with diminished IGF contents but with normal contractile function and no apparent endoplasmic reticulum stress response. This result shows that muscles rely on GRP94 primarily to support local production of IGFs, a pool that is necessary for normal muscle growth. In addition, body weights were ∼30% smaller than those of littermate controls, and circulating IGF-I also decreased significantly, yet glucose homeostasis was maintained with little disruption to the growth hormone pathway. The growth defect was complemented on administration of recombinant IGF-I. Thus, unlike liver production of IGF-I, muscle IGF-I is necessary not only locally but also globally for whole-body growth.


Subject(s)
Growth , Membrane Glycoproteins/physiology , Muscle, Skeletal/growth & development , Somatomedins/antagonists & inhibitors , Animals , Blood Glucose/analysis , Cells, Cultured , Immunohistochemistry , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Muscle, Skeletal/physiology , Reverse Transcriptase Polymerase Chain Reaction , Somatomedins/biosynthesis
10.
J Appl Physiol (1985) ; 108(5): 1069-76, 2010 May.
Article in English | MEDLINE | ID: mdl-20133429

ABSTRACT

Insulin-like growth factor I (IGF-I) coordinates proliferation and differentiation in a wide variety of cell types. The igf1 gene not only produces IGF-I, but also generates multiple carboxy-terminal extensions, the E-peptides, through alternative splicing leading to different isoforms. It is not known if the IGF-I isoforms share a common pathway for their actions, or if there are specific actions of each protein. Viral administration of murine IGF-IA, IGF-IB, and mature IGF, which lacked an E-peptide extension, was utilized to identify IGF-I isoform-specific responsive genes in muscles of young growing mice. Microarray analysis revealed responses that were driven by increased IGF-I regardless of the presence of E-peptide, such as Bcl-XL. In contrast, distinct expression patterns were observed after viral delivery of IGF-IA or IGF-IB, which included matrix metalloproteinase 13 (MMP13). Expression of Bcl-XL was prevented when viral administration of the IGF-I isoforms was performed into muscles of MKR mice, which lack functional IGF-I receptors on the muscle fibers. However, MMP13 expression persisted under the same conditions after viral injection of IGF-IB. At 4 mo after viral delivery, expression of IGF-IA or IGF-IB promoted muscle hypertrophy, but viral delivery of mature IGF-I failed to increase muscle mass. These studies provide evidence that local production of IGF-I requires the E-peptides to drive hypertrophy in growing muscle and that both common and unique pathways exist for the IGF-I isoforms to promote biological effects.


Subject(s)
Cell Enlargement , Insulin-Like Growth Factor I/biosynthesis , Muscle Development , Muscle, Skeletal/metabolism , Signal Transduction , Animals , Gene Expression Profiling/methods , Gene Expression Regulation , Gene Transfer Techniques , Genetic Vectors , Hindlimb , Hypertrophy , Insulin-Like Growth Factor I/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Development/genetics , Muscle, Skeletal/growth & development , Muscle, Skeletal/pathology , Oligonucleotide Array Sequence Analysis , Protein Isoforms , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Signal Transduction/genetics , Time Factors
11.
Mol Biol Cell ; 20(17): 3810-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19605562

ABSTRACT

Insulin-like growth factor (IGF)-I is a critical protein for cell development and growth. Alternative splicing of the igf1 gene gives rise to multiple isoforms. In rodents, proIGF-IA and proIGF-IB have different carboxy-terminal extensions called the E-peptides (EA and EB) and upon further posttranslational processing, produce the identical mature IGF-I protein. Rodent EB has been reported to have mitogenic and motogenic effects independent of IGF-I. However, effects of EA or EB on mature IGF-I, or whether proIGF-IA and proIGF-IB have different properties, have not been addressed. To determine whether the presence of EA or EB affected the distribution and stability of mature IGF-I protein, transient transfections of cDNAs encoding murine IGF-IA, IGF-IB, and mature IGF-I were performed in C2C12 cells, a skeletal muscle cell line. IGF-I secretion was measured by enzyme-linked immunosorbent assay of the media, and did not differ between expression of proIGF-IA, proIGF-IB, or mature IGF-I expression. Next, epitope-tagged constructs were transfected to determine cellular distribution of IGF-I, EA, and EB in the cells throughout the culture. IGF-I was detected in significantly fewer nontransfected cells in cultures transfected with mature IGF-I compared with transfection of proIGF-IA or proIGF-IB. These results demonstrate that EA and EB are not required for IGF-I secretion but that they increase cell entry of IGF-I from the media. This study provides evidence that the EA and EB may modulate IGF-I in addition to having independent activity.


Subject(s)
Endocytosis/physiology , Insulin-Like Growth Factor I/metabolism , Peptide Fragments/metabolism , Protein Isoforms/metabolism , Protein Precursors/metabolism , Animals , Cell Line , Humans , Insulin-Like Growth Factor I/genetics , Mice , Peptide Fragments/genetics , Protein Isoforms/genetics , Protein Precursors/genetics , Transfection
12.
Cancer Res ; 67(3): 1221-7, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17283158

ABSTRACT

HIV protease inhibitors (HIV PI) are a class of antiretroviral drugs that are designed to target the viral protease. Unexpectedly, this class of drugs is also reported to have antitumor activity. In this study, we have evaluated the in vitro activity of nelfinavir, a HIV PI, against human melanoma cells. Nelfinavir inhibits the growth of melanoma cell lines at low micromolar concentrations that are clinically attainable. Nelfinavir promotes apoptosis and arrests cell cycle at G(1) phase. Cell cycle arrest is attributed to inhibition of cyclin-dependent kinase 2 (CDK2) and concomitant dephosphorylation of retinoblastoma tumor suppressor. We further show that nelfinavir inhibits CDK2 through proteasome-dependent degradation of Cdc25A phosphatase. Our results suggest that nelfinavir is a promising candidate chemotherapeutic agent for advanced melanoma, for which novel and effective therapies are urgently needed.


Subject(s)
Melanoma/drug therapy , Melanoma/pathology , Nelfinavir/pharmacology , Apoptosis/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/metabolism , G1 Phase/drug effects , HIV Protease Inhibitors/pharmacology , Humans , Melanoma/enzymology , Melanoma/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Retinoblastoma Protein/metabolism , Signal Transduction/drug effects , cdc25 Phosphatases/metabolism
13.
Cancer Biol Ther ; 4(7): 709-15, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15970687

ABSTRACT

Significant growth inhibition and induction of apoptosis by IFN-beta in cancer cells including colorectal cancer cells have been observed. We and others have previously reported the Stat 1 induction of TRAIL is a crucial step in the IFN-beta induced apoptosis pathway. However, when evaluating the sensitivity of a panel of colorectal cancer cell lines, we found no clear correlation between activation of the Jak/Stat signaling pathway and response to interferon. In the present study, we have evaluated the interaction of the PI3k/Akt pathway and IFN-beta induced apoptosis in human colorectal cancer cells. The results demonstrate a correlation between Akt activity, phosphorylation of Bad and resistance to interferon-induced apoptosis in these cells. The association of activation of Akt, phosphorylation of Bad and resistance to IFN-beta-induced apoptosis was further supported by the observation that disruption of the pathway in a more resistant cell line led to sensitization, and expression of an activated Akt in a more sensitive cell line led to increased resistance. Taken together, this data indicates that the PI3/Akt kinase pathway may be an important contributor to IFN-beta sensitivity and resistance in colorectal cancer cells. This data demonstrates a potential pathway by which cells may develop resistance to IFN, and further elucidation of this process may allow us to better target IFN therapy.


Subject(s)
Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Enzyme Activation/drug effects , Interferon Type I/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Apoptosis/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Luciferases , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics , Recombinant Proteins , Tumor Cells, Cultured , bcl-Associated Death Protein/metabolism
14.
Clin Cancer Res ; 10(4): 1535-44, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14977858

ABSTRACT

Preclinical in vitro and in vivo studies have demonstrated synergistic interactions between 5-fluorouracil (5-FU) and type I and II IFNs against human colorectal cancer cells. Despite these activities, randomized human trials have failed to identify a clinical benefit for this combination treatment. These limited clinical results may be secondary to the short half-life of recombinant IFN protein and the increased systemic toxicities of 5-FU/IFN combinations. We have previously reported an adenoviral-mediated IFN-beta gene therapy strategy, which may circumvent the pitfalls of recombinant IFN therapy. However, a dose-dependent toxicity and acute inflammatory response to systemically administered adenovirus vectors may limit the clinical application of this therapy. The combination of adenoviral-mediated IFN-beta gene therapy and 5-FU resulted in tumor regression, apoptosis, and improved survival in an established liver metastases model. These therapeutic effects were observed at a significantly lower vector dose than we had previously reported and with limited toxicity. This approach may allow for an effective clinical application of this therapy and warrants additional investigation.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Fluorouracil/pharmacology , Genetic Therapy , Interferon-beta/genetics , Liver Neoplasms/secondary , Liver Neoplasms/therapy , Adenoviridae/genetics , Animals , Apoptosis , Cell Line, Tumor , Colorectal Neoplasms/mortality , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , In Situ Nick-End Labeling , Inflammation , Interferon-beta/blood , Interferon-beta/metabolism , Liver/metabolism , Liver Neoplasms/mortality , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Time Factors , Treatment Outcome
15.
Cancer Res ; 63(17): 5299-307, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14500361

ABSTRACT

Type I IFNs are known to inhibit tumor cell growth and stimulate the immune system. However, little is known of the mechanism of type I IFN-induced apoptosis in human cancer cells. In this study, we have IFN-beta treatment of a human colorectal cell line (KM12L4) and a resistant clone of this cell line, L4RIFN. We demonstrate the induction of apoptosis in the parent cell line. This process was associated with the induction of the Jak-Stat signaling pathway, induction of the proapoptotic mediator tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), and activation of procaspase-3, -8, -9, and -10. Additionally, we evaluated the role of Stat1 in mediating IFN-beta induction of these proapoptotic signals in a fibrosarcoma cell line (2ftgh) and a Stat1-deficient clone (U3A). Our results demonstrate that IFN-beta induction of apoptosis and the induction of proapoptotic mediator TRAIL is Stat1 dependent. Evaluation of a stable transfectant of the KM12L4 cell line expressing c-FLIP supports the role of TRAIL and the cell-surface death signaling pathways in IFN-beta induction of apoptosis. Studies evaluating the TRAIL promoter indicate induction of TRAIL promoter activity by IFN-beta. These results may represent a novel pathway by which IFN-beta may induce therapeutic effects.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , DNA-Binding Proteins/physiology , Interferon Type I/pharmacology , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/biosynthesis , Trans-Activators/physiology , Tumor Necrosis Factor-alpha/biosynthesis , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/biosynthesis , Caspase Inhibitors , Caspases/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Membrane Glycoproteins/genetics , Promoter Regions, Genetic/drug effects , Recombinant Proteins , STAT1 Transcription Factor , Signal Transduction/drug effects , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...