Entry - *603109 - SMAD FAMILY MEMBER 3; SMAD3 - OMIM
 
* 603109

SMAD FAMILY MEMBER 3; SMAD3


Alternative titles; symbols

MOTHERS AGAINST DECAPENTAPLEGIC, DROSOPHILA, HOMOLOG OF, 3; MADH3
SMA- AND MAD-RELATED PROTEIN 3


HGNC Approved Gene Symbol: SMAD3

Cytogenetic location: 15q22.33     Genomic coordinates (GRCh38): 15:67,065,602-67,195,169 (from NCBI)


Gene-Phenotype Relationships
Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
15q22.33 Loeys-Dietz syndrome 3 613795 AD 3

TEXT

Cloning and Expression

Drosophila Mad is required for signaling by the TGF-beta (e.g., 190180)-related factor decapentaplegic. Zhang et al. (1996) cloned a human cDNA encoding MADH3, a homolog of Drosophila Mad. The deduced 425-amino acid MADH3 protein (GenBank 2522267) is 92% identical to MADH2 (601366).

By searching an expressed sequence tag database with the protein sequences of Mad and Mad homologs, Riggins et al. (1996) isolated human cDNAs encoding MADH3, which they called JV15-2. The C terminus of MADH3 shows significant homology to that of Drosophila Mad.


Gene Structure

Arai et al. (1998) determined the genomic structure of SMAD3, which contains 9 exons.


Mapping

Riggins et al. (1996) mapped the MADH3 gene to 15q21-q22 by somatic cell hybrid analysis and screening of YAC clones.


Gene Function

Zhang et al. (1996) showed that MADH3 and MADH4 (SMAD4; 600993) synergized to induce strong ligand-independent TGF-beta-like responses. MADH3 containing a C-terminal truncation acted as a dominant-negative inhibitor of the normal TGF-beta response. The activity of MADH3 was regulated by the TGF-beta receptors (e.g., 190181), and MADH3 was phosphorylated and associated with the ligand-bound receptor complex. Zhang et al. (1996) stated that these results define MADH3 as an effector of the TGF-beta response.

Zawel et al. (1998) found that human SMAD3 and SMAD4 proteins could specifically recognize an identical 8-bp palindromic sequence (GTCTAGAC). Tandem repeats of this palindrome conferred striking TGF-beta responsiveness to a minimal promoter. This responsiveness was abrogated by targeted deletion of the cellular SMAD4 gene. These results showed that SMAD proteins are involved in the biologic responses to TGF-beta and related ligands.

You and Kruse (2002) studied corneal myofibroblast differentiation and signal transduction induced by the TGFB family members activin A (147290) and bone morphogenetic protein-7 (BMP7; 112267). They found that activin A induced phosphorylation of SMAD2 (601366), and BMP7 induced SMAD1 (601595), both of which were inhibited by follistatin (136470). Transfection with antisense SMAD2/SMAD3 prevented activin-induced expression and accumulation of alpha-smooth muscle actin. The authors concluded that TGFB proteins have different functions in the cornea. Activin A and TGFB1, but not BMP7, are regulators of keratocyte differentiation and might play a role during myofibroblast transdifferentiation. SMAD2/SMAD3 signal transduction appeared to be important in the regulation of muscle-specific genes.

SMAD3 is a direct mediator of transcriptional activation by the TGF-beta receptor. Its target genes in epithelial cells include cyclin-dependent kinase (CDK; see 116953) inhibitors that generate a cytostatic response. Chen et al. (2002) defined how, in the same context, SMAD3 can mediate transcriptional repression of the growth-promoting gene MYC (190080). A complex containing SMAD3, the transcription factors E2F4 (600659), E2F5 (600967), and DP1 (189902), and the corepressor p107 (116957) preexists in the cytoplasm. In response to TGF-beta, this complex moves into the nucleus and associates with SMAD4, recognizing a composite SMAD-E2F site on MYC for repression. Previously known as the ultimate recipients of CDK regulatory signals, E2F4/E2F5 and p107 act here as transducers of TGF-beta receptor signals upstream of CDK. SMAD proteins therefore mediate transcriptional activation or repression depending on their associated partners.

TGFB (190180) stimulation leads to phosphorylation and activation of SMAD2 and SMAD3, which form complexes with SMAD4 that accumulate in the nucleus and regulate transcription of target genes. Inman et al. (2002) demonstrated that following TGFB stimulation of epithelial cells, receptors remain active for at least 3 to 4 hours, and continuous receptor activity is required to maintain active SMADs in the nucleus and for TGFB-induced transcription. Continuous nucleocytoplasmic shuttling of the SMADs during active TGFB signaling provides the mechanism whereby the intracellular transducers of the signal continuously monitor receptor activity. These data explain how, at all times, the concentration of active SMADs in the nucleus is directly dictated by the levels of activated receptors in the cytoplasm.

Based upon molecular allelotyping and comparative genomic hybridization studies, chromosome 15q is the likely location of a tumor suppressor gene important in the pathogeneses of sporadic enteropancreatic endocrine tumors and parathyroid adenomas. To determine whether SMAD3 plays a primary role in the development of these tumors, Shattuck et al. (2002) investigated 20 enteropancreatic tumors and 67 parathyroid adenomas for LOH at DNA markers surrounding SMAD3. Twenty percent of enteropancreatic tumors and 24% of parathyroid adenomas showed loss. All 9 coding exons and intron-exon boundaries of the SMAD3 gene were then sequenced in genomic DNA from all 20 enteropancreatic and 25 parathyroid tumors, including every case with LOH. No acquired clonal mutations, insertions, or microdeletions in SMAD3 were detected in any tumors. Because inactivating somatic mutation is the hallmark of an authentic tumor suppressor, SMAD3 is unlikely to function as a classic tumor suppressor gene in the pathogenesis of sporadic parathyroid or enteropancreatic endocrine tumors.

Matsuura et al. (2004) showed that SMAD3 is a major physiologic substrate of the G1 cyclin-dependent kinases CDK4 (123829) and CDK2 (116953). Except for the retinoblastoma protein family, SMAD3 was the only CDK4 substrate demonstrated to that time. Matsuura et al. (2004) mapped CDK4 and CDK2 phosphorylation sites to thr8, thr178, and ser212 in SMAD3. Mutation of the CDK phosphorylation sites increased Smad3 transcriptional activity, leading to higher expression of the CDK inhibitor p15 (600431). Mutation of the CDK phosphorylation sites of Smad3 also increased its ability to downregulate the expression of c-myc. Using Smad3 knockout mouse embryonic fibroblasts and other epithelial cell lines, Matsuura et al. (2004) further showed that Smad3 inhibits cell cycle progression from G1 to S phase and that mutation of the CDK phosphorylation sites in Smad3 increases this ability. They concluded that CDK phosphorylation of SMAD3 inhibits its transcriptional activity and antiproliferative function.

To determine the role of SMAD3 in the pathogenesis of lymphoid neoplasia, Wolfraim et al. (2004) measured SMAD3 mRNA and protein in leukemia cells obtained at diagnosis from 19 children with acute leukemia: 10 with T-cell acute lymphoblastic leukemia (ALL), 7 with pre-B-cell ALL, and 2 with acute nonlymphoblastic leukemia (ANLL). SMAD3 protein was absent in T-cell ALL but present in pre-B-cell ALL and ANLL. No mutations in the SMAD3 gene were identified in T-cell ALL, and SMAD3 mRNA was present in T-cell ALL and normal T cells at similar levels. Wolfraim et al. (2004) concluded that loss of SMAD3 protein is a specific feature of pediatric T-cell lymphoblastic leukemia.

In experiments using mouse muscle, Carlson et al. (2008) found that, in addition to the loss of Notch (190198) activation, old muscle produces excessive TGF-beta (but not myostatin, 601788), which induces unusually high levels of Smad3 in resident satellite cells and interferes with the regenerative capacity. Importantly, endogenous Notch and Smad3 antagonize each other in the control of satellite cell proliferation, such that activation of Notch blocks the TGF-beta-dependent upregulation of the cyclin-dependent kinase (CDK) inhibitors p15, p16 (600160), p21 (116899), and p27 (600778), whereas inhibition of Notch induces them. Furthermore, in muscle stem cells, Notch activity determined the binding of Smad3 to the promoters of these negative regulators of cell cycle progression. Attenuation of TGF-beta/Smad3 in old, injured muscle restored regeneration to satellite cells in vivo. Thus, a balance between endogenous Smad3 and active Notch controls the regenerative competence of muscle stem cells, and deregulation of this balance in the old muscle microniche interferes with regeneration.

Davis et al. (2008) demonstrated that induction of a contractile phenotype in human vascular smooth muscle cells by TGF-beta (190180) and BMPs (see 112264) is mediated by miR21 (611020). miR21 downregulates PDCD4 (608610), which in turn acts as a negative regulator of smooth muscle contractile genes. Surprisingly, TGF-beta and BMP signaling promoted a rapid increase in expression of mature miR21 through a posttranscriptional step, promoting the processing of primary transcripts of miR21 (pri-miR21) into precursor miR21 (pre-miR21) by the Drosha complex (see 608828). TGF-beta and BMP-specific SMAD signal transducers SMAD1 (601595), SMAD2 (601366), SMAD3, and SMAD5 (603110) are recruited to pri-miR21 in a complex with the RNA helicase p68 (DDX5; 180630), a component of the Drosha microprocessor complex. The shared cofactor SMAD4 (600993) is not required for this process. Thus, Davis et al. (2008) concluded that regulation of microRNA biogenesis by ligand-specific SMAD proteins is critical for control of the vascular smooth muscle cell phenotype and potentially for SMAD4-independent responses mediated by the TGF-beta and BMP signaling pathways.

Chuderland et al. (2008) identified an SPS motif in ERK2 (MAPK1; 176948) and SMAD3 and a similar TPT motif in MEK1 (MAP2K1; 176872) that directed protein nuclear accumulation when phosphorylated.

Using coimmunoprecipitation and in vitro binding assays, Liu et al. (2017) found that human BRD7 (618489) interacted with SMAD3 and SMAD4 in HEK293T cells. The MH1 and MH2 domains of the SMADs were sufficient for BRD7 binding, and the N-terminal region preceding the bromodomain in BRD7 was required for SMAD binding. Overexpression of BRD7 significantly increased TGF-beta-induced transcriptional activation of p21, whereas knockdown of BRD7 reduced it. Chromatin immunoprecipitation assays demonstrated that, via its bromodomain, BRD7 increased SMAD3/SMAD4 binding to the p21 promoter in the presence of TGF-beta. BRD7 also enhanced TGF-beta-induced transcriptional activity of SMAD4 by interacting and cooperating with p300 (EP300; 602700). BRD7 knockdown attenuated the TGF-beta-induced antiproliferation phenotype in tumor cells, whereas expression of BRD7 had a suppressive effect on tumor formation and enhanced TGF-beta-mediated epithelial-mesenchymal transition responses.

Bertero et al. (2018) described the interactome of SMAD2/3 in human pluripotent stem cells. This analysis revealed that SMAD2/3 is involved in multiple molecular processes in addition to its role in transcription. In particular, Bertero et al. (2018) identified a functional interaction with the METTL3 (612472)-METTL14 (616504)-WTAP (605442) complex, which mediates the conversion of adenosine to N6-methyladenosine (m6A) on RNA. Bertero et al. (2018) showed that SMAD2/3 promotes binding of the m6A methyltransferase complex to a subset of transcripts involved in early cell fate decisions. This mechanism destabilizes specific SMAD2/3 transcriptional targets, including the pluripotency factor gene NANOG (607937), priming them for rapid downregulation upon differentiation to enable timely exit from pluripotency. Bertero et al. (2018) concluded that their findings revealed the mechanism by which extracellular signaling can induce rapid cellular responses through regulation of the epitranscriptome. These aspects of TGF-beta signaling could have far-reaching implications in many other cell types and in diseases such as cancer.


Molecular Genetics

Loeys-Dietz Syndrome 3

In a 4-generation Dutch family with arterial aneurysms and dissections and early-onset osteoarthritis mapping to chromosome 15q22.2-q24.2, van de Laar et al. (2011) analyzed the candidate gene SMAD3 and identified heterozygosity for a missense mutation (R287W; 603109.0001) that segregated with disease. The authors designated the disorder aneurysms-osteoarthritis syndrome (AOS), but it is here incorporated into the Loeys-Dietz phenotypic series as Loeys-Dietz syndrome-3 (LDS3; 613795). Analysis of SMAD3 in 99 patients with thoracic aortic aneurysms and dissections and Marfan-like features, who were known to be negative for mutation in the FBN1 (134797), TGFBR1 (190181), and TGFBR2 (190182) genes, revealed 2 additional probands with heterozygous SMAD3 mutations (603109.0002; 603109.0003). All 3 mutations were located in the MH2 domain, which mediates oligomerization of SMAD3 with SMAD4 (600993) and SMAD-dependent transcriptional activation.

Regalado et al. (2011) reported 4 new mutations in SMAD3. One mutation (603109.0004) was a frameshift mutation in exon 5 segregating in a family with LDS3 phenotype. The other 3 were missense mutations in invariant codons.

Van de Laar et al. (2012) identified 5 novel SMAD3 mutations in 5 additional families with aneurysms-osteoarthritis syndrome (see, e.g., 603109.0008-603109.0010).

Associations Pending Confirmation

For discussion of a possible association between variation in the SMAD3 gene and dizygotic twinning, see 276400.

Exclusion Studies

Using cDNA, Roth et al. (2000) conducted mutation analysis of the SMAD2, SMAD3, and SMAD4 genes in 14 Finnish kindreds with hereditary nonpolyposis colon cancer (see 120435). They found no mutations.


Animal Model

Zhu et al. (1998) reported the targeted disruption of the mouse Smad3 gene. Smad3 mutant mice were viable and fertile. Between 4 and 6 months of age, the Smad3 mutant mice became moribund with colorectal adenocarcinomas. The neoplasms penetrated through the intestinal wall and metastasized to lymph nodes. Since TGF-beta transduces its signal to the interior of the cell via Smad2, Smad3, and Smad4, these results directly implicate TGF-beta signaling in the pathogenesis of colorectal cancer and provide a compelling animal model for the study of human colorectal cancer.

Yang et al. (1999) found that Smad3-null (ex8/ex8) mice died between 1 and 8 months due to a primary defect in immune function. The mice exhibited inflammatory lesions in a number of organs, including the nasal mucosa, stomach, pancreas, colon, and small intestine, as well as enlarged lymph nodes, an involuted thymus, and the formation of bacterial abscesses adjacent to mucosal surfaces. Immunostaining revealed a significant increase in T-cell activation, suggesting that Smad3 has a role in TGFB-mediated regulation of T-cell activation.

Renal tubulointerstitial fibrosis is a chronic inflammatory condition in which renal fibrosis is associated with epithelial-mesenchymal transition of the renal tubules and synthesis of extracellular matrix in response to multiple entities, including ureteral obstruction. TGFB plays a pivotal role in the disease process. Sato et al. (2003) found that Smad3-null mice with ureteral obstruction were protected against tubulointerstitial fibrosis, presumably by blocking the downstream effects of TGFB. Levels of TGFB mRNA and mature protein were decreased in the mutant animals compared to experimental controls, indicating that the Smad3 pathway is also essential for autoinduction of TGFB.

Wolfraim et al. (2004) used mice in which 1 or both alleles of Smad3 were inactivated to evaluate the role of Smad3 in the response of normal T cells to TGF-beta and in the susceptibility to spontaneous leukemogenesis in mice in which both alleles of the tumor suppressor p27(Kip1) (CDKN1B; 600778) were deleted. The loss of 1 allele for Smad3 impaired the inhibitory effect of TGF-beta on the proliferation of normal T cells and worked in tandem with the homozygous inactivation of p27(Kip1) to promote T-cell leukemogenesis. Wolfraim et al. (2004) concluded that a reduction in Smad3 expression and the loss of p27(Kip1) work synergistically to promote T-cell leukemogenesis in mice.

Ashcroft et al. (1999) generated Smad3-null mice and observed accelerated cutaneous wound healing, with complete reepithelialization by day 2 compared to day 5 in wildtype mice, and significantly reduced local infiltration of monocytes. Smad3 -/- keratinocytes showed altered patterns of growth and migration, and Smad3 -/- monocytes exhibited a selectively blunted chemotactic response to TGF-beta (190180).

Arany et al. (2006) created excisional ear wounds in Smad3 -/- mice and observed wound enlargement compared to wildtype controls. Levels of elastin and glycosaminoglycans were increased, collagen fibers were more compactly organized, and integrins, TGFB1, and matrix metalloproteinases were altered both basally and after wounding in Smad3-knockout mice. Mechanical testing revealed an increased modulus of elasticity, suggesting an imbalance of tissue forces. Arany et al. (2006) proposed that the altered mechanical elastic properties lead to a persistent retractile force that is opposed by decreased wound contractile forces.

Kanamaru et al. (2005) found that bone marrow-derived mast cells (BMMCs) from Smad3-null mice had an augmented capacity to produce proinflammatory cytokines upon stimulation with lipopolysaccharide. Mast cell-deficient mice reconstituted with Smad3-null BMMCs survived significantly longer in an acute peritonitis model than mast cell-deficient mice reconstituted with wildtype BMMCs. Kanamaru et al. (2005) proposed that SMAD3 in mast cells inhibits mast cell-mediated immune responses against gram-negative bacteria.


History

Gupta et al. (2006) retracted their paper describing the identification of a microRNA in the latency-associated transcript (Lat) of herpes simplex virus (HSV)-1 (miR-Lat) that targets TGFB and SMAD3 via sequences in their 3-prime UTRs that show partial homology to miR-Lat.

The article in which Dong et al. (2002) suggested that alterations in the SMAD pathway, including marked SMAD7 (602932) deficiency and SMAD3 upregulation, may be responsible for the TGFB1 (191080) hyperresponsiveness observed in scleroderma (181750) was retracted because some of the elements in figure 3 may have been fabricated.


ALLELIC VARIANTS ( 10 Selected Examples):

.0001 LOEYS-DIETZ SYNDROME 3

SMAD3, ARG287TRP
  
RCV000023241...

In 20 affected members of a 4-generation Dutch family with arterial aneurysms and dissections and early-onset osteoarthritis (LDS3; 613795), van de Laar et al. (2011) identified heterozygosity for an 859C-T transition in exon 6 of the SMAD3 gene, resulting in an arg287-to-trp (R287W) substitution at a highly conserved residue within the MH2 domain. The mutation was not found in 7 unaffected family members or in 544 Dutch control chromosomes. Immunohistochemical analysis of aortic wall tissue from 2 patients showed increased expression of key proteins in the TGF-beta (see TGFB1, 190180) pathway.


.0002 LOEYS-DIETZ SYNDROME 3

SMAD3, 2-BP DEL, 741AT
  
RCV000023242

In 3 Dutch sibs with arterial aneurysms and dissections and early-onset osteoarthritis (LDS3; 613795), van de Laar et al. (2011) identified heterozygosity for a 2-bp deletion (741delAT) in exon 6 of the SMAD3 gene, resulting in a frameshift and a premature termination sequence at codon 309 in exon 7 that removes nearly the complete MH2 domain (Thr247ProfsTer61). The deletion, which was presumably present in their affected deceased father, was not found in their unaffected mother or in 544 Dutch control chromosomes. Analysis of patient cDNA showed very weak mutant signal compared to wildtype, and treatment of patient fibroblast cultures with cycloheximide markedly increased the mutant signal, indicating that most of the abnormal RNA was subjected to nonsense messenger RNA decay and that a truncated SMAD3 protein was barely formed.


.0003 LOEYS-DIETZ SYNDROME 3

SMAD3, THR261ILE
  
RCV000023243

In a Dutch male patient with arterial aneurysm and early-onset osteoarthritis (LDS3; 613795), van de Laar et al. (2011) identified heterozygosity for a 783C-T transition in exon 6 of the SMAD3 gene, resulting in a thr261-to-ile (T261I) substitution at a highly conserved residue in the MH2 domain. The mutation was not found in 544 Dutch control chromosomes.


.0004 LOEYS-DIETZ SYNDROME 3

SMAD3, 1-BP DEL, 652A
  
RCV000023244...

In a 3-generation pedigree segregating autosomal dominant thoracic aortic aneurysms and dissections with intracranial and other arterial aneurysms (LDS3; 613795), Regalado et al. (2011) identified a deletion of an A at nucleotide 652 in exon 5 of the SMAD3 gene, resulting in frameshift leading to premature termination following asparagine-218 (N218fs). This mutation was present in all individuals with vascular disease in the family and segregated with a lod score of 2.52. The pedigree had originally been reported by Regalado et al. (2011). The mutation was absent from 2,300 control exomes.


.0005 LOEYS-DIETZ SYNDROME 3

SMAD3, ARG279LYS
  
RCV000023245

In 2 unrelated families of European descent with autosomal dominant thoracic aortic and other aneurysms (LDS3; 613795), Regalado et al. (2011) identified a G-to-A transition at nucleotide 836 in exon 6 of the SMAD3 gene, resulting in an arg-to-lys substitution at codon 279 (R279K). Arg279 is completely conserved from human to Drosophila, and the R279K mutation was predicted to disrupt protein function. The mutation was not identified in 2,300 control exomes. There was decreased penetrance in younger family members.


.0006 LOEYS-DIETZ SYNDROME 3

SMAD3, GLU239LYS
  
RCV000023246...

In a small family with 3 sibs affected with thoracic aortic aneurysm and dissection (LDS3; 613795), Regalado et al. (2011) identified a G-to-A transition at nucleotide 715 in exon 6 of the SMAD3 gene, resulting in a glutamine-to-lysine substitution at codon 239 (E239K). Exon 6 encodes the MH2 protein-protein binding domain. Glu239 is completely conserved from human to Drosophila, and the E239K mutation was predicted to disrupt protein function. The mutation was not identified in 2,300 control exomes.


.0007 LOEYS-DIETZ SYNDROME 3

SMAD3, ALA112VAL
  
RCV000023247...

In a family segregating autosomal dominant thoracic aortic aneurysm with dissection as well as other features of Loeys-Dietz syndrome (LDS3; 613795) including bifid uvula and scoliosis, and early-onset osteoarthritis, Regalado et al. (2011) identified a heterozygous alanine-to-valine substitution at codon 112 (A112V). The mutation segregated with disease with reduced penetrance in this family and was not identified in 2,300 control exomes. Guo (2012) stated that the correct nucleotide change for the A112V mutation is 335C-T in exon 2 rather than 235C-T as cited in Regalado et al. (2011).


.0008 LOEYS-DIETZ SYNDROME 3

SMAD3, 1-BP DEL, 313G
  
RCV000023248...

In a patient with aneurysms-osteoarthritis syndrome (LCS3; 613795), van de Laar et al. (2012) identified a 1-bp deletion at nucleotide 313 of the SMAD3 gene (313delG), resulting in a frameshift (Ala105ProfsTer11).


.0009 LOEYS-DIETZ SYNDROME 3

SMAD3, PRO263LEU
  
RCV000023249...

In a patient with aneurysms-osteoarthritis syndrome (LCS3; 613795), van de Laar et al. (2012) identified a 788C-T transition in the SMAD3 gene, resulting in a pro263-to-leu (P263L) substitution.


.0010 LOEYS-DIETZ SYNDROME 3

SMAD3, GLU361TER
  
RCV000023250

In affected members of a family segregating aneurysms-osteoarthritis syndrome (LCS3; 613795), van de Laar et al. (2012) identified a 1-bp duplication at nucleotide 1080 in the SMAD3 gene (1080dupT), resulting in a glu361-to-ter (E361X) substitution.


REFERENCES

  1. Arai, T., Akiyama, Y., Okabe, S., Ando, M., Endo, M., Yuasa, Y. Genomic structure of the human Smad3 gene and its infrequent alterations in colorectal cancers. Cancer Lett. 122: 157-163, 1998. [PubMed: 9464505, related citations] [Full Text]

  2. Arany, P. R., Flanders, K. C., Kobayashi, T., Kuo, C. K., Stuelten, C., Desai, K. V., Tuan, R., Rennard, S. I., Roberts, A. B. Smad3 deficiency alters key structural elements of the extracellular matrix and mechanotransduction of wound closure. Proc. Nat. Acad. Sci. 103: 9250-9255, 2006. [PubMed: 16754864, images, related citations] [Full Text]

  3. Ashcroft, G. S., Yang, X., Glick, A. B., Weinstein, M., Letterio, J. L., Mizel, D. E., Anzano, M., Greenwell-Wild, T., Wahl, S. M., Deng, C., Roberts, A. B. Mice lacking Smad3 show accelerated wound healing and an impaired local inflammatory response. Nature Cell Biol. 1: 260-266, 1999. [PubMed: 10559937, related citations] [Full Text]

  4. Bertero, A., Brown, S., Madrigal, P., Osnato, A., Ortmann, D., Yiangou, L., Kadiwala, J., Hubner, N. C., de los Mozos, I. R., Sadee, C., Lenaerts, A.-S., Nakanoh, S., Grandy, R., Farnell, E., Ule, J., Stunnenberg, H. G., Mendjan, S., Vallier, L. The SMAD2/3 interactome reveals that TGF-beta controls m6A mRNA methylation in pluripotency. Nature 555: 256-259, 2018. [PubMed: 29489750, related citations] [Full Text]

  5. Carlson, M. E., Hsu, M., Conboy, I. M. Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells. Nature 454: 528-532, 2008. Note: Erratum: Nature 538: 274 only, 2016. [PubMed: 18552838, related citations] [Full Text]

  6. Chen, C.-R., Kang, Y., Siegel, P. M., Massague, J. E2F4/5 and p107 as Smad cofactors linking the TGF-beta receptor to c-myc repression. Cell 110: 19-32, 2002. [PubMed: 12150994, related citations] [Full Text]

  7. Chuderland, D., Konson, A., Seger, R. Identification and characterization of a general nuclear translocation signal in signaling proteins. Molec. Cell 31: 850-861, 2008. [PubMed: 18760948, related citations] [Full Text]

  8. Davis, B. N., Hilyard, A. C., Lagna, G., Hata, A. SMAD proteins control DROSHA-mediated microRNA maturation. Nature 454: 56-61, 2008. [PubMed: 18548003, images, related citations] [Full Text]

  9. Dong, C., Zhu, S., Wang, T., Yoon, W., Li, Z., Alvarez, R. J., ten Dijke, P., White, B., Wigley, F. M., Goldschmidt-Clermont, P. J. Deficient Smad7 expression: a putative molecular defect in scleroderma. Proc. Nat. Acad. Sci. 99: 3908-3913, 2002. Note: Retraction: Proc. Nat. Acad. Sci. 113: E2208, 2016. [PubMed: 11904440, related citations] [Full Text]

  10. Guo, D.-C. Personal Communication. Houston, Tex. 2/7/2012.

  11. Gupta, A., Gartner, J. J., Sethupathy, P., Hatzigeorgiou, A. G., Fraser, N. W. Anti-apoptotic function of a microRNA encoded by the HSV-1 latency-associated transcript. Nature 442: 82-85, 2006. Note: Retraction: Nature 451: 600 only, 2008. [PubMed: 16738545, related citations] [Full Text]

  12. Inman, G. J., Nicolas, F. J., Hill, C. S. Nucleocytoplasmic shuttling of Smads 2, 3, and 4 permits sensing of TGF-beta receptor activity. Molec. Cell 10: 283-294, 2002. [PubMed: 12191474, related citations] [Full Text]

  13. Kanamaru, Y., Sumiyoshi, K., Ushio, H., Ogawa, H., Okumura, K., Nakao, A. Smad3 deficiency in mast cells provides efficient host protection against acute septic peritonitis. J. Immun. 174: 4193-4197, 2005. [PubMed: 15778380, related citations] [Full Text]

  14. Liu, T., Zhao, M., Liu, J., He, Z., Zhang, Y., You, H., Huang, J., Lin, X., Feng, X.-H. Tumor suppressor bromodomain-containing protein 7 cooperates with Smads to promote transforming growth factor-beta responses. Oncogene 36: 362-372, 2017. [PubMed: 27270427, related citations] [Full Text]

  15. Matsuura, I., Denissova, N. G., Wang, G., He, D., Long, J., Liu, F. Cyclin-dependent kinases regulate the antiproliferative function of Smads. Nature 430: 226-231, 2004. [PubMed: 15241418, related citations] [Full Text]

  16. Regalado, E., Medrek, S., Tran-Fadulu, V., Guo, D.-C., Pannu, H., Golabbakhsh, H., Smart, S., Chen, J. H., Shete, S., Kim, D. H., Stern, R., Braverman, A. C., Milewicz, D. M. Autosomal dominant inheritance of a predisposition to thoracic aortic aneurysms and dissections and intracranial saccular aneurysms. Am. J. Med. Genet. 155A: 2125-2130, 2011. [PubMed: 21815248, related citations] [Full Text]

  17. Regalado, E. S., Guo, D., Villamizar, C., Avidan, N., Gilchrist, D., McGillivray, B., Clarke, L., Bernier, F., Santos-Cortez, R. L., Leal, S. M., Bertoli-Avella, A. M., Shendure, J., Rieder, M. J., Nickerson, D. A., NHLBI GO Exome Sequencing Project, Milewicz, D. M. Exome sequencing identifies SMAD3 mutations as a cause of familial thoracic aortic aneurysm and dissection with intracranial and other arterial aneurysms. Circ. Res. 109: 680-686, 2011. [PubMed: 21778426, images, related citations] [Full Text]

  18. Riggins, G. J., Thiagalingam, S., Rozenblum, E., Weinstein, C. L., Kern, S. E., Hamilton, S. R., Willson, J. K. V., Markowitz, S. D., Kinzler, K. W., Vogelstein, B. Mad-related genes in the human. Nature Genet. 13: 347-349, 1996. [PubMed: 8673135, related citations] [Full Text]

  19. Roth, S., Johansson, M., Loukola, A., Peltomaki, P., Jarvinen, H., Mecklin, J.-P., Aaltonen, L. A. Mutation analysis of SMAD2, SMAD3, and SMAD4 genes in hereditary non-polyposis colorectal cancer. J. Med. Genet. 37: 298-300, 2000. [PubMed: 10819637, related citations] [Full Text]

  20. Sato, M., Muragaki, Y., Saika, S., Roberts, A. B., Ooshima, A. Targeted disruption of TGF-beta-1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J. Clin. Invest. 112: 1486-1494, 2003. [PubMed: 14617750, images, related citations] [Full Text]

  21. Shattuck, T. M., Costa, J., Bernstein, M., Jensen, R. T., Chung, D. C., Arnold, A. Mutational analysis of Smad3, a candidate tumor suppressor implicated in TGF-beta and menin pathways, in parathyroid adenomas and enteropancreatic endocrine tumors. J. Clin. Endocr. Metab. 87: 3911-3914, 2002. [PubMed: 12161532, related citations] [Full Text]

  22. van de Laar, I. M. B. H., Oldenburg, R. A., Pals, G., Roos-Hesselink, J. W., de Graaf, B. M., Verhagen, J. M. A., Hoedemaekers, Y. M., Willemsen, R., Severijnen, L.-A., Venselaar, H., Vriend, G., Pattynama, P. M., and 14 others. Mutations in SMAD3 cause a syndrome form of aortic aneurysms and dissections with early-onset osteoarthritis. Nature Genet. 43: 121-126, 2011. [PubMed: 21217753, related citations] [Full Text]

  23. van de Laar, I. M. B. H., van der Linde, D., Oei, E. H. G., Bos, P. K., Bessems, J. H., Bierma-Zeinstra, S. M., van Meer, B. L., Pals, G., Oldenburg, R. A., Bekkers, J. A., Moelker, A., de Graaf, B. M., and 17 others. Phenotypic spectrum of the SMAD3-related aneurysms-osteoarthritis syndrome. J. Med. Genet. 49: 47-57, 2012. [PubMed: 22167769, related citations] [Full Text]

  24. Wolfraim, L. A., Fernandez, T. M., Mamura, M., Fuller, W. L., Kumar, R., Cole, D. E., Byfield, S., Felici, A., Flanders, K. C., Walz, T. M., Roberts, A. B., Aplan, P. D., Balis, F. M., Letterio, J. J. Loss of Smad3 in acute T-cell lymphoblastic leukemia. New Eng. J. Med. 351: 552-559, 2004. [PubMed: 15295048, related citations] [Full Text]

  25. Yang, X., Letterio, J. J., Lechleider, R. J., Chen, L., Hayman, R., Gu, H., Roberts, A. B., Deng, C. Targeted disruption of SMAD3 results in impaired mucosal immunity and diminished T cell responsiveness to TGF-beta. EMBO J. 18: 1280-1291, 1999. [PubMed: 10064594, related citations] [Full Text]

  26. You, L., Kruse, F. E. Differential effect of activin A and BMP-7 on myofibroblast differentiation and the role of the Smad signaling pathway. Invest. Ophthal. Vis. Sci. 43: 72-81, 2002. [PubMed: 11773015, related citations]

  27. Zawel, L., Dai, J. L., Buckhaults, P., Zhou, S., Kinzler, K. W., Vogelstein, B., Kern, S. E. Human Smad3 and Smad4 are sequence-specific transcription activators. Molec. Cell 1: 611-617, 1998. [PubMed: 9660945, related citations] [Full Text]

  28. Zhang, Y., Feng, X.-H., Wu, R.-Y., Derynck, R. Receptor-associated Mad homologues synergize as effectors of the TGF-beta response. Nature 383: 168-172, 1996. [PubMed: 8774881, related citations] [Full Text]

  29. Zhu, Y., Richardson, J. A., Parada, L. F., Graff, J. M. Smad3 mutant mice develop metastatic colorectal cancer. Cell 94: 703-714, 1998. [PubMed: 9753318, related citations] [Full Text]


Bao Lige - updated : 06/28/2019
Ada Hamosh - updated : 08/13/2018
Marla J. F. O'Neill - updated : 6/14/2016
Ada Hamosh - updated : 2/6/2012
Ada Hamosh - updated : 9/26/2011
Marla J. F. O'Neill - updated : 3/7/2011
Patricia A. Hartz - updated : 5/29/2009
Ada Hamosh - updated : 8/29/2008
Ada Hamosh - updated : 8/13/2008
Ada Hamosh - updated : 4/4/2008
Paul J. Converse - updated : 11/1/2006
Marla J. F. O'Neill - updated : 7/28/2006
Patricia A. Hartz - updated : 7/20/2006
Ada Hamosh - updated : 9/29/2004
Victor A. McKusick - updated : 9/13/2004
Ada Hamosh - updated : 8/26/2004
Cassandra L. Kniffin - updated : 12/4/2003
John A. Phillips, III - updated : 4/8/2003
Stylianos E. Antonarakis - updated : 9/11/2002
Stylianos E. Antonarakis - updated : 7/26/2002
Jane Kelly - updated : 7/8/2002
Victor A. McKusick - updated : 4/25/2002
Michael J. Wright - updated : 1/8/2001
Ada Hamosh - updated : 8/31/2000
Stylianos E. Antonarakis - updated : 1/31/1999
Stylianos E. Antonarakis - updated : 10/13/1998
Creation Date:
Patti M. Sherman : 10/9/1998
carol : 01/08/2020
mgross : 06/28/2019
alopez : 08/13/2018
carol : 08/17/2017
alopez : 12/19/2016
carol : 06/20/2016
carol : 6/17/2016
carol : 6/14/2016
carol : 11/14/2014
alopez : 4/22/2014
carol : 9/6/2012
carol : 2/27/2012
terry : 2/7/2012
carol : 2/7/2012
terry : 2/6/2012
alopez : 10/24/2011
alopez : 10/5/2011
terry : 9/26/2011
carol : 3/7/2011
terry : 3/7/2011
terry : 6/3/2009
mgross : 6/2/2009
terry : 5/29/2009
alopez : 9/11/2008
terry : 8/29/2008
alopez : 8/20/2008
terry : 8/13/2008
alopez : 4/14/2008
terry : 4/4/2008
wwang : 12/28/2007
terry : 12/11/2007
mgross : 11/7/2006
terry : 11/1/2006
wwang : 8/7/2006
terry : 7/28/2006
mgross : 7/20/2006
carol : 4/28/2005
carol : 4/28/2005
mgross : 4/13/2005
terry : 9/29/2004
tkritzer : 9/14/2004
terry : 9/13/2004
tkritzer : 8/30/2004
terry : 8/26/2004
carol : 12/8/2003
ckniffin : 12/4/2003
cwells : 4/29/2003
terry : 4/8/2003
mgross : 9/11/2002
mgross : 7/26/2002
mgross : 7/26/2002
mgross : 7/8/2002
mgross : 4/25/2002
alopez : 1/8/2001
alopez : 9/5/2000
terry : 8/31/2000
carol : 1/31/1999
carol : 10/13/1998
carol : 10/13/1998

* 603109

SMAD FAMILY MEMBER 3; SMAD3


Alternative titles; symbols

MOTHERS AGAINST DECAPENTAPLEGIC, DROSOPHILA, HOMOLOG OF, 3; MADH3
SMA- AND MAD-RELATED PROTEIN 3


HGNC Approved Gene Symbol: SMAD3

Cytogenetic location: 15q22.33     Genomic coordinates (GRCh38): 15:67,065,602-67,195,169 (from NCBI)


Gene-Phenotype Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
15q22.33 Loeys-Dietz syndrome 3 613795 Autosomal dominant 3

TEXT

Cloning and Expression

Drosophila Mad is required for signaling by the TGF-beta (e.g., 190180)-related factor decapentaplegic. Zhang et al. (1996) cloned a human cDNA encoding MADH3, a homolog of Drosophila Mad. The deduced 425-amino acid MADH3 protein (GenBank 2522267) is 92% identical to MADH2 (601366).

By searching an expressed sequence tag database with the protein sequences of Mad and Mad homologs, Riggins et al. (1996) isolated human cDNAs encoding MADH3, which they called JV15-2. The C terminus of MADH3 shows significant homology to that of Drosophila Mad.


Gene Structure

Arai et al. (1998) determined the genomic structure of SMAD3, which contains 9 exons.


Mapping

Riggins et al. (1996) mapped the MADH3 gene to 15q21-q22 by somatic cell hybrid analysis and screening of YAC clones.


Gene Function

Zhang et al. (1996) showed that MADH3 and MADH4 (SMAD4; 600993) synergized to induce strong ligand-independent TGF-beta-like responses. MADH3 containing a C-terminal truncation acted as a dominant-negative inhibitor of the normal TGF-beta response. The activity of MADH3 was regulated by the TGF-beta receptors (e.g., 190181), and MADH3 was phosphorylated and associated with the ligand-bound receptor complex. Zhang et al. (1996) stated that these results define MADH3 as an effector of the TGF-beta response.

Zawel et al. (1998) found that human SMAD3 and SMAD4 proteins could specifically recognize an identical 8-bp palindromic sequence (GTCTAGAC). Tandem repeats of this palindrome conferred striking TGF-beta responsiveness to a minimal promoter. This responsiveness was abrogated by targeted deletion of the cellular SMAD4 gene. These results showed that SMAD proteins are involved in the biologic responses to TGF-beta and related ligands.

You and Kruse (2002) studied corneal myofibroblast differentiation and signal transduction induced by the TGFB family members activin A (147290) and bone morphogenetic protein-7 (BMP7; 112267). They found that activin A induced phosphorylation of SMAD2 (601366), and BMP7 induced SMAD1 (601595), both of which were inhibited by follistatin (136470). Transfection with antisense SMAD2/SMAD3 prevented activin-induced expression and accumulation of alpha-smooth muscle actin. The authors concluded that TGFB proteins have different functions in the cornea. Activin A and TGFB1, but not BMP7, are regulators of keratocyte differentiation and might play a role during myofibroblast transdifferentiation. SMAD2/SMAD3 signal transduction appeared to be important in the regulation of muscle-specific genes.

SMAD3 is a direct mediator of transcriptional activation by the TGF-beta receptor. Its target genes in epithelial cells include cyclin-dependent kinase (CDK; see 116953) inhibitors that generate a cytostatic response. Chen et al. (2002) defined how, in the same context, SMAD3 can mediate transcriptional repression of the growth-promoting gene MYC (190080). A complex containing SMAD3, the transcription factors E2F4 (600659), E2F5 (600967), and DP1 (189902), and the corepressor p107 (116957) preexists in the cytoplasm. In response to TGF-beta, this complex moves into the nucleus and associates with SMAD4, recognizing a composite SMAD-E2F site on MYC for repression. Previously known as the ultimate recipients of CDK regulatory signals, E2F4/E2F5 and p107 act here as transducers of TGF-beta receptor signals upstream of CDK. SMAD proteins therefore mediate transcriptional activation or repression depending on their associated partners.

TGFB (190180) stimulation leads to phosphorylation and activation of SMAD2 and SMAD3, which form complexes with SMAD4 that accumulate in the nucleus and regulate transcription of target genes. Inman et al. (2002) demonstrated that following TGFB stimulation of epithelial cells, receptors remain active for at least 3 to 4 hours, and continuous receptor activity is required to maintain active SMADs in the nucleus and for TGFB-induced transcription. Continuous nucleocytoplasmic shuttling of the SMADs during active TGFB signaling provides the mechanism whereby the intracellular transducers of the signal continuously monitor receptor activity. These data explain how, at all times, the concentration of active SMADs in the nucleus is directly dictated by the levels of activated receptors in the cytoplasm.

Based upon molecular allelotyping and comparative genomic hybridization studies, chromosome 15q is the likely location of a tumor suppressor gene important in the pathogeneses of sporadic enteropancreatic endocrine tumors and parathyroid adenomas. To determine whether SMAD3 plays a primary role in the development of these tumors, Shattuck et al. (2002) investigated 20 enteropancreatic tumors and 67 parathyroid adenomas for LOH at DNA markers surrounding SMAD3. Twenty percent of enteropancreatic tumors and 24% of parathyroid adenomas showed loss. All 9 coding exons and intron-exon boundaries of the SMAD3 gene were then sequenced in genomic DNA from all 20 enteropancreatic and 25 parathyroid tumors, including every case with LOH. No acquired clonal mutations, insertions, or microdeletions in SMAD3 were detected in any tumors. Because inactivating somatic mutation is the hallmark of an authentic tumor suppressor, SMAD3 is unlikely to function as a classic tumor suppressor gene in the pathogenesis of sporadic parathyroid or enteropancreatic endocrine tumors.

Matsuura et al. (2004) showed that SMAD3 is a major physiologic substrate of the G1 cyclin-dependent kinases CDK4 (123829) and CDK2 (116953). Except for the retinoblastoma protein family, SMAD3 was the only CDK4 substrate demonstrated to that time. Matsuura et al. (2004) mapped CDK4 and CDK2 phosphorylation sites to thr8, thr178, and ser212 in SMAD3. Mutation of the CDK phosphorylation sites increased Smad3 transcriptional activity, leading to higher expression of the CDK inhibitor p15 (600431). Mutation of the CDK phosphorylation sites of Smad3 also increased its ability to downregulate the expression of c-myc. Using Smad3 knockout mouse embryonic fibroblasts and other epithelial cell lines, Matsuura et al. (2004) further showed that Smad3 inhibits cell cycle progression from G1 to S phase and that mutation of the CDK phosphorylation sites in Smad3 increases this ability. They concluded that CDK phosphorylation of SMAD3 inhibits its transcriptional activity and antiproliferative function.

To determine the role of SMAD3 in the pathogenesis of lymphoid neoplasia, Wolfraim et al. (2004) measured SMAD3 mRNA and protein in leukemia cells obtained at diagnosis from 19 children with acute leukemia: 10 with T-cell acute lymphoblastic leukemia (ALL), 7 with pre-B-cell ALL, and 2 with acute nonlymphoblastic leukemia (ANLL). SMAD3 protein was absent in T-cell ALL but present in pre-B-cell ALL and ANLL. No mutations in the SMAD3 gene were identified in T-cell ALL, and SMAD3 mRNA was present in T-cell ALL and normal T cells at similar levels. Wolfraim et al. (2004) concluded that loss of SMAD3 protein is a specific feature of pediatric T-cell lymphoblastic leukemia.

In experiments using mouse muscle, Carlson et al. (2008) found that, in addition to the loss of Notch (190198) activation, old muscle produces excessive TGF-beta (but not myostatin, 601788), which induces unusually high levels of Smad3 in resident satellite cells and interferes with the regenerative capacity. Importantly, endogenous Notch and Smad3 antagonize each other in the control of satellite cell proliferation, such that activation of Notch blocks the TGF-beta-dependent upregulation of the cyclin-dependent kinase (CDK) inhibitors p15, p16 (600160), p21 (116899), and p27 (600778), whereas inhibition of Notch induces them. Furthermore, in muscle stem cells, Notch activity determined the binding of Smad3 to the promoters of these negative regulators of cell cycle progression. Attenuation of TGF-beta/Smad3 in old, injured muscle restored regeneration to satellite cells in vivo. Thus, a balance between endogenous Smad3 and active Notch controls the regenerative competence of muscle stem cells, and deregulation of this balance in the old muscle microniche interferes with regeneration.

Davis et al. (2008) demonstrated that induction of a contractile phenotype in human vascular smooth muscle cells by TGF-beta (190180) and BMPs (see 112264) is mediated by miR21 (611020). miR21 downregulates PDCD4 (608610), which in turn acts as a negative regulator of smooth muscle contractile genes. Surprisingly, TGF-beta and BMP signaling promoted a rapid increase in expression of mature miR21 through a posttranscriptional step, promoting the processing of primary transcripts of miR21 (pri-miR21) into precursor miR21 (pre-miR21) by the Drosha complex (see 608828). TGF-beta and BMP-specific SMAD signal transducers SMAD1 (601595), SMAD2 (601366), SMAD3, and SMAD5 (603110) are recruited to pri-miR21 in a complex with the RNA helicase p68 (DDX5; 180630), a component of the Drosha microprocessor complex. The shared cofactor SMAD4 (600993) is not required for this process. Thus, Davis et al. (2008) concluded that regulation of microRNA biogenesis by ligand-specific SMAD proteins is critical for control of the vascular smooth muscle cell phenotype and potentially for SMAD4-independent responses mediated by the TGF-beta and BMP signaling pathways.

Chuderland et al. (2008) identified an SPS motif in ERK2 (MAPK1; 176948) and SMAD3 and a similar TPT motif in MEK1 (MAP2K1; 176872) that directed protein nuclear accumulation when phosphorylated.

Using coimmunoprecipitation and in vitro binding assays, Liu et al. (2017) found that human BRD7 (618489) interacted with SMAD3 and SMAD4 in HEK293T cells. The MH1 and MH2 domains of the SMADs were sufficient for BRD7 binding, and the N-terminal region preceding the bromodomain in BRD7 was required for SMAD binding. Overexpression of BRD7 significantly increased TGF-beta-induced transcriptional activation of p21, whereas knockdown of BRD7 reduced it. Chromatin immunoprecipitation assays demonstrated that, via its bromodomain, BRD7 increased SMAD3/SMAD4 binding to the p21 promoter in the presence of TGF-beta. BRD7 also enhanced TGF-beta-induced transcriptional activity of SMAD4 by interacting and cooperating with p300 (EP300; 602700). BRD7 knockdown attenuated the TGF-beta-induced antiproliferation phenotype in tumor cells, whereas expression of BRD7 had a suppressive effect on tumor formation and enhanced TGF-beta-mediated epithelial-mesenchymal transition responses.

Bertero et al. (2018) described the interactome of SMAD2/3 in human pluripotent stem cells. This analysis revealed that SMAD2/3 is involved in multiple molecular processes in addition to its role in transcription. In particular, Bertero et al. (2018) identified a functional interaction with the METTL3 (612472)-METTL14 (616504)-WTAP (605442) complex, which mediates the conversion of adenosine to N6-methyladenosine (m6A) on RNA. Bertero et al. (2018) showed that SMAD2/3 promotes binding of the m6A methyltransferase complex to a subset of transcripts involved in early cell fate decisions. This mechanism destabilizes specific SMAD2/3 transcriptional targets, including the pluripotency factor gene NANOG (607937), priming them for rapid downregulation upon differentiation to enable timely exit from pluripotency. Bertero et al. (2018) concluded that their findings revealed the mechanism by which extracellular signaling can induce rapid cellular responses through regulation of the epitranscriptome. These aspects of TGF-beta signaling could have far-reaching implications in many other cell types and in diseases such as cancer.


Molecular Genetics

Loeys-Dietz Syndrome 3

In a 4-generation Dutch family with arterial aneurysms and dissections and early-onset osteoarthritis mapping to chromosome 15q22.2-q24.2, van de Laar et al. (2011) analyzed the candidate gene SMAD3 and identified heterozygosity for a missense mutation (R287W; 603109.0001) that segregated with disease. The authors designated the disorder aneurysms-osteoarthritis syndrome (AOS), but it is here incorporated into the Loeys-Dietz phenotypic series as Loeys-Dietz syndrome-3 (LDS3; 613795). Analysis of SMAD3 in 99 patients with thoracic aortic aneurysms and dissections and Marfan-like features, who were known to be negative for mutation in the FBN1 (134797), TGFBR1 (190181), and TGFBR2 (190182) genes, revealed 2 additional probands with heterozygous SMAD3 mutations (603109.0002; 603109.0003). All 3 mutations were located in the MH2 domain, which mediates oligomerization of SMAD3 with SMAD4 (600993) and SMAD-dependent transcriptional activation.

Regalado et al. (2011) reported 4 new mutations in SMAD3. One mutation (603109.0004) was a frameshift mutation in exon 5 segregating in a family with LDS3 phenotype. The other 3 were missense mutations in invariant codons.

Van de Laar et al. (2012) identified 5 novel SMAD3 mutations in 5 additional families with aneurysms-osteoarthritis syndrome (see, e.g., 603109.0008-603109.0010).

Associations Pending Confirmation

For discussion of a possible association between variation in the SMAD3 gene and dizygotic twinning, see 276400.

Exclusion Studies

Using cDNA, Roth et al. (2000) conducted mutation analysis of the SMAD2, SMAD3, and SMAD4 genes in 14 Finnish kindreds with hereditary nonpolyposis colon cancer (see 120435). They found no mutations.


Animal Model

Zhu et al. (1998) reported the targeted disruption of the mouse Smad3 gene. Smad3 mutant mice were viable and fertile. Between 4 and 6 months of age, the Smad3 mutant mice became moribund with colorectal adenocarcinomas. The neoplasms penetrated through the intestinal wall and metastasized to lymph nodes. Since TGF-beta transduces its signal to the interior of the cell via Smad2, Smad3, and Smad4, these results directly implicate TGF-beta signaling in the pathogenesis of colorectal cancer and provide a compelling animal model for the study of human colorectal cancer.

Yang et al. (1999) found that Smad3-null (ex8/ex8) mice died between 1 and 8 months due to a primary defect in immune function. The mice exhibited inflammatory lesions in a number of organs, including the nasal mucosa, stomach, pancreas, colon, and small intestine, as well as enlarged lymph nodes, an involuted thymus, and the formation of bacterial abscesses adjacent to mucosal surfaces. Immunostaining revealed a significant increase in T-cell activation, suggesting that Smad3 has a role in TGFB-mediated regulation of T-cell activation.

Renal tubulointerstitial fibrosis is a chronic inflammatory condition in which renal fibrosis is associated with epithelial-mesenchymal transition of the renal tubules and synthesis of extracellular matrix in response to multiple entities, including ureteral obstruction. TGFB plays a pivotal role in the disease process. Sato et al. (2003) found that Smad3-null mice with ureteral obstruction were protected against tubulointerstitial fibrosis, presumably by blocking the downstream effects of TGFB. Levels of TGFB mRNA and mature protein were decreased in the mutant animals compared to experimental controls, indicating that the Smad3 pathway is also essential for autoinduction of TGFB.

Wolfraim et al. (2004) used mice in which 1 or both alleles of Smad3 were inactivated to evaluate the role of Smad3 in the response of normal T cells to TGF-beta and in the susceptibility to spontaneous leukemogenesis in mice in which both alleles of the tumor suppressor p27(Kip1) (CDKN1B; 600778) were deleted. The loss of 1 allele for Smad3 impaired the inhibitory effect of TGF-beta on the proliferation of normal T cells and worked in tandem with the homozygous inactivation of p27(Kip1) to promote T-cell leukemogenesis. Wolfraim et al. (2004) concluded that a reduction in Smad3 expression and the loss of p27(Kip1) work synergistically to promote T-cell leukemogenesis in mice.

Ashcroft et al. (1999) generated Smad3-null mice and observed accelerated cutaneous wound healing, with complete reepithelialization by day 2 compared to day 5 in wildtype mice, and significantly reduced local infiltration of monocytes. Smad3 -/- keratinocytes showed altered patterns of growth and migration, and Smad3 -/- monocytes exhibited a selectively blunted chemotactic response to TGF-beta (190180).

Arany et al. (2006) created excisional ear wounds in Smad3 -/- mice and observed wound enlargement compared to wildtype controls. Levels of elastin and glycosaminoglycans were increased, collagen fibers were more compactly organized, and integrins, TGFB1, and matrix metalloproteinases were altered both basally and after wounding in Smad3-knockout mice. Mechanical testing revealed an increased modulus of elasticity, suggesting an imbalance of tissue forces. Arany et al. (2006) proposed that the altered mechanical elastic properties lead to a persistent retractile force that is opposed by decreased wound contractile forces.

Kanamaru et al. (2005) found that bone marrow-derived mast cells (BMMCs) from Smad3-null mice had an augmented capacity to produce proinflammatory cytokines upon stimulation with lipopolysaccharide. Mast cell-deficient mice reconstituted with Smad3-null BMMCs survived significantly longer in an acute peritonitis model than mast cell-deficient mice reconstituted with wildtype BMMCs. Kanamaru et al. (2005) proposed that SMAD3 in mast cells inhibits mast cell-mediated immune responses against gram-negative bacteria.


History

Gupta et al. (2006) retracted their paper describing the identification of a microRNA in the latency-associated transcript (Lat) of herpes simplex virus (HSV)-1 (miR-Lat) that targets TGFB and SMAD3 via sequences in their 3-prime UTRs that show partial homology to miR-Lat.

The article in which Dong et al. (2002) suggested that alterations in the SMAD pathway, including marked SMAD7 (602932) deficiency and SMAD3 upregulation, may be responsible for the TGFB1 (191080) hyperresponsiveness observed in scleroderma (181750) was retracted because some of the elements in figure 3 may have been fabricated.


ALLELIC VARIANTS 10 Selected Examples):

.0001   LOEYS-DIETZ SYNDROME 3

SMAD3, ARG287TRP
SNP: rs387906850, ClinVar: RCV000023241, RCV000195645, RCV000699559, RCV002276569

In 20 affected members of a 4-generation Dutch family with arterial aneurysms and dissections and early-onset osteoarthritis (LDS3; 613795), van de Laar et al. (2011) identified heterozygosity for an 859C-T transition in exon 6 of the SMAD3 gene, resulting in an arg287-to-trp (R287W) substitution at a highly conserved residue within the MH2 domain. The mutation was not found in 7 unaffected family members or in 544 Dutch control chromosomes. Immunohistochemical analysis of aortic wall tissue from 2 patients showed increased expression of key proteins in the TGF-beta (see TGFB1, 190180) pathway.


.0002   LOEYS-DIETZ SYNDROME 3

SMAD3, 2-BP DEL, 741AT
SNP: rs587776880, ClinVar: RCV000023242

In 3 Dutch sibs with arterial aneurysms and dissections and early-onset osteoarthritis (LDS3; 613795), van de Laar et al. (2011) identified heterozygosity for a 2-bp deletion (741delAT) in exon 6 of the SMAD3 gene, resulting in a frameshift and a premature termination sequence at codon 309 in exon 7 that removes nearly the complete MH2 domain (Thr247ProfsTer61). The deletion, which was presumably present in their affected deceased father, was not found in their unaffected mother or in 544 Dutch control chromosomes. Analysis of patient cDNA showed very weak mutant signal compared to wildtype, and treatment of patient fibroblast cultures with cycloheximide markedly increased the mutant signal, indicating that most of the abnormal RNA was subjected to nonsense messenger RNA decay and that a truncated SMAD3 protein was barely formed.


.0003   LOEYS-DIETZ SYNDROME 3

SMAD3, THR261ILE
SNP: rs387906851, ClinVar: RCV000023243

In a Dutch male patient with arterial aneurysm and early-onset osteoarthritis (LDS3; 613795), van de Laar et al. (2011) identified heterozygosity for a 783C-T transition in exon 6 of the SMAD3 gene, resulting in a thr261-to-ile (T261I) substitution at a highly conserved residue in the MH2 domain. The mutation was not found in 544 Dutch control chromosomes.


.0004   LOEYS-DIETZ SYNDROME 3

SMAD3, 1-BP DEL, 652A
SNP: rs587776881, ClinVar: RCV000023244, RCV001385143

In a 3-generation pedigree segregating autosomal dominant thoracic aortic aneurysms and dissections with intracranial and other arterial aneurysms (LDS3; 613795), Regalado et al. (2011) identified a deletion of an A at nucleotide 652 in exon 5 of the SMAD3 gene, resulting in frameshift leading to premature termination following asparagine-218 (N218fs). This mutation was present in all individuals with vascular disease in the family and segregated with a lod score of 2.52. The pedigree had originally been reported by Regalado et al. (2011). The mutation was absent from 2,300 control exomes.


.0005   LOEYS-DIETZ SYNDROME 3

SMAD3, ARG279LYS
SNP: rs387906852, ClinVar: RCV000023245

In 2 unrelated families of European descent with autosomal dominant thoracic aortic and other aneurysms (LDS3; 613795), Regalado et al. (2011) identified a G-to-A transition at nucleotide 836 in exon 6 of the SMAD3 gene, resulting in an arg-to-lys substitution at codon 279 (R279K). Arg279 is completely conserved from human to Drosophila, and the R279K mutation was predicted to disrupt protein function. The mutation was not identified in 2,300 control exomes. There was decreased penetrance in younger family members.


.0006   LOEYS-DIETZ SYNDROME 3

SMAD3, GLU239LYS
SNP: rs387906853, ClinVar: RCV000023246, RCV001703421, RCV003528136

In a small family with 3 sibs affected with thoracic aortic aneurysm and dissection (LDS3; 613795), Regalado et al. (2011) identified a G-to-A transition at nucleotide 715 in exon 6 of the SMAD3 gene, resulting in a glutamine-to-lysine substitution at codon 239 (E239K). Exon 6 encodes the MH2 protein-protein binding domain. Glu239 is completely conserved from human to Drosophila, and the E239K mutation was predicted to disrupt protein function. The mutation was not identified in 2,300 control exomes.


.0007   LOEYS-DIETZ SYNDROME 3

SMAD3, ALA112VAL
SNP: rs387906854, ClinVar: RCV000023247, RCV002313719

In a family segregating autosomal dominant thoracic aortic aneurysm with dissection as well as other features of Loeys-Dietz syndrome (LDS3; 613795) including bifid uvula and scoliosis, and early-onset osteoarthritis, Regalado et al. (2011) identified a heterozygous alanine-to-valine substitution at codon 112 (A112V). The mutation segregated with disease with reduced penetrance in this family and was not identified in 2,300 control exomes. Guo (2012) stated that the correct nucleotide change for the A112V mutation is 335C-T in exon 2 rather than 235C-T as cited in Regalado et al. (2011).


.0008   LOEYS-DIETZ SYNDROME 3

SMAD3, 1-BP DEL, 313G
SNP: rs587776882, ClinVar: RCV000023248, RCV000767318

In a patient with aneurysms-osteoarthritis syndrome (LCS3; 613795), van de Laar et al. (2012) identified a 1-bp deletion at nucleotide 313 of the SMAD3 gene (313delG), resulting in a frameshift (Ala105ProfsTer11).


.0009   LOEYS-DIETZ SYNDROME 3

SMAD3, PRO263LEU
SNP: rs387906855, ClinVar: RCV000023249, RCV002313720

In a patient with aneurysms-osteoarthritis syndrome (LCS3; 613795), van de Laar et al. (2012) identified a 788C-T transition in the SMAD3 gene, resulting in a pro263-to-leu (P263L) substitution.


.0010   LOEYS-DIETZ SYNDROME 3

SMAD3, GLU361TER
SNP: rs387906856, ClinVar: RCV000023250

In affected members of a family segregating aneurysms-osteoarthritis syndrome (LCS3; 613795), van de Laar et al. (2012) identified a 1-bp duplication at nucleotide 1080 in the SMAD3 gene (1080dupT), resulting in a glu361-to-ter (E361X) substitution.


REFERENCES

  1. Arai, T., Akiyama, Y., Okabe, S., Ando, M., Endo, M., Yuasa, Y. Genomic structure of the human Smad3 gene and its infrequent alterations in colorectal cancers. Cancer Lett. 122: 157-163, 1998. [PubMed: 9464505] [Full Text: https://doi.org/10.1016/s0304-3835(97)00384-4]

  2. Arany, P. R., Flanders, K. C., Kobayashi, T., Kuo, C. K., Stuelten, C., Desai, K. V., Tuan, R., Rennard, S. I., Roberts, A. B. Smad3 deficiency alters key structural elements of the extracellular matrix and mechanotransduction of wound closure. Proc. Nat. Acad. Sci. 103: 9250-9255, 2006. [PubMed: 16754864] [Full Text: https://doi.org/10.1073/pnas.0602473103]

  3. Ashcroft, G. S., Yang, X., Glick, A. B., Weinstein, M., Letterio, J. L., Mizel, D. E., Anzano, M., Greenwell-Wild, T., Wahl, S. M., Deng, C., Roberts, A. B. Mice lacking Smad3 show accelerated wound healing and an impaired local inflammatory response. Nature Cell Biol. 1: 260-266, 1999. [PubMed: 10559937] [Full Text: https://doi.org/10.1038/12971]

  4. Bertero, A., Brown, S., Madrigal, P., Osnato, A., Ortmann, D., Yiangou, L., Kadiwala, J., Hubner, N. C., de los Mozos, I. R., Sadee, C., Lenaerts, A.-S., Nakanoh, S., Grandy, R., Farnell, E., Ule, J., Stunnenberg, H. G., Mendjan, S., Vallier, L. The SMAD2/3 interactome reveals that TGF-beta controls m6A mRNA methylation in pluripotency. Nature 555: 256-259, 2018. [PubMed: 29489750] [Full Text: https://doi.org/10.1038/nature25784]

  5. Carlson, M. E., Hsu, M., Conboy, I. M. Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells. Nature 454: 528-532, 2008. Note: Erratum: Nature 538: 274 only, 2016. [PubMed: 18552838] [Full Text: https://doi.org/10.1038/nature07034]

  6. Chen, C.-R., Kang, Y., Siegel, P. M., Massague, J. E2F4/5 and p107 as Smad cofactors linking the TGF-beta receptor to c-myc repression. Cell 110: 19-32, 2002. [PubMed: 12150994] [Full Text: https://doi.org/10.1016/s0092-8674(02)00801-2]

  7. Chuderland, D., Konson, A., Seger, R. Identification and characterization of a general nuclear translocation signal in signaling proteins. Molec. Cell 31: 850-861, 2008. [PubMed: 18760948] [Full Text: https://doi.org/10.1016/j.molcel.2008.08.007]

  8. Davis, B. N., Hilyard, A. C., Lagna, G., Hata, A. SMAD proteins control DROSHA-mediated microRNA maturation. Nature 454: 56-61, 2008. [PubMed: 18548003] [Full Text: https://doi.org/10.1038/nature07086]

  9. Dong, C., Zhu, S., Wang, T., Yoon, W., Li, Z., Alvarez, R. J., ten Dijke, P., White, B., Wigley, F. M., Goldschmidt-Clermont, P. J. Deficient Smad7 expression: a putative molecular defect in scleroderma. Proc. Nat. Acad. Sci. 99: 3908-3913, 2002. Note: Retraction: Proc. Nat. Acad. Sci. 113: E2208, 2016. [PubMed: 11904440] [Full Text: https://doi.org/10.1073/pnas.062010399]

  10. Guo, D.-C. Personal Communication. Houston, Tex. 2/7/2012.

  11. Gupta, A., Gartner, J. J., Sethupathy, P., Hatzigeorgiou, A. G., Fraser, N. W. Anti-apoptotic function of a microRNA encoded by the HSV-1 latency-associated transcript. Nature 442: 82-85, 2006. Note: Retraction: Nature 451: 600 only, 2008. [PubMed: 16738545] [Full Text: https://doi.org/10.1038/nature04836]

  12. Inman, G. J., Nicolas, F. J., Hill, C. S. Nucleocytoplasmic shuttling of Smads 2, 3, and 4 permits sensing of TGF-beta receptor activity. Molec. Cell 10: 283-294, 2002. [PubMed: 12191474] [Full Text: https://doi.org/10.1016/s1097-2765(02)00585-3]

  13. Kanamaru, Y., Sumiyoshi, K., Ushio, H., Ogawa, H., Okumura, K., Nakao, A. Smad3 deficiency in mast cells provides efficient host protection against acute septic peritonitis. J. Immun. 174: 4193-4197, 2005. [PubMed: 15778380] [Full Text: https://doi.org/10.4049/jimmunol.174.7.4193]

  14. Liu, T., Zhao, M., Liu, J., He, Z., Zhang, Y., You, H., Huang, J., Lin, X., Feng, X.-H. Tumor suppressor bromodomain-containing protein 7 cooperates with Smads to promote transforming growth factor-beta responses. Oncogene 36: 362-372, 2017. [PubMed: 27270427] [Full Text: https://doi.org/10.1038/onc.2016.204]

  15. Matsuura, I., Denissova, N. G., Wang, G., He, D., Long, J., Liu, F. Cyclin-dependent kinases regulate the antiproliferative function of Smads. Nature 430: 226-231, 2004. [PubMed: 15241418] [Full Text: https://doi.org/10.1038/nature02650]

  16. Regalado, E., Medrek, S., Tran-Fadulu, V., Guo, D.-C., Pannu, H., Golabbakhsh, H., Smart, S., Chen, J. H., Shete, S., Kim, D. H., Stern, R., Braverman, A. C., Milewicz, D. M. Autosomal dominant inheritance of a predisposition to thoracic aortic aneurysms and dissections and intracranial saccular aneurysms. Am. J. Med. Genet. 155A: 2125-2130, 2011. [PubMed: 21815248] [Full Text: https://doi.org/10.1002/ajmg.a.34050]

  17. Regalado, E. S., Guo, D., Villamizar, C., Avidan, N., Gilchrist, D., McGillivray, B., Clarke, L., Bernier, F., Santos-Cortez, R. L., Leal, S. M., Bertoli-Avella, A. M., Shendure, J., Rieder, M. J., Nickerson, D. A., NHLBI GO Exome Sequencing Project, Milewicz, D. M. Exome sequencing identifies SMAD3 mutations as a cause of familial thoracic aortic aneurysm and dissection with intracranial and other arterial aneurysms. Circ. Res. 109: 680-686, 2011. [PubMed: 21778426] [Full Text: https://doi.org/10.1161/CIRCRESAHA.111.248161]

  18. Riggins, G. J., Thiagalingam, S., Rozenblum, E., Weinstein, C. L., Kern, S. E., Hamilton, S. R., Willson, J. K. V., Markowitz, S. D., Kinzler, K. W., Vogelstein, B. Mad-related genes in the human. Nature Genet. 13: 347-349, 1996. [PubMed: 8673135] [Full Text: https://doi.org/10.1038/ng0796-347]

  19. Roth, S., Johansson, M., Loukola, A., Peltomaki, P., Jarvinen, H., Mecklin, J.-P., Aaltonen, L. A. Mutation analysis of SMAD2, SMAD3, and SMAD4 genes in hereditary non-polyposis colorectal cancer. J. Med. Genet. 37: 298-300, 2000. [PubMed: 10819637] [Full Text: https://doi.org/10.1136/jmg.37.4.298]

  20. Sato, M., Muragaki, Y., Saika, S., Roberts, A. B., Ooshima, A. Targeted disruption of TGF-beta-1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction. J. Clin. Invest. 112: 1486-1494, 2003. [PubMed: 14617750] [Full Text: https://doi.org/10.1172/JCI19270]

  21. Shattuck, T. M., Costa, J., Bernstein, M., Jensen, R. T., Chung, D. C., Arnold, A. Mutational analysis of Smad3, a candidate tumor suppressor implicated in TGF-beta and menin pathways, in parathyroid adenomas and enteropancreatic endocrine tumors. J. Clin. Endocr. Metab. 87: 3911-3914, 2002. [PubMed: 12161532] [Full Text: https://doi.org/10.1210/jcem.87.8.8707]

  22. van de Laar, I. M. B. H., Oldenburg, R. A., Pals, G., Roos-Hesselink, J. W., de Graaf, B. M., Verhagen, J. M. A., Hoedemaekers, Y. M., Willemsen, R., Severijnen, L.-A., Venselaar, H., Vriend, G., Pattynama, P. M., and 14 others. Mutations in SMAD3 cause a syndrome form of aortic aneurysms and dissections with early-onset osteoarthritis. Nature Genet. 43: 121-126, 2011. [PubMed: 21217753] [Full Text: https://doi.org/10.1038/ng.744]

  23. van de Laar, I. M. B. H., van der Linde, D., Oei, E. H. G., Bos, P. K., Bessems, J. H., Bierma-Zeinstra, S. M., van Meer, B. L., Pals, G., Oldenburg, R. A., Bekkers, J. A., Moelker, A., de Graaf, B. M., and 17 others. Phenotypic spectrum of the SMAD3-related aneurysms-osteoarthritis syndrome. J. Med. Genet. 49: 47-57, 2012. [PubMed: 22167769] [Full Text: https://doi.org/10.1136/jmedgenet-2011-100382]

  24. Wolfraim, L. A., Fernandez, T. M., Mamura, M., Fuller, W. L., Kumar, R., Cole, D. E., Byfield, S., Felici, A., Flanders, K. C., Walz, T. M., Roberts, A. B., Aplan, P. D., Balis, F. M., Letterio, J. J. Loss of Smad3 in acute T-cell lymphoblastic leukemia. New Eng. J. Med. 351: 552-559, 2004. [PubMed: 15295048] [Full Text: https://doi.org/10.1056/NEJMoa031197]

  25. Yang, X., Letterio, J. J., Lechleider, R. J., Chen, L., Hayman, R., Gu, H., Roberts, A. B., Deng, C. Targeted disruption of SMAD3 results in impaired mucosal immunity and diminished T cell responsiveness to TGF-beta. EMBO J. 18: 1280-1291, 1999. [PubMed: 10064594] [Full Text: https://doi.org/10.1093/emboj/18.5.1280]

  26. You, L., Kruse, F. E. Differential effect of activin A and BMP-7 on myofibroblast differentiation and the role of the Smad signaling pathway. Invest. Ophthal. Vis. Sci. 43: 72-81, 2002. [PubMed: 11773015]

  27. Zawel, L., Dai, J. L., Buckhaults, P., Zhou, S., Kinzler, K. W., Vogelstein, B., Kern, S. E. Human Smad3 and Smad4 are sequence-specific transcription activators. Molec. Cell 1: 611-617, 1998. [PubMed: 9660945] [Full Text: https://doi.org/10.1016/s1097-2765(00)80061-1]

  28. Zhang, Y., Feng, X.-H., Wu, R.-Y., Derynck, R. Receptor-associated Mad homologues synergize as effectors of the TGF-beta response. Nature 383: 168-172, 1996. [PubMed: 8774881] [Full Text: https://doi.org/10.1038/383168a0]

  29. Zhu, Y., Richardson, J. A., Parada, L. F., Graff, J. M. Smad3 mutant mice develop metastatic colorectal cancer. Cell 94: 703-714, 1998. [PubMed: 9753318] [Full Text: https://doi.org/10.1016/s0092-8674(00)81730-4]


Contributors:
Bao Lige - updated : 06/28/2019
Ada Hamosh - updated : 08/13/2018
Marla J. F. O'Neill - updated : 6/14/2016
Ada Hamosh - updated : 2/6/2012
Ada Hamosh - updated : 9/26/2011
Marla J. F. O'Neill - updated : 3/7/2011
Patricia A. Hartz - updated : 5/29/2009
Ada Hamosh - updated : 8/29/2008
Ada Hamosh - updated : 8/13/2008
Ada Hamosh - updated : 4/4/2008
Paul J. Converse - updated : 11/1/2006
Marla J. F. O'Neill - updated : 7/28/2006
Patricia A. Hartz - updated : 7/20/2006
Ada Hamosh - updated : 9/29/2004
Victor A. McKusick - updated : 9/13/2004
Ada Hamosh - updated : 8/26/2004
Cassandra L. Kniffin - updated : 12/4/2003
John A. Phillips, III - updated : 4/8/2003
Stylianos E. Antonarakis - updated : 9/11/2002
Stylianos E. Antonarakis - updated : 7/26/2002
Jane Kelly - updated : 7/8/2002
Victor A. McKusick - updated : 4/25/2002
Michael J. Wright - updated : 1/8/2001
Ada Hamosh - updated : 8/31/2000
Stylianos E. Antonarakis - updated : 1/31/1999
Stylianos E. Antonarakis - updated : 10/13/1998

Creation Date:
Patti M. Sherman : 10/9/1998

Edit History:
carol : 01/08/2020
mgross : 06/28/2019
alopez : 08/13/2018
carol : 08/17/2017
alopez : 12/19/2016
carol : 06/20/2016
carol : 6/17/2016
carol : 6/14/2016
carol : 11/14/2014
alopez : 4/22/2014
carol : 9/6/2012
carol : 2/27/2012
terry : 2/7/2012
carol : 2/7/2012
terry : 2/6/2012
alopez : 10/24/2011
alopez : 10/5/2011
terry : 9/26/2011
carol : 3/7/2011
terry : 3/7/2011
terry : 6/3/2009
mgross : 6/2/2009
terry : 5/29/2009
alopez : 9/11/2008
terry : 8/29/2008
alopez : 8/20/2008
terry : 8/13/2008
alopez : 4/14/2008
terry : 4/4/2008
wwang : 12/28/2007
terry : 12/11/2007
mgross : 11/7/2006
terry : 11/1/2006
wwang : 8/7/2006
terry : 7/28/2006
mgross : 7/20/2006
carol : 4/28/2005
carol : 4/28/2005
mgross : 4/13/2005
terry : 9/29/2004
tkritzer : 9/14/2004
terry : 9/13/2004
tkritzer : 8/30/2004
terry : 8/26/2004
carol : 12/8/2003
ckniffin : 12/4/2003
cwells : 4/29/2003
terry : 4/8/2003
mgross : 9/11/2002
mgross : 7/26/2002
mgross : 7/26/2002
mgross : 7/8/2002
mgross : 4/25/2002
alopez : 1/8/2001
alopez : 9/5/2000
terry : 8/31/2000
carol : 1/31/1999
carol : 10/13/1998
carol : 10/13/1998