Sei sulla pagina 1di 11

J Am Soc Nephrol 13: 2600–2610, 2002

REVIEW

TGF-ß Signaling in Renal Disease


ERWIN P. BÖTTINGER and MARKUS BITZER
Unified Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine and Montefiore
Medical Center, Department of Molecular Genetics, Albert Einstein College of Medicine, Bronx, New York.

Abstract. Since discovery over a decade ago of a role for the evidence in support of nonlinear models and functional roles of
cytokine TGF-ß as key mediator of glomerular and tubuloin- TGF-ß signaling in mediating apoptosis and epithelial-to-mes-
terstitial pathobiology in chronic kidney diseases, studies of enchymal transdifferentiation (EMT) in chronic progressive
TGF-ß signaling in the kidney have focused on the molecular renal disease. Emphasis is placed on cell context-dependent
biology of fibrogenesis. In recent years, glomerular and tubular models of TGF-ß signaling providing a conceptual framework
epithelial cell apoptosis and cellular transdifferentiation have to consolidate seemingly distinct pathomechanisms of progres-
been proposed as putative primary pathomechanisms that may sion of glomerular and tubulointerstitial disease.
underlie progression of renal disease. This review describes

The progression of chronic renal diseases is an increasingly remodeling, depending on physiological context. In light of
common condition, often leading to complete destruction of this multifunctionality, it is not surprising that an astonishing
functional kidney tissue and dependency of affected individu- array of different cytoplasmic and nuclear proteins and mech-
als on life-long treatments with dialysis or renal allograft anisms have been discovered during recent years, which are
transplantation (1). Pioneering studies of Border et al. (2– 4), exerting direct or indirect agonist or antagonist transmodula-
Ziyadeh and Sharma (5,6), and many others (reviewed in tion on the central TGF-ß/Smad signaling axis. In addition, it
reference 7) indicate a central role for TGF-ß and its down- is increasingly apparent that TGF-ß receptors can activate
stream signaling cascades in activating cellular pathomecha- Smad-independent signaling mechanisms, although it remains
nisms that underlie the progression of renal diseases. In general unclear how non-Smad pathways are connected to TGF-ß
terms, the TGF-ß superfamily consists of secreted peptides, of receptors at a molecular level (11). A comprehensive overview
which the three TGF-ß isoforms (TGF-ß1, -␤2, and -␤3), of the emerging complexity of TGF-ß/Smad signaling
activins, and bone morphogenetic proteins (BMP) are best transmodulators is shown in Figure 2, which is extensively
known in mammalian development, homeostasis, and pathobi- annotated in the corresponding figure legend. Discussion of
ology. The TGF-ß isoforms are widely expressed and act on molecular details of individual TGF-ß/Smad transmodulator
virtually every cell type in mammals by engaging a ubiquitous action is clearly beyond the scope and intent of this review.
intracellular signaling cascade of SMAD family proteins The interested reader is referred to outstanding general re-
through ligand-induced activation of heteromeric transmem- views, emphasizing molecular details of the emerging com-
brane TGF-ß receptor kinases. Receptor-activated Smad pro- plexity and context-dependence of TGF-ß/Smad signaling (see
tein complexes accumulate in the nucleus, where they partic- references 12–15). A major focus of the present review is to
ipate directly in transcriptional activation of target genes. evaluate critically the TGF-ß/Smad signaling system in the
Figure 1 shows the basic TGF-ß/Smad signaling axis. context of renal disease progression, not as a monolithic, linear
Although this seemingly simple linear model has emerged in inducer of fibrogenesis, but rather as a segment of dynamically
recent years as a central view of the TGF-ß/Smad signaling regulated, versatile signaling networks.
axis (8), it has become clear that it is insufficient to explain the
extensive and context-dependent multifunctionality of TGF-ß Reevaluating Paradigms of Renal Disease Progression
that is a hallmark of these peptides (9,10). Thus, TGF-ß are Excessive renal fibrogenesis, the process leading to tissue
considered important regulators of cell proliferation, differen- fibrosis, is considered a dominant pathomechanism induced by
tiation, apoptosis, immune response, and extracellular matrix TGF-ß in the kidney, largely on the basis of the observation
that glomerular and tubulointerstitial scarring are universal
outcomes of renal disease progression. In part, the current
Correspondence to Dr. Erwin P. Böttinger, Albert Einstein College of
scientific focus on renal fibrogenesis may be attributable to the
Medicine, Jack and Pearl Resnick Research Campus, 1300 Morris Park
Ave, Bronx, NY 10461. Phone: 718 430-3158; Fax: 718 430-8963; E-mail: availability of molecular targets, i.e., extracellular matrix com-
bottinge@aecom.yu.edu ponents, and assays that allow renal researchers to monitor
1046-6673/1310-2600 fibrotic reactions easily at a molecular and whole organ level.
Journal of the American Society of Nephrology In contrast, it has been more challenging to examine cellular
Copyright © 2002 by the American Society of Nephrology pathomechanisms, such as apoptosis, proliferation, and trans-
DOI: 10.1097/01.ASN.0000033611.79556.AE differentiation, largely because chronic progressive kidney dis-
J Am Soc Nephrol 13: 2600–2610, 2002 TGF-ß Signaling 2601

Figure 1. A simplified cartoon of the SMAD protein family and the basic TGF-ß/Smad signaling axis. TGF-ß ligand-binding with type II
receptor (RII) serine/threonine kinases (constitutively active) induces recruitment of type I receptor (RI) kinases into a heteromeric
ligand-receptor complex, leading to activation of RI kinase. The activated RI then signals to SMAD family members by phosphorylation of
two COOH-terminal serine residues, which are conserved in the receptor-regulated subgroup of Smads, R-Smads (Smad1, 2, 3, 5, and 8).
Smad1, 5, and 8 are substrates of bone morphogenetic proteins (BMP) receptor kinases. Smad2 and 3 are substrates of TGF-ß and activin
receptor kinases. Phospho-Smads interact with cytoplasmic common-partner Smad (Co-Smad), Smad4, which lacks COOH-terminal serines
and is not phosphorylated. R-Smad/Co-Smad complexes translocate to the nucleus and bind to specific DNA sequence motifs in target genes
to participate in transcriptional activation. R-Smads and Co-Smads are characterized by two major domains of homology, MH1 and MH2,
which are connected by a variable linker region. A subfamily of inhibitory Smads (I-Smads) lacks MH1 domains and consists of Smad6 and
Smad7, which inhibit TGF-ß/Smad signaling.

ease in humans frequently is a protracted, focal, and/or seg- proposed, stating that abnormal post-glomerular hypertension
mental process, making it difficult to detect significant single and vasoconstriction reduce peritubular capillary blood flow
cell alterations. However, along with readily apparent renal and cause rarification of peritubular capillaries, resulting in
scarring, a striking phenotype of chronic progressive kidney local hypoxia and tubular atrophy (19). Other reports suggest
disease is the disappearance of differentiated glomerular, tu- that tubular atrophy and interstitial fibrosis may be induced by
bular, and vascular cells that constitute the normal nephron increased protein content of ultrafiltrate, giving rise to an
(16). Thus, tubular cell atrophy and dilatation are invariably inflammatory reaction and interstitial fibrosis (20,21). Upregu-
associated with progressive nephron loss and onset of tubulo- lation of TGF-ß is consistently associated with these post-
interstitial fibrosis, a robust histopathologic predictor of glomerular events, irrespective of the proposed model of
chronic progressive renal disease (17,18). In addition, loss of pathogenesis (22–24).
peritubular capillaries is noted in progressive renal disease in Sclerosing glomeruli are characterized by progressive deple-
humans (18). Indeed, a chronic hypoxia hypothesis has been tion of podocytes (16,25) and striking loss of glomerular cap-
2602 Journal of the American Society of Nephrology J Am Soc Nephrol 13: 2600–2610, 2002

Figure 2. Mediator and modifier steps regulating the TGF-ß/Smad signaling axis. Green arrows indicate agonist, and red arrows/lines indicate antagonist
function of the depicted steps, respectively. Upon activation of latent TGF-ß complexes and release of TGF-ß ligand in the extracellular space, ligand binding
induces heteromeric TGF-ß receptor complexes and type I receptor kinase activity. Recruitment and C-terminal phosphorylation of cytoplasmic R-Smads,
Smad2 and Smad3, by activated TßRI is regulated by cytoskeletal proteins and multiple proteins with scaffold, adaptor, anchoring, and/or chaperone function
(pink boxes). Green lettering and (⫹) indicate positive regulation. Red lettering and (⫺) indicate negative regulation of TGF-ß/Smad signaling. Activated
R-Smads and cytoplasmic Co-Smad Smad4 form heteromeric complexes that translocate to the nucleus via nuclear transporters (yellow box). Nuclear Smad
complexes bind to consensus Smad3/4 DNA binding elements (SBE) (beige box) in TGF-ß/Smad immediate-early target genes. Example SBE core sequences
and their respective positions in select TGF-ß target gene promoters are shown (beige box). Smads regulate target gene transcription together with nuclear
transcription factors (brown box), co-activators (grey box), and/or co-repressors (olive box). A list of gene symbols for new immediate-early TGF-ß target
genes, identified by microarray screens (11), is shown (green box). Gene symbols can be searched in NCBI Unigene (http://www.ncbi.nlm.nih.gov/UniGene/).
Gene activation may be limited and/or terminated by targeting of nuclear R-Smads for proteasome degradation via E2 or E3 enzyme complexes (light blue
box). Cytokine and TGF-ß–inducible I-Smads inhibit the TGF-ß/Smad signaling axis by competitive interaction with R-Smads and TßRI, and/or by
recruitment of Smurf ubiquitin-ligases (light blue boxes) to activated TßR complexes, leading to its removal via proteasomal degradation. Receptor tyrosine
kinase–activated MAPK Erk may phosphorylate motifs in the linker regions of R-Smads to prevent nuclear translocation of activated Smad complexes and
inhibit the TGF-ß/Smad signaling axis. Extracellular modulators: TSP1, thrombospondin 1; LAP, latency-associated peptide; IFN-␥, interferon ␥; TNF-␣,
tumor necrosis factor ␣; IL-1ß, interleukin 1ß; HGF, hepatocyte growth factor; EGF, epidermal growth factor. Cross-regulating signal transducers: Stat1,
signal transducer and activator of transcription 1; NF-␬B, nuclear factor–␬B; Erk, extracellular signal-regulated kinase. Chaperones/Anchors/Adaptors:
SARA, Smad anchor for receptor activation; Hrs/Hgs, hepatic growth factor-regulated tyrosine kinase substrate; Cav-1, caveolin-1; Dab-2, disabled-2; SNIX,
sorting nexin; STRAP, serine-threonine kinase receptor-associated protein; TRAP-1, TGF-ß receptor-associated protein-1. Nuclear transporters: Crm1,
chromosome region maintenance 1. Ubiquitin ligases: Ubc3, ubiquitin conjugating enzyme 3; UbcH5b/c, ubiquitin conjugating enzyme H5b/c; ROC1-
SCF(Fbw1a): ROC1, Skp1, Cullin1, and Fbw1a complex. Transcription factors: AR, androgen receptor; ATF-2, activating transcription factor-2; BF-1, brain
factor-1; E1A, early region 1A; ER, estrogen receptor; Evi-1, ectopic viral integration site-1; FAST/FoxH1, forkhead activin signal transducer; Gli3,
glioblastoma gene product 3; GR, glucocorticoid receptor; HNF4, hepatocyte nuclear factor 4; LEF/TCF, lymphoid enhancer factor/T-cell factor; MEF2,
myocyte enhancer-binding factor 2; Menin, multiple endocrine neoplasia-type 1 tumor suppressor protein; Miz1, Myc interacting zinc finger protein 1; OAZ,
Olf-1/EBF associated zinc-finger ; PEBP2/CBFA/AML, polyoma-virus-enhancer-binding protein/core-binding factor A/acute myeloid leukemia; SNIP1,
Smad nuclear interacting protein 1; TFE3, transcription factor mu E3; Sp1, specificity factor 1; Sp3, specificity factor 3; VDR, vitamin D receptor.
Co-Activators: MSG1, melanocyte-specific gene 1; P/CAF, p300/CBP-associated factor. Co-Repressors: SnoN, ski-related novel gene; TGIF, TG-interacting
factor; HDACs, histone deacetylases; Ski, Sloan-Kettering Institute proto-oncogene.
J Am Soc Nephrol 13: 2600–2610, 2002 TGF-ß Signaling 2603

illaries associated with depletion of endothelial cells (26 –28). tubulointerstitial lesions in TGF-ß1 transgenic mice, a model
Kriz et al. (25) proposed that podocyte depletion and/or de- of progressive glomerulosclerosis and tubulointerstitial fibrosis
tachment lead to subsequent synechiae formation and tuft (40,41), indicates that increased tubular cell apoptosis precedes
adhesions as initial lesions of glomerular injury, consistent manifestations of tubular atrophy, tubular dilatation, and
with the frequently observed segmental nature of early glomer- perivascular inflammation [Wenjun Ju, Markus Bitzer, and
ular disease. Their concept of podocyte depletion as initiating Erwin Böttinger; personal communication, August 2002].
lesion in glomerulosclerosis is further supported by observa- Work by Neilson and colleagues and others (42– 44) suggests
tions in humans with diabetes. Podocyte numbers are reduced that tubular epithelial cells may transdifferentiate to acquire
in otherwise normal appearing glomeruli and predict long-term (myo)fibroblast phenotypes associated with interstitial fibrosis
urinary albumin excretion (29). Together, these phenotypic in experimental models and human renal biopsies (45). Be-
observations suggest that segmental loss of differentiated cause TGF-ß is a well-known inducer of epithelial-to-mesen-
podocytes and tubular and microvascular endothelial cells may chymal transdifferentiation in several organs (11,46,47), it is
be initial and irreversible lesions in progressive nephron loss. perhaps not surprising that recent reports also implicate a direct
Thus, we propose that a comprehensive model of progression role for TGF-ß in mediating EMT of renal tubular epithelial
of renal disease must provide pathomechanisms to account for cells in vitro and in vivo (48,49).
progressive loss of differentiated renal cells.
The common fibrocentric paradigm hypothesizes that fibro- TGF-ß and Podocyte Depletion
sis is the primary pathomechanism mediating renal disease As described before, glomerulosclerosis in animal models
progression and that the primary pathogenetic role of TGF-ß and humans is characterized in part by depletion of visceral
signaling lies in promotion of fibrogenesis (30). However, in epithelial cells (podocytes) (25,50). Mechanical detachment of
the context of the aforementioned observations, this paradigm podocytes from glomerular basement membranes (GBM) and
is apparently insufficient in that it fails to provide molecular loss in urinary space, possibly due to altered cell adhesion
and cellular links to explain the loss of differentiated renal and/or increased mechanical stress of injured podocytes, has
cells. There is no direct experimental evidence to support the been proposed as a potential mechanism. In addition, it is
central tenet of the fibrocentric paradigm, namely that ex- thought that podocytes are unable to proliferate and to replace
panded and/or altered extracellular matrix leads to demise of the “lost” podocytes in most forms of glomerular injury, lead-
differentiated renal cells and ultimately organ dysfunction. ing to a state of relative podocyte insufficiency (25,51,52). We
Interestingly, recent analyses of a classical condition of exces- have shown that TGF-ß and Smad7 synergize to induce apo-
sive scarring, systemic sclerosis, suggest that autoantibody- ptosis in podocytes in vitro (53). Our in vivo studies indicate
mediated endothelial cell apoptosis is a primary pathomecha- that time of peak podocyte apoptosis coincides with expression
nism leading to secondary sclerotic skin and organ destruction of TGF-ß1 and Smad7, and with the onset of albuminuria, but
(31,32). Other conditions associated with excessive scarring, precedes mesangial expansion in a TGF-ß1 transgenic model
such as posttraumatic hypertrophic scars and desmoplastic of progressive glomerulosclerosis (53). Recent results from our
tumors, are characterized by fibroplasia and increased vascu- lab, obtained in CD2AP knockout mice, a new murine model
larization, indicating that extracellular matrix expansion per se of focal segmental glomerulosclerosis (54), are similar to and
is not sufficient to induce atrophy and/or depletion of resident confirm these observations in a second, independent experi-
cells (33,34). If renal fibrosis is not causing nephron loss, what mental model of chronic progressive glomerulosclerosis
is the evidence for alternative pathomechanisms mediated by [Mario Schiffer, Andrey Shaw, and Erwin Böttinger; personal
TGF-ß? Interestingly, in contrast with renal research, studies of communication, August 2002].
TGF-ß/Smad signaling in non-renal diseases commonly exam-
ine cellular responses other then fibrogenesis, such as cell TGF-ß and Loss of Capillary Endothelial Cells
cycle control, apoptosis, and differentiation. Can nephron loss Progressive renal disease is characterized in part by a pro-
thus be explained in part on the basis of apoptosis and/or gressive loss of the glomerular and peritubular microvascula-
transdifferentiation induced by TGF-ß signaling? ture (27). Loss of glomerular capillaries is associated with
increased apoptosis of glomerular endothelial cells and corre-
TGF-ß and Tubular Atrophy lates with the development of glomerulosclerosis in rodent
There is indeed increasing evidence indicating a role for models of progressive anti-GBM disease, in remnant kidney
apoptosis in tubular epithelial cells, causing tubular atrophy in models, and in aging kidney (28,55,56). Similarly, endothelial
various experimental models and human forms of progressive cell (EC) apoptosis may underlie the loss of peritubular capil-
renal disease, including chronic obstructive nephropathy laries characteristically associated with tubulointerstitial fibro-
(CON), cyclosporine-induced allograft nephropathy, diabetic sis and tubular atrophy (57). Several reports demonstrate that
kidneys, and others (35–37). In these studies, TGF-ß1 expres- TGF-ß and thrombospondin 1 (TSP-1), a putative activator of
sion was associated with apoptotic tubular cells. Angiotensin II extracellular TGF-ß, induce apoptosis in microvascular endo-
blockade, or inhibition of TGF-ß using anti-TGF-ß antibodies, thelial cells, including glomerular endothelial cells (58,59). In
reduced tubular epithelial apoptosis and reduced the extent of addition, loss of paracrine signaling of angiogenic survival
tubular atrophy in models of CON and diabetic kidneys factor vascular endothelial growth factor (VEGF), acting on
(38,39). Interestingly, a detailed analysis from our group of endothelial cells, enhances EC susceptibility to undergo apo-
2604 Journal of the American Society of Nephrology J Am Soc Nephrol 13: 2600–2610, 2002

ptosis and causes EC growth arrest (27). Loss of expression of gressive glomerulosclerosis (Figure 3). Concomitant loss of
VEGF in glomerular diseases and progressive renal disease has glomerular capillaries may be a result of decreased EC survival
been described (27). VEGF is constitutively and selectively caused by direct, pro-apoptotic signaling networks of TGF-ß in
expressed in podocytes and tubular epithelial cells in normal EC and by a decrease of VEGF associated with progressive
kidneys (60,61); therefore, TGF–ß-induced depletion of podo- podocyte depletion (Figure 3).
cytes and/or tubular epithelial cells may be causing the de- This model of multiple pathogenetic events mediated by
creased VEGF expression. TGF-ß may thus indirectly promote TGF-ß further implies engagement of distinct signaling mod-
EC apoptosis by depleting VEGF-producing cell types in pro- ules/networks for the TGF-ß/Smad segment to specify pro-
gressive renal disease. apoptotic signals in podocytes, tubular epithelial cells, and/or
endothelial cells, as opposed to EMT signaling cascades in
Revised Model of TGF-ß-Mediated Pathomechanisms tubular epithelial cells or pro-fibrotic signals in mesenchymal
of Progressive Nephron Loss myofibroblasts and/or mesangial cells. The proposed shift of
On the basis of these observations, we propose a substan- functional endpoints of TGF-ß/Smad signaling from fibrosis to
tially revised model, including new functional roles and mul- apoptosis and EMT as putative primary pathomechanisms for
tiple pathogenetic endpoints, of TGF-ß signaling in renal dis- progressive nephron loss has important implications for future
ease progression (Figure 3). Whereas interstitial and mesangial experimental approaches and selection of therapeutic targets in
matrix expansion may result from direct activation of “fibro- progression of renal disease. In the remaining sections, we
genic” signaling networks by TGF-ß in mesangial cells and review recent advances in our understanding of distinct, re-
interstitial (myo)fibroblasts, additional primary pathomecha- sponse-specific signaling networks activated by TGF-ß/Smad
nisms mediated by TGF-ß signaling should include apoptosis to mediate EMT and apoptosis.
and EMT. Thus, TGF-ß signaling may initiate pro-apoptotic
effectors and/or EMT in tubular epithelial cells, resulting in TGF-ß Signaling Networks in Epithelial-to-
tubular degeneration and tubular atrophy (Figure 3). In addi- Mesenchymal Transdifferentiation
tion, TGF-ß–induced EMT may convert tubular epithelial cells Activation of TGF-ß signaling is sufficient to induce EMT in
into activated (myo)fibroblasts, which may be responsible for cultured epithelial cells, including a non-transformed mouse
increased deposition of interstitial matrix in response to TGF- mammary cell line (NMuMG) and human keratinocytes (Ha-
ß/Smad signaling (Figure 3). Decline of tubular VEGF expres- Cat) (11,46,62,63). A role for EMT in tubular atrophy and
sion as a result of tubular degeneration/atrophy may indirectly appearance of myofibroblasts in renal disease was first pro-
contribute to decreased EC survival in peritubular capillaries posed several years ago (64). However, evidence for TGF-ß as
and peritubular capillary loss (Figure 3). Thus, nonlinear, re- mediator of renal tubular EMT has only recently been reported
sponse-specific TGF-ß signaling networks may lead to tubu- (49,65). For example, advanced glycation end products (AGE)
lointerstitial fibrosis by inducing multiple pathogenetic pro- were found to induce EMT in vitro and in diabetic rats through
cesses in tubular and microvascular cells. In addition, we activation of TGF-ß signaling, indicating an important role for
propose that podocyte and endothelial cell apoptosis are di- this TGF-ß–induced response in progression of diabetic ne-
rectly induced by TGF-ß signaling in glomeruli exposed to phropathy (49). On the basis of recent studies of signaling
various forms of injury (Figure 3). Apoptosis of podocytes may pathways activated by TGF-ß to induce EMT in various types
lead to depletion of podocytes and formation of synechiae of epithelial cells, a model of this response-specific TGF-ß
between bare GBM and Bowman’s capsule. This results in signaling network is emerging (Figure 4). EMT is a coordi-
segmental tuft adhesions characteristic of early lesions in pro- nated cellular response that involves several distinct processes,
including disruption and disassembly of desmosomes and E-
cadherin adherens junctions, remodeling of actin cytoskeleton
and stress fiber formation, alteration of cell-matrix adhesion,
and increase in cell motility (Figure 4). To date, Smads have
been implicated in some aspects of TGF-ß–induced EMT. For
example, overexpression of Smad2, Smad3, and Smad4 in
NMuMG murine mammary gland epithelial cells induces stress
fiber formation (46). In addition, TGF-ß induces Net1A, a
RhoA-specific guanine exchange factor, in a Smad-dependent
manner (11,66). Net1A is required for TGF-ß–induced F-actin
remodeling (66). This is consistent with observations that
TGF-ß can rapidly induce the activation of the small GTPase
RhoA, a regulator of actin cytoskeleton and adhesion junctions
in NMuMG cells (67). Inhibitors of Rho kinase can block
F-actin remodeling and relocalization of E-cadherin in adher-
Figure 3. Profile of putative TGF-ß–mediated response-specific cel- ens junctions induced by TGF-ß (67). RhoA and phosphatidyl-
lular pathomechanisms in progressive nephron loss. See text for inositol 3-kinase (PI3K), signaling through the serine-threo-
details. nine kinase p160ROCK and Akt/PKB, are both required for
J Am Soc Nephrol 13: 2600–2610, 2002 TGF-ß Signaling 2605

Figure 4. Signaling modules and genetic programs underlying epithelial-to-mesenchymal transdifferentiation induced by TGF-␤. TGF-ß is
sufficient to induce characteristic processes of EMT, including desmosome disassembly, cell-matrix adhesion remodeling, E-cadherin adherens
junction disassembly, cell motility, F-actin remodeling (stress fiber formation), and activation of transitional progenitor cell factors. TGF-ß
receptor activated pathways/mediators implicated in EMT include Smads, MEK/ERK, Net1A, RhoA, PI3K, PKB/Akt, p160ROCK, and
hypothetical mediators (⫹/⫺ Other mediators) (shaded boxes). Genes induced by TGF-ß within 4 h (11) are denoted by Unigene Symbols
colored in red (searchable at http://www.ncbi.nlm.nih.gov/UniGene/). Genes and pathways are aligned according to time of peak activation.
Solid line arrows indicate pathways, programs and cellular responses suggested in reference 11. Broken line arrows show mediators and
dependent responses demonstrated in other reports. Dotted line arrows show putative pathways proposed in reference 11.

TGF-ß–induced disassembly of cell-cell junctions and F-actin TGF-ß [Jiri Zavadil, Lukas Cermak, and Erwin Böttinger;
remodeling (47,67). TGF-ß induces Smad-interacting zinc fin- personal communication, August 2002]. In contrast, chemical
ger protein SIP1 (68) and SLUG (11), transcriptional repres- inhibition of mitogen-activated protein kinase Erk (MAPK
sors of E-cadherin capable of mediating disassembly of cell Erk) blocks selectively TGF-ß–mediated regulation of genes
adherens junctions in tubular epithelial MDCK cells. We re- with functional roles in cell-matrix interactions, cell motility,
ported a microarray-based screen of transcriptional profiles of and endocytosis (11). This is consistent with functional inhi-
TGF-ß–induced EMT in human HaCat keratinocytes (11). bition of TGF-ß–induced disassembly of adherens junctions
These studies suggest a rapid (within 4 h) and dynamic change and cell motility in the presence of MEK/ERK inhibitor (11).
in expression of approximately 4000 transcripts, including Signaling networks underlying EMT in many tissues, in-
factors with roles in mesenchymal progenitor cell types, and cluding kidney, is a young field. Most studies of this cellular
Wnt and Notch signaling. Our recent results demonstrate that response are conducted in the context of embryonic develop-
functional inactivation of ␥-secretase, an activator of Notch ment and tumor invasiveness and metastasis formation. The
receptor, completely prevents EMT induced by TGF-ß, indi- described knowledge of TGF-ß signaling mediating EMT has
cating a role for Notch signaling components downstream of been obtained largely in these contexts. However, a detailed
2606 Journal of the American Society of Nephrology J Am Soc Nephrol 13: 2600–2610, 2002

understanding of the molecular signaling mechanisms that a tissue section may be associated with a significant loss of cell
mediate tubular epithelial cell EMT in nephrons exposed to mass (69,70). Interestingly, glomerular cell apoptosis is mark-
pathogenic stimuli will likely be essential for development of edly increased in renal biopsy specimen from patients with IgA
therapies that can prevent renal disease progression. nephropathy, with the greatest number, up to one apoptotic cell
per cross section, found in lesions described as “predominantly
TGF-ß Signaling Networks in Apoptosis sclerosing” (71,72). The number of apoptotic cells per glomer-
It has been difficult to establish apoptosis as a candidate ular cross-section correlates with semiquantitative “sclerosis”
pathomechanism mediated by TGF-ß in progression of renal scores in IgA nephropathy and lupus nephritis (73,74).
disease, because detection and reliable quantitation of apopto- TGF-ß can induce apoptosis in many cell types, including
tic cells in chronic progressive renal injury is complicated by renal cells (reviewed in references 53,75,76). A review of
several factors. First, the half-life of apoptotic cells is only a apoptotic signaling networks of TGF-ß is best guided by or-
few hours, limiting the window of detection. In contrast with ganization of apoptotic signaling in three phases (Figure 5). An
extracellular matrix components, cellular remnants of apopto- Initiation phase is triggered by stress signals and/or specific
sis are not accumulating in tissue but are often removed by factors (including TGF-ß) acting through a subset of receptors.
phagocytosis. In addition, detached apoptotic podocytes and During Initiation and Integration phase, cells balance signals
tubular epithelial cells may be flushed away by urine. As a from several signaling pathways (pro- and anti-apoptotic) to
consequence, a low percentage of apoptotic cells detectable in determine whether the Execution phase of cell death should be

Figure 5. Signaling networks mediating TGF-ß–induced apoptosis. Apoptosis signaling is comprised of three phases: Initiation, Integration, and
Execution. TGF-ß receptor activation (RI/RII) leads to activation of Smad3. Smad3 is required for upregulation of pro-apoptotic signaling
mediators death-associated protein kinase (DAPK), TGF-ß–induced early gene (TIEG), TGF-ß–stimulated clone 22 (TSC22), and Smad7.
Smad7 inhibits anti-apoptotic signaling by NF-␬B, and may be responsive to TGF-ß-independent signals from pro-apoptotic factors. Smad7
also activates pro-apoptotic JNK. TGF-ß–induced activation of pro-apoptotic MAPK p38 and JNK may be mediated by MAPK cascades via
hematopoietic progenitor kinase 1 (HPK1), TGF-ß–activated kinase 1 (TAK1), and TAK1 activator (TAB1). Daxx interacts with RII and
activates JNK. TGF-ß induces apoptosis in murine podocytes through activation of p38 and induction of pro-apoptotic protein Bad, leading to
activation of caspases. TGF-ß stimulates nuclear translocation of pro-apoptotic ARTS from mitochondria.
J Am Soc Nephrol 13: 2600–2610, 2002 TGF-ß Signaling 2607

activated. Critical regulators of the execution phase are the (53,89). Because Smad7 is regulated by TGF-ß in a Smad3-
Bcl2 family of proteins, which include anti-apoptotic (e.g., dependent manner (90), and by other extracellular stress and
Bcl-2, Bcl-XL) and pro-apoptotic (e.g., Bax, Bad, and Bid) receptor signals (91–93), it may have an important role in
members. Relative expression levels of Bcl proteins can trigger enhancing TGF-ß– dependent pro-apoptotic signaling, as well
the irreversible execution phase by regulating release of cyto- as inhibition of anti-apoptotic pathways independent of TGF-ß
chrome c from the outer mitochondrial membrane, leading to (93). A recent report by Larisch et al. (94) identifies a septin
activation of effector protease cascades of the caspase family. family protein, apoptosis-related protein in the TGF-ß signal-
There is growing evidence that Smad3 is an important sig- ing pathway (ARTS), which is required for TGF-ß–induced
naling anchor for apoptotic networks. Thus, TGF-ß–induced apoptosis in rat prostate cells. ARTS is normally localized to
apoptosis is enhanced by overexpression of R-Smads in some mitochondria and translocates to the nucleus when TGF-ß
cells (77). In addition, a number of target genes of Smad3 induces apoptosis. Together, these TGF-ß–regulated signaling
appear to mediate TGF-ß–induced apoptosis. Smad3 is re- mediators of apoptosis participate in the Integration and Exe-
quired for transcriptional activation of death-associated protein cution phases of apoptotic signaling networks by stimulating
kinase (DAPK), which is essential for TGF-ß–induced apopto- expression of pro-apoptotic members and by suppressing ex-
sis in human hepatoma cells and Burkitts Lymphoma cells pression of anti-apoptotic members of the Bcl protein family,
(78). We also found Smad3-dependent regulation of DAPK in respectively (95–97).
murine podocytes (Markus Bitzer and Erwin Böttinger; per-
sonal communication). In addition, activation of several target Concluding Remarks
genes of TGF-ß, including TGF-ß–induced early gene (TIEG) In this review, we attempt to provide a rationale for a revised
in mink lung epithelial and Hep3B cells, and TGF-ß–stimu- model of TGF-ß signaling in progressive renal disease. The
lated clone 22 (TSC-22) in gastric carcinoma cells (79 – 81) role of TGF-ß in renal fibrosis is widely accepted and targets
have been implicated in apoptosis. Because TGF-ß activates of fibrogenic signaling have been discussed (98,99); we have
transcription of these genes, it is likely that Smad3 is required therefore focused on TGF-ß–mediated apoptosis and EMT.
for these signals. In addition, TGF-ß–induced activation of Apoptosis and EMT are considered primary cellular patho-
pro-apoptotic Bcl member Bad in FaO rat hepatoma cells is mechanisms mediated by response-specific TGF-ß signaling
mediated by Smad3 (82). Daxx is a Fas-receptor–associated networks in various forms of tissue injury. We propose that
protein that can interact directly with TGF-ß type II receptor. these mechanisms may cause progressive loss of differentiated
Daxx activates the JNK pathway and mediates apoptosis in- renal cells, a hallmark of chronic progression of renal disease.
duced by Fas, but dominant negative mutant Daxx or func- TGF-ß–induced apoptosis is likely to have a pathogenetic role
tional inactivation with Daxx antisense oligonucleotides also in podocyte depletion and glomerulosclerosis, tubular degen-
inhibit TGF-ß–induced apoptosis in B cells and mouse hepa- eration/atrophy, and loss of glomerular and peritubular capil-
tocytes (83). Thus, Daxx may mediate a direct biochemical laries. In addition, EMT induced by TGF-ß may contribute to
connection between the TGF-ß receptors and the apoptotic tubular atrophy and generation of interstitial myofibroblasts,
signaling network. leading to concomitant tubulointerstitial fibrosis. The revised
Several reports implicate MAPK pathways in apoptotic sig- model of TGF-ß signaling in renal disease is intended to
naling by TGF-ß. Activation of TGF-ß–activated kinase-1 stimulate new perspectives on selection of scientific and ther-
(TAK-1), a protein of the MAP kinase kinase kinase family, apeutic approaches to understand and prevent chronic progres-
activates p38 and JNK signaling in TGF-ß family–induced sion of renal disease.
apoptosis (84). In addition, the upstream activator of TAK1,
TAB1, may link pro-apoptotic MAPK to TGF-ß receptors Acknowledgements
through receptor-mediated activation of hematopoietic progen- We thank members and technicians of our laboratory for
itor kinase 1 (HPK1) (85,86). p38 signaling is required for stimulating discussions and support. This work was supported
TGF-ß–induced apoptosis in murine podocytes (53). It is un- with funding from NIH grants RO1 DK56607 and RO1
clear how TGF-ß receptors signal to MAPK pathways or HPK1 DK60043 to Erwin P. Böttinger. Markus Bitzer is recipient of
to induce apoptosis. There is also evidence that Smad7 may a research fellowship from the National Kidney Foundation of
induce apoptosis by activation of JNK (87). Indeed, the I- New York/New Jersey.
Smad7 has been implicated in apoptosis signaling by TGF-ß
and TGF-ß–independent pathways. TGF-ß–stimulated expres- References
sion of Smad7 is required for induction of apoptosis via p38 1. Wolfe RA, Port FK, Webb RL, Bloembergen WE, Hirth R,
activation in prostatic carcinoma cells (88), indicating that Young EW, Ojo AO, Strawderman RL, Parekh R, Stack A,
Tedeschi PJ, Hulbert-Shearon T, Ashby VB, Callard S, Hanson
Smad7 may function downstream of TGF-ß in apoptosis sig-
J, Jain A, Meyers-Purkiss A, Roys E, Brown P, Wheeler JR,
naling networks. However, ectopic expression of Smad7 can Jones CA, Greer JW, Agodoa LY: Introduction to the excerpts
induce apoptosis independently of TGF-ß pathways in murine from the United States Renal Data System 1999 Annual Data
podocytes (53) and distal tubular epithelial MDCK cells (89). Report. Am J Kidney Dis 34: S1–S3, 1999
Both reports indicate that Smad7 inhibits nuclear translocation 2. Border WA, Okuda S, Languino LR, Ruoslahti E: Transforming
and transcriptional activator function of NF-␬B, a major acti- growth factor-beta regulates production of proteoglycans by
vator of transcription of genes with anti-apoptotic functions mesangial cells. Kidney Int 37: 689 – 695, 1990
2608 Journal of the American Society of Nephrology J Am Soc Nephrol 13: 2600–2610, 2002

3. Border WA, Okuda S, Languino LR, Sporn MB, Ruoslahti E: 25. Kriz W, Gretz N, Lemley KV: Progression of glomerular dis-
Suppression of experimental glomerulonephritis by antiserum eases: is the podocyte the culprit? Kidney Int 54:687– 697, 1998
against transforming growth factor beta 1. Nature 346: 371–374, 26. Lee LK, Meyer TW, Pollock AS, Lovett DH: Endothelial cell
1990 injury initiates glomerular sclerosis in the rat remnant kidney.
4. Border WA, Okuda S, Nakamura T, Languino LR, Ruoslahti E: J Clin Invest 96:953–964, 1995
Role of TGF-beta 1 in experimental glomerulonephritis. Ciba 27. Kang DH, Kanellis J, Hugo C, Truong L, Anderson S, Kerjaschki
Found Symp 157:178 – 89; discus, 1991 D, Schreiner GF, Johnson RJ: Role of the microvascular endo-
5. Sharma K, Ziyadeh FN, Alzahabi B, McGowan TA, Kapoor S, thelium in progressive renal disease. J Am Soc Nephrol 13:806 –
Kurnik BRC, Kurnik PB, Weisberg LS: Increased renal produc- 816, 2002
tion of transforming growth factor-b1 in patients with type II 28. Kang DH, Hughes J, Mazzali M, Schreiner GF, Johnson RJ:
diabetes mellitus. Diabetes 46:854 – 859, 1997 Impaired angiogenesis in the remnant kidney model: II. Vascular
6. Ziyadeh FN: Role of transforming growth factor beta in diabetic endothelial growth factor administration reduces renal fibrosis
nephropathy. Exp Nephrol 2:137, 1994 and stabilizes renal function. J Am Soc Nephrol 12:1448 –1457,
7. Bitzer M, Sterzel RB, Bottinger EP: Transforming growth factor- 2001
beta in renal disease. Kidney Blood Press Res 21:1–12, 1998 29. Meyer TW, Bennett PH, Nelson RG: Podocyte number predicts
8. Massague J: How cells read TGF-beta signals. Nat Rev Mol Cell long-term urinary albumin excretion in Pima Indians with Type
Biol 1:169 –178, 2000 II diabetes and microalbuminuria. Diabetologia 42:1341–1344,
9. Roberts AB, Sporn MB: The transforming growth factors-␤. In: 1999
Handbook of Experimental Pharmacology. Peptide Growth Fac- 30. Border WA, Noble NA: TGF-beta in kidney fibrosis: a target for
tors and Their Receptors., 95 edn., edited by Sporn MB, Roberts gene therapy. [Review] [40 refs]. Kidney Int 51:1388 –1396,
AB, Berlin Heidelberg New York London Paris Tokyo Hong 1997
Kong, Springer-Verlag, 1990, p 419 – 472. 31. Lunardi C, Bason C, Navone R, Millo E, Damonte G, Corrocher
10. Roberts AB: Molecular and cell biology of TGF-beta. Miner R, Puccetti A: Systemic sclerosis immunoglobulin G autoanti-
Electrolyte Metab 24:111–119, 1998 bodies bind the human cytomegalovirus late protein UL94 and
11. Zavadil J, Bitzer M, Liang D, Yang YC, Massimi A, Kneitz S, induce apoptosis in human endothelial cells. Nat Med 6:1183–
Piek E, Bottinger EP: Genetic programs of epithelial cell plas- 1186, 2000
ticity directed by transforming growth factor-beta. Proc Natl 32. Sgonc R: The vascular perspective of systemic sclerosis: of
Acad Sci U.S.A 98:6686 – 6691, 2001 chickens, mice and men. Int Arch Allergy Immunol 120:169 –
12. Roberts AB: The ever-increasing complexity of TGF-beta sig- 176, 1999
naling. Cytokine Growth Factor Rev 13:3–5, 2002 33. Tuan TL, Nichter LS: The molecular basis of keloid and hyper-
13. ten Dijke P, Goumans MJ, Itoh F, Itoh S: Regulation of cell trophic scar formation. Mol Med Today 4:19 –24, 1998
proliferation by Smad proteins. J Cell Physiol 191:1–16, 2002 34. Lieubeau B, Garrigue L, Barbieux I, Meflah K, Gregoire M: The
14. Derynck R, Akhurst RJ, Balmain A: TGF-beta signaling in tumor role of transforming growth factor beta 1 in the fibroblastic
suppression and cancer progression. Nat Genet 29:117–129, reaction associated with rat colorectal tumor development. Can-
2001 cer Res 54:6526 – 6532, 1994
15. Massague J, Chen YG: Controlling TGF-beta signaling. Genes 35. Gobe GC, Axelsen RA: Genesis of renal tubular atrophy in
Dev 14:627– 644, 2000 experimental hydronephrosis in the rat. Role of apoptosis. Lab
16. Rennke HG, Anderson S, Brenner BM: The progression of renal Invest 56:273–281, 1987
disease: structural and functional correlations. In: Renal Pathol- 36. Ito H, Kasagi N, Shomori K, Osaki M, Adachi H: Apoptosis in
ogy edited by Tisher CC, Brenner BM, Philadelphia, J.B. Lip- the human allografted kidney. Analysis by terminal deoxynu-
pincott Company, 1994, pp 116 –150 cleotidyl transferase-mediated DUTP-botin nick end labeling.
17. Harris DC: Tubulointerstitial renal disease. Curr Opin Nephrol Transplantation 60:794 –798, 1995
Hypertens 10:303–313, 2001 37. Ortiz A, Ziyadeh FN, Neilson EG: Expression of apoptosis-
18. Bohle A, Wehrmann M, Bogenschutz O, Batz C, Vogl W, regulatory genes in renal proximal tubular epithelial cells ex-
Schmitt H, Muller CA, Muller GA: The long-term prognosis of posed to high ambient glucose and in diabetic kidneys. J Investig
the primary glomerulonephritides. A morphological and clinical Med 45:50 –56, 1997
analysis of 1747 cases. Pathol Res Pract 188:908 –924, 1992 38. Kelly DJ, Cox AJ, Tolcos M, Cooper ME, Wilkinson-Berka JL,
19. Fine LG, Orphanides C, Norman JT: Progressive renal disease: Gilbert RE: Attenuation of tubular apoptosis by blockade of the
the chronic hypoxia hypothesis. Kidney Int Suppl 65:S74 –S78, renin-angiotensin system in diabetic Ren-2 rats. Kidney Int 61:
1998 31–39, 2002
20. Remuzzi G, Ruggenenti P, Benigni A: Understanding the nature 39. Miyajima A, Chen J, Lawrence C, Ledbetter S, Soslow RA, Stern
of renal disease progression. Kidney Int 51:2-15, 1997 J, Jha S, Pigato J, Lemer ML, Poppas DP, Vaughan ED, Felsen
21. Burton C, Harris KP: The role of proteinuria in the progression D: Antibody to transforming growth factor-beta ameliorates tu-
of chronic renal failure. Am J Kidney Dis 27:765–775, 1996 bular apoptosis in unilateral ureteral obstruction. Kidney Int
22. Johnson RJ: Cytokines, growth factors and renal injury: where do 58:2301–2313, 2000
we go now? Kidney Int Suppl 63:S2–S6, 1997 40. Sanderson N, Factor V, Nagy P, Kopp J, Kondaiah P, Wakefield
23. Wu LL, Cox A, Roe CJ, Dziadek M, Cooper ME, Gilbert RE: L, Roberts AB, Sporn MB, Thorgeirsson SS: Hepatic expression
Transforming growth factor beta 1 and renal injury following of mature transforming growth factor beta 1 in transgenic mice
subtotal nephrectomy in the rat: role of the renin-angiotensin results in multiple tissue lesions. Proc Natl Acad Sci U.S.A.
system. Kidney Int 51:1553–1567, 1997 92:2572–2576, 1995
24. Eddy AA: Molecular basis of renal fibrosis. Pediatr Nephrol 41. Kopp JB, Factor VM, Mozes M, Nagy P, Sanderson N, Bottinger
15:290 –301, 2000 EP, Klotman PE, Thorgeirsson SS: Transgenic mice with in-
J Am Soc Nephrol 13: 2600–2610, 2002 TGF-ß Signaling 2609

creased plasma levels of TGF-beta 1 develop progressive renal induce apoptosis of endothelial cells. Cancer Res 57:1735–1742,
disease. Lab Invest 74:991-1003, 1996 1997
42. Strutz F, Okada H, Lo CW, Danoff T, Carone RL, Tomaszewski 59. Choi ME, Ballermann BJ: Inhibition of capillary morphogenesis
JE, Neilson EG: Identification and characterization of a fibro- and associated apoptosis by dominant negative mutant trans-
blast marker: FSP1. J Cell Biol 130:393– 405, 1995 forming growth factor-beta receptors. J Biol Chem 270:21144 –
43. Strutz F, Muller GA, Neilson EG: Transdifferentiation: a new 21150, 1995
angle on renal fibrosis. [Review] [32 refs]. Experimental Ne- 60. Simon M, Grone HJ, Johren O, Kullmer J, Plate KH, Risau W,
phrology 4:267–270, 1996 Fuchs E: Expression of vascular endothelial growth factor and its
44. Ng YY, Huang TP, Yang WC, Chen ZP, Yang AH, Mu W, receptors in human renal ontogenesis and in adult kidney. Am J
Nikolic-Paterson DJ, Atkins RC, Lan HY: Tubular epithelial- Physiol 268:F240 –F250, 1995
myofibroblast transdifferentiation in progressive tubulointersti- 61. Berse B, Brown LF, van de WL, Dvorak HF, Senger DR:
tial fibrosis in 5/6 nephrectomized rats. Kidney Int 54:864 – 876, Vascular permeability factor (vascular endothelial growth factor)
1998 gene is expressed differentially in normal tissues, macrophages,
45. Rastaldi MP, Ferrario F, Giardino L, Dell’Antonio G, Grillo C, and tumors. Mol Biol Cell 3:211–220, 1992
Grillo P, Strutz F, Muller GA, Colasanti G, D’Amico G: Epithe- 62. Miettinen PJ, Ebner R, Lopez AR, Derynck R: TGF-beta induced
lial-mesenchymal transition of tubular epithelial cells in human transdifferentiation of mammary epithelial cells to mesenchymal
renal biopsies. Kidney Int 62:137–146, 2002 cells: involvement of type I receptors. J Cell Biol 127:2021–
46. Piek E, Moustakas A, Kurisaki A, Heldin CH, ten Dijke P: 2036, 1994
TGF-(beta) type I receptor/ALK-5 and Smad proteins mediate 63. Portella G, Cumming SA, Liddell J, Cui W, Ireland H, Akhurst
epithelial to mesenchymal transdifferentiation in NMuMG breast RJ, Balmain A: Transforming growth factor beta is essential for
epithelial cells. J Cell Sci 112 ( Pt 24):4557– 4568, 1999 spindle cell conversion of mouse skin carcinoma in vivo: impli-
47. Bakin AV, Tomlinson AK, Bhowmick NA, Moses HL, Arteaga cations for tumor invasion. Cell Growth Differ 9:393– 404, 1998
CL: Phosphatidylinositol-3 Kinase Function is Required for 64. Strutz F, Muller GA, Neilson EG: Transdifferentiation: a new
TGF␤- mediated Epithelial to Mesenchymal Transition and Cell angle on renal fibrosis. Exp Nephrol 4:267–270, 1996
Migration. J Biol Chem 2000 65. Fan JM, Ng YY, Hill PA, Nikolic-Paterson DJ, Mu W, Atkins
RC, Lan HY: Transforming growth factor-beta regulates tubular
48. Strutz F, Zeisberg M, Ziyadeh FN, Yang CQ, Kalluri R, Muller GA,
epithelial-myofibroblast transdifferentiation in vitro. Kidney Int
Neilson EG: Role of basic fibroblast growth factor-2 in epithelial-
56:1455–1467, 1999
mesenchymal transformation. Kidney Int 61:1714 –1728, 2002
66. Shen X, Li J, Hu PP, Waddell D, Zhang J, Wang XF: The activity
49. Oldfield MD, Bach LA, Forbes JM, Nikolic-Paterson D, McRob-
of guanine exchange factor NET1 is essential for transforming
ert A, Thallas V, Atkins RC, Osicka T, Jerums G, Cooper ME:
growth factor-beta-mediated stress fiber formation. J Biol Chem
Advanced glycation end products cause epithelial-myofibroblast
276:15362–15368, 2001
transdifferentiation via the receptor for advanced glycation end
67. Bhowmick NA, Ghiassi M, Bakin A, Aakre M, Lundquist CA,
products (RAGE). J Clin Invest 108:1853–1863, 2001
Engel ME, Arteaga CL, Moses HL: Transforming Growth Fac-
50. Mundel P, Schwarz K, Reiser J: Podocyte biology: a footstep
tor-beta1 Mediates Epithelial to Mesenchymal Transdifferentia-
further. Adv Nephrol Necker Hosp 31:235–241, 2001
tion through a RhoA-dependent Mechanism. Mol Biol Cell 12:
51. Petermann AT, Hiromura K, Blonski M, Pippin J, Monkawa T,
27–36, 2001
Durvasula R, Couser WG, Shankland SJ: Mechanical stress 68. Comijn J, Berx G, Vermassen P, Verschueren K, van Grunsven
reduces podocyte proliferation in vitro. Kidney Int 61:40 –50, L, Bruyneel E, Mareel M, Huylebroeck D, van Roy F: The
2002 two-handed E box binding zinc finger protein SIP1 downregu-
52. Shankland SJ: Cell-cycle control and renal disease. Kidney Int lates E-cadherin and induces invasion. Mol Cell 7:1267–1278,
52:294 –308, 1997 2001
53. Schiffer M, Bitzer M, Roberts IS, Kopp JB, ten Dijke P, Mundel 69. Barres BA, Hart IK, Coles HS, Burne JF, Voyvodic JT, Rich-
P, Bottinger EP: Apoptosis in podocytes induced by TGF-beta ardson WD, Raff MC: Cell death and control of cell survival in
and Smad7. J Clin Invest 108:807– 816, 2001 the oligodendrocyte lineage. Cell 70:31– 46, 1992
54. Shih NY, Li J, Karpitskii V, Nguyen A, Dustin ML, Kanagawa 70. Coles HS, Burne JF, Raff MC: Large-scale normal cell death in
O, Miner JH, Shaw AS: Congenital nephrotic syndrome in mice the developing rat kidney and its reduction by epidermal growth
lacking CD2-associated protein. Science 286:312–315, 1999 factor. Development 118:777–784, 1993
55. Shimizu A, Kitamura H, Masuda Y, Ishizaki M, Sugisaki Y, 71. Sugiyama H, Kashihara N, Makino H, Yamasaki Y, Ota A:
Yamanaka N: Rare glomerular capillary regeneration and subse- Apoptosis in glomerular sclerosis. Kidney Int 49:103–111, 1996
quent capillary regression with endothelial cell apoptosis in 72. Wagrowska-Danilewicz M, Danilewicz M: A study of apoptosis
progressive glomerulonephritis. Am J Pathol 151:1231–1239, in human glomerulonephritis as determined by in situ non-
1997 radioactive labelling of DNA strand breaks. Acta Histochem
56. Kitamura H, Shimizu A, Masuda Y, Ishizaki M, Sugisaki Y, 99:257–266, 1997
Yamanaka N: Apoptosis in glomerular endothelial cells during 73. Sugiyama H, Kashihara N, Makino H, Yamasaki Y, Ota A:
the development of glomerulosclerosis in the remnant-kidney Apoptosis in glomerular sclerosis. Kidney Int 49:103–111, 1996
model. Exp Nephrol 6:328 –336, 1998 74. Wagrowska-Danilewicz M, Danilewicz M: A study of apoptosis
57. Ohashi R, Kitamura H, Yamanaka N: Peritubular capillary injury in human glomerulonephritis as determined by in situ non-
during the progression of experimental glomerulonephritis in radioactive labelling of DNA strand breaks. Acta Histochem
rats. J Am Soc Nephrol 11:47–56, 2000 99:257–266, 1997
58. Guo N, Krutzsch HC, Inman JK, Roberts DD: Thrombospondin 75. Schuster N, Krieglstein K: Mechanisms of TGF-beta-mediated
1 and type I repeat peptides of thrombospondin 1 specifically apoptosis. Cell Tissue Res 307:1–14, 2002
2610 Journal of the American Society of Nephrology J Am Soc Nephrol 13: 2600–2610, 2002

76. Ten Dijke P, Goumans MJ, Itoh F, Itoh S: Regulation of cell growth factor beta in prostatic carcinoma cells. Curr Biol 10:
proliferation by Smad proteins. J Cell Physiol 191:1–16, 2002 535–538, 2000
77. Yanagisawa K, Osada H, Masuda A, Kondo M, Saito T, Yatabe 89. Lallemand F, Mazars A, Prunier C, Bertrand F, Kornprost M,
Y, Takagi K, Takahashi T, Takahashi T: Induction of apoptosis Gallea S, Roman-Roman S, Cherqui G, Atfi A: Smad7 inhibits
by Smad3 and down-regulation of Smad3 expression in response the survival nuclear factor kappaB and potentiates apoptosis in
to TGF-beta in human normal lung epithelial cells. Oncogene epithelial cells. Oncogene 20:879 – 884, 2001
17:1743–1747, 1998 90. von Gersdorff G, Susztak K, Rezvani F, Bitzer M, Liang D,
78. Jang CW, Chen CH, Chen CC, Chen JY, Su YH, Chen RH: Bottinger EP: Smad3 and Smad4 mediate transcriptional activa-
TGF-beta induces apoptosis through Smad-mediated expression tion of the human Smad7 promoter by transforming growth
of DAP-kinase. Nat Cell Biol 4:51–58, 2002 factor beta. J Biol Chem 275:11320 –11326, 2000
79. Chalaux E, Lopez-Rovira T, Rosa JL, Pons G, Boxer LM, 91. Topper JN, Cai J, Qiu Y, Anderson KR, Xu YY, Deeds JD,
Bartrons R, Ventura F: A zinc-finger transcription factor induced Feeley R, Gimeno CJ, Woolf EA, Tayber O, Mays GG, Sampson
by TGF-beta promotes apoptotic cell death in epithelial Mv1Lu BA, Schoen FJ, Gimbrone MA, Jr., Falb D: Vascular MADs: two
cells. FEBS Lett. 457:478 – 482, 1999 novel MAD-related genes selectively inducible by flow in human
80. Ribeiro A, Bronk SF, Roberts PJ, Urrutia R, Gores GJ: The vascular endothelium. Proc Natl Acad Sci U.S.A 94:9314 –9319,
transforming growth factor beta(1)-inducible transcription factor 1997
TIEG1, mediates apoptosis through oxidative stress. Hepatology 92. Ulloa L, Doody J, Massague J: Inhibition of transforming growth
30:1490 –1497, 1999 factor-beta/SMAD signalling by the interferon-gamma/STAT
81. Ohta S, Yanagihara K, Nagata K: Mechanism of apoptotic cell pathway. Nature 397:710 –713, 1999
death of human gastric carcinoma cells mediated by transforming 93. Bitzer M, von Gersdorff G, Liang D, Dominguez-Rosales A, Beg
growth factor beta. Biochem J 324 ( Pt 3):777–782, 1997 AA, Rojkind M, ttinger EP: A mechanism of suppression of
82. Kim BC, Mamura M, Choi KS, Calabretta B, Kim SJ: Trans- TGF-beta/SMAD signaling by NF-kappaB/RelA. Genes Dev 14:
forming growth factor beta 1 induces apoptosis through cleavage 187–197, 2000
of BAD in a Smad3-dependent mechanism in FaO hepatoma 94. Larisch S, Yi Y, Lotan R, Kerner H, Eimerl S, Tony PW,
cells. Mol Cell Biol 22:1369 –1378, 2002 Gottfried Y, Birkey RS, de Caestecker MP, Danielpour D, Book-
83. Perlman R, Schiemann WP, Brooks MW, Lodish HF, Weinberg Melamed N, Timberg R, Duckett CS, Lechleider RJ, Steller H,
RA: TGF-beta-induced apoptosis is mediated by the adapter Orly J, Kim SJ, Roberts AB: A novel mitochondrial septin-like
protein Daxx that facilitates JNK activation. Nat Cell Biol protein, ARTS, mediates apoptosis dependent on its P-loop mo-
3:708 –714, 2001 tif. Nat Cell Biol 2:915–921, 2000
84. Yamaguchi K, Shirakabe K, Shibuya H, Irie K, Oishi I, Ueno N, 95. Motyl T, Grzelkowska K, Zimowska W, Skierski J, Wareski P,
Taniguchi T, Nishida E, Matsumoto K: Identification of a mem- Ploszaj T, Trzeciak L: Expression of bcl-2 and bax in TGF-beta
ber of the MAPKKK family as a potential mediator of TGF-beta 1-induced apoptosis of L1210 leukemic cells. Eur J Cell Biol
signal transduction. Science 270:2008 –2011, 1995 75:367–374, 1998
85. Shibuya H, Yamaguchi K, Shirakabe K, Tonegawa A, Gotoh Y, 96. Francis JM, Heyworth CM, Spooncer E, Pierce A, Dexter TM,
Ueno N, Irie K, Nishida E, Matsumoto K: TAB1: an activator of Whetton AD: Transforming growth factor-beta 1 induces apo-
the TAK1 MAPKKK in TGF-beta signal transduction. Science ptosis independently of p53 and selectively reduces expression of
272:1179 –1182, 1996 Bcl-2 in multipotent hematopoietic cells. J Biol Chem 275:
86. Wang W, Zhou G, Hu MC, Yao Z, Tan TH: Activation of the 39137–39145, 2000
hematopoietic progenitor kinase-1 (HPK1)-dependent, stress-ac- 97. Chipuk JE, Bhat M, Hsing AY, Ma J, Danielpour D: Bcl-xL
tivated c-Jun N-terminal kinase (JNK) pathway by transforming blocks transforming growth factor-beta 1-induced apoptosis by
growth factor beta (TGF-beta)-activated kinase (TAK1), a kinase inhibiting cytochrome c release and not by directly antagonizing
mediator of TGF beta signal transduction. J Biol Chem 272: Apaf-1-dependent caspase activation in prostate epithelial cells.
22771–22775, 1997 J Biol Chem 276:26614 –26621, 2001
87. Mazars A, Lallemand F, Prunier C, Marais J, Ferrand N, Pessah 98. Schnaper HW, Hayashida T, Poncelet AC: It’s a Smad World:
M, Cherqui G, Atfi A: Evidence for a role of the JNK cascade in Regulation of TGF-beta Signaling in the Kidney. J Am Soc
Smad7-mediated apoptosis. J.Biol.Chem. 276:36797-36803, Nephrol 13:1126 –1128, 2002
2001 99. Verrecchia F, Mauviel A: Control of connective tissue gene
88. Landstrom M, Heldin NE, Bu S, Hermansson A, Itoh S, Dijke P, expression by TGF beta: role of Smad proteins in fibrosis. Curr
Heldin CH: Smad7 mediates apoptosis induced by transforming Rheumatol Rep 4:143–149, 2002

Access to UpToDate on-line is available for additional clinical information


at http://www.jasn.org/

Potrebbero piacerti anche