16
Kidney International, Vol. 59 (2001), pp. 2023–2038 PERSPECTIVES IN BASIC SCIENCE Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases KUNIO MATSUMOTO and TOSHIKAZU NAKAMURA Division of Biochemistry, Biomedical Research Center, Osaka University Graduate School of Medicine, Osaka, Japan Hepatocyte growth factor: Renotropic role and potential thera- unrecognized. Understanding the molecular mechanisms peutics for renal diseases. Hepatocyte growth factor (HGF), a involved in regeneration systems will lead to new clinical ligand for the c-Met receptor tyrosine kinase, has mitogenic, strategies to combat renal disease, as is now the case for motogenic, anti-apoptotic, and morphogenic (for example, in- the immune and neuroendocrine systems. duction of branching tubulogenesis) activities for renal tubular Patterns of tissue regeneration are mainly divided into cells, while it has angiogenic and angioprotective actions for two distinct systems. One is the “stem cell system.” In endothelial cells. Stromal cells such as mesangial cells, endothe- lial cells, and macrophages are sources of renal HGF; thus, tissues, such as nervous and muscle, stem cells usually HGF mediates epithelial–stromal and endothelial–mesangial remain in a quiescent and undifferentiated state (but interactions in the kidney. In response to acute renal injury, committed to differentiate into specific cell types), yet the expression of HGF increases in the injured kidney and in they proliferate to produce daughter cells that will differ- distant intact organs such as the lung and spleen. Locally and entiate into the required tissues, in response to injuries. systemically increased HGF supports renal regeneration, possi- The other is a “simple duplication system.” In parenchy- bly not only by enhancing cell growth but also by promoting morphogenesis of renal tissue. During progression of chronic mal organs such as the kidney and liver, differentiated renal failure/renal fibrosis, the expression of HGF decreases cells proliferate without dedifferentiation to replace ex- in a manner reciprocal to the increase in expression of trans- cised or injured tissues. Thus, to understand mechanisms forming growth factor-b (TGF-b), a key player in tissue fibrosis. involved in renal and hepatic regeneration, the following A decrease in endogenous HGF, as well as increase in TGF-b, issues must be addressed: (1) how differentiated cells augments susceptibility to the onset of chronic renal failure/ recognize injury or partial removal of tissues even though renal fibrosis. On the other hand, supplements of exogenous HGF have preventive and therapeutic effects in cases of acute the remaining cells are apparently intact; (2) which tropic and chronic renal failure/renal fibrosis in laboratory animals. factors are responsible for regeneration; and (3) how HGF prevents epithelial cell death and enhances regeneration differentiated cells proliferate and reorganize tissue-spe- and remodeling of renal tissue with injury or fibrosis. A reno- cific multicellular architectures. tropic system underlies the vital potential of the kidney to Both the kidney and the liver have a vital capacity to regenerate, while an impaired renotropic system may confer regenerate. After unilateral nephrectomy, a compensa- susceptibility to the onset of renal diseases. Thus, HGF supple- mentation may be one therapeutic strategy to treat subjects tory renal enlargement occurs and blood-borne “reno- with renal diseases, as it enhances the intrinsic ability of the tropin” is present that enhances renal regeneration [1, 2]. kidney to regenerate. Although renotropin(s) has yet to be clearly identified, the renotropic system is considered to support the intrin- sic ability of the kidney to regenerate in response to Animals, including mammals, share the intrinsic abil- injury. Studies done in the 1990s have shown that hepato- ity to regenerate specific tissues and organs. This unique cyte growth factor (HGF) qualifies as a candidate for potential to regenerate damaged or lost tissues is a bio- renotropin. An increase and a decrease in expression of logical defense system, and many therapies depend on renotropic HGF are respectively involved in renal regen- the intrinsic regeneration potential (for example, treat- eration and susceptibility to the onset of diseases such ment for bone fracture), although this notion often goes as fibrosis of the kidney. This review focuses on the renotropic functions of HGF for renal regeneration/pro- tection and the potential of HGF to treat humans with Key words: acute renal failure, cell regeneration, chronic renal failure, renal disorders. renal fibrosis, morphogenesis. Received for publication August 31, 2000 BACKGROUND and in revised form November 14, 2000 Accepted for publication November 16, 2000 Hepatocyte growth factor, first identified [3, 4], puri- fied [5–8], and cloned [9, 10] as a potent mitogen for fully 2001 by the International Society of Nephrology 2023

Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Embed Size (px)

Citation preview

Page 1: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Kidney International, Vol. 59 (2001), pp. 2023–2038

PERSPECTIVES IN BASIC SCIENCE

Hepatocyte growth factor: Renotropic role and potentialtherapeutics for renal diseases

KUNIO MATSUMOTO and TOSHIKAZU NAKAMURA

Division of Biochemistry, Biomedical Research Center, Osaka University Graduate School of Medicine, Osaka, Japan

Hepatocyte growth factor: Renotropic role and potential thera- unrecognized. Understanding the molecular mechanismspeutics for renal diseases. Hepatocyte growth factor (HGF), a involved in regeneration systems will lead to new clinicalligand for the c-Met receptor tyrosine kinase, has mitogenic, strategies to combat renal disease, as is now the case formotogenic, anti-apoptotic, and morphogenic (for example, in-

the immune and neuroendocrine systems.duction of branching tubulogenesis) activities for renal tubularPatterns of tissue regeneration are mainly divided intocells, while it has angiogenic and angioprotective actions for

two distinct systems. One is the “stem cell system.” Inendothelial cells. Stromal cells such as mesangial cells, endothe-lial cells, and macrophages are sources of renal HGF; thus, tissues, such as nervous and muscle, stem cells usuallyHGF mediates epithelial–stromal and endothelial–mesangial remain in a quiescent and undifferentiated state (butinteractions in the kidney. In response to acute renal injury, committed to differentiate into specific cell types), yetthe expression of HGF increases in the injured kidney and in

they proliferate to produce daughter cells that will differ-distant intact organs such as the lung and spleen. Locally andentiate into the required tissues, in response to injuries.systemically increased HGF supports renal regeneration, possi-The other is a “simple duplication system.” In parenchy-bly not only by enhancing cell growth but also by promoting

morphogenesis of renal tissue. During progression of chronic mal organs such as the kidney and liver, differentiatedrenal failure/renal fibrosis, the expression of HGF decreases cells proliferate without dedifferentiation to replace ex-in a manner reciprocal to the increase in expression of trans- cised or injured tissues. Thus, to understand mechanismsforming growth factor-b (TGF-b), a key player in tissue fibrosis.

involved in renal and hepatic regeneration, the followingA decrease in endogenous HGF, as well as increase in TGF-b,issues must be addressed: (1) how differentiated cellsaugments susceptibility to the onset of chronic renal failure/recognize injury or partial removal of tissues even thoughrenal fibrosis. On the other hand, supplements of exogenous

HGF have preventive and therapeutic effects in cases of acute the remaining cells are apparently intact; (2) which tropicand chronic renal failure/renal fibrosis in laboratory animals. factors are responsible for regeneration; and (3) howHGF prevents epithelial cell death and enhances regeneration differentiated cells proliferate and reorganize tissue-spe-and remodeling of renal tissue with injury or fibrosis. A reno-

cific multicellular architectures.tropic system underlies the vital potential of the kidney toBoth the kidney and the liver have a vital capacity toregenerate, while an impaired renotropic system may confer

regenerate. After unilateral nephrectomy, a compensa-susceptibility to the onset of renal diseases. Thus, HGF supple-mentation may be one therapeutic strategy to treat subjects tory renal enlargement occurs and blood-borne “reno-with renal diseases, as it enhances the intrinsic ability of the tropin” is present that enhances renal regeneration [1, 2].kidney to regenerate. Although renotropin(s) has yet to be clearly identified,

the renotropic system is considered to support the intrin-sic ability of the kidney to regenerate in response to

Animals, including mammals, share the intrinsic abil- injury. Studies done in the 1990s have shown that hepato-ity to regenerate specific tissues and organs. This unique cyte growth factor (HGF) qualifies as a candidate forpotential to regenerate damaged or lost tissues is a bio- renotropin. An increase and a decrease in expression oflogical defense system, and many therapies depend on renotropic HGF are respectively involved in renal regen-the intrinsic regeneration potential (for example, treat- eration and susceptibility to the onset of diseases suchment for bone fracture), although this notion often goes as fibrosis of the kidney. This review focuses on the

renotropic functions of HGF for renal regeneration/pro-tection and the potential of HGF to treat humans with

Key words: acute renal failure, cell regeneration, chronic renal failure, renal disorders.renal fibrosis, morphogenesis.

Received for publication August 31, 2000 BACKGROUNDand in revised form November 14, 2000Accepted for publication November 16, 2000 Hepatocyte growth factor, first identified [3, 4], puri-

fied [5–8], and cloned [9, 10] as a potent mitogen for fully 2001 by the International Society of Nephrology

2023

Page 2: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems2024

Fig. 1. Schematic structures. (A) Prohepato-cyte growth factor (HGF) and mature HGF.(B) Typical biological activities of HGF medi-ated by c-Met/HGF receptor and intracellularsignal transducers which associate with tyro-sine-phosphorylated c-Met.

differentiated hepatocytes, is a heterodimeric molecule single-chain precursor form, and processing by specificserine proteases into the two-chain form is coupled tocomposed of a 69 kD a-chain and a 34 kD b-chain [6–8].

The a-chain contains the N-terminal hairpin domain and its activation (Fig. 1A). HGF activator, a serine proteaseresponsible for activation of HGF, has structural similar-the subsequent four kringle domains, and the b-chain

contains a serine protease-like domain (Fig. 1A) [9, 10]. ity to blood coagulation factor XII [11]. Involvementof urokinase-type plasminogen activator (uPA) in theThe kringle domain was initially found in serine proteases

involved in blood coagulation or thrombolysis, and HGF activation of pro-HGF was also reported [12].Hepatocyte growth factor has multiple biological activi-has a 38% amino acid sequence homology to plasminogen.

Plasminogen is composed of a five kringle-containing ties on a wide variety of cells, including mitogenic, moto-genic (enhancement of cell movement), morphogenic, andA-chain and B-chain of the serine protease catalytic sub-

unit, whereas HGF has no serine protease activity because anti-apoptotic activities (Fig. 1B) [13–19]. The motogenicaction of HGF was deduced from the unexpected findingamino acids are substituted in the catalytic center. HGF

is biosynthesized and secreted in a biologically inactive that characterization of scatter factor showed it to be

Page 3: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems 2025

identical to HGF [20–22]. Scatter factor was originally creasing cadherin-mediated cell–cell adhesion, respec-tively. In addition to transphosphorylation-dependentidentified and purified as a fibroblast-derived epithelial

cell motility factor [23–25]. HGF affects cell–cell interac- signaling pathways, the induction of gene expression ofproteins involved in cell-associated proteolytic break-tion and cell–extracellular matrix (ECM) interaction and

stimulates or activates proteolytic networks involved in down of ECM components plays an important role inbranching tubulogenesis, including uPA, uPA receptor,the breakdown of ECM proteins [26–29]. Thus, typical

biological activities of HGF are involved in construction, and membrane-type 1 matrix metalloprotease (MT1-MMP) [26–28]. In terms of anti-apoptosis, the activationremodeling, and protection of tissue structures during

development and regeneration. of PI-3 kinase and downstream Akt (protein kinase B)[47] and the induction of Bcl-2/Bcl-xL are likely path-The receptor for HGF was identified in 1991 to be a

c-met protooncogene product [30, 31]. The c-Met receptor ways responsible for protection of cells from apoptosisby HGF [48, 49].is composed of a 50 kD a-chain and a 145 kD b-chain

[32]. The a-chain is exposed extracellularly, while the The essential role of HGF and the c-Met receptor inmammalian development was defined by disruption ofb-chain is a transmembrane subunit containing an intra-

cellular tyrosine kinase domain (Fig. 1B). Binding of HGF or the c-Met gene in mice; these animals died onembryonic days 13 to 15 because of impaired organogene-HGF to the Met receptor induces activation of tyrosine

kinase, which results in the subsequent phosphorylation sis of the placenta and liver [50–52]. HGF is also involvedin the development of epithelial tissues, including theof C-terminally clustered tyrosine residues [33–36]. Phos-

phorylation of these tyrosine residues recruits intracellu- kidney, lung, mammary gland, and teeth, as a mediator inepithelial–mesenchymal interactions for organogenesislar signaling molecules containing the src homology (SH)

domain, including Gab-1, phospholipase C-g (PLC-g), (discussed later in this article) [18, 53]. Concerning neu-roneal development, HGF supports the projection ofRas-GTPase activating protein (Ras-GAP), phosphati-

dylinositol 4,5-bisphosphate 3-kinase (PI-3 kinase), c-Src, motor neurons [54].Shp-2, Crk-2, and Grb-2 (Fig. 1B). Physiologically, HGF has an organotrophic role in the

Although the intracellular signaling pathways leading regeneration and protection of various organs, includingto specific or preferential activation of each biological the liver, lung, stomach, pancreas, heart, neurons, andresponse (that is, mitogenic, motogenic, morphogenic, kidney [16, 55]. HGF levels increase in tissues and bloodor anti-apoptotic) driven by HGF-Met receptor coupling in response to various acute injuries and diseases. Thehave yet to be fully defined, preferential activation of administration of HGF into laboratory animals has po-signaling molecules and induction of gene expression tent therapeutic effects on various models of acute andresponsible for activation of specific cellular responses chronic organ diseases [part of related publications, ex-are evident. Phosphotyrosine-dependent recruitment of cept for renal diseases; 48, 49, 56–76]. Likewise, HGFthe Grb-2/SOS complex activates Ras and subsequent has angiogenic activity for vascular endothelial cells andphosphorylation events, including extracellular signal- induces extensive blood vessels when administered intoregulated kinase (ERK). Activation of the Ras-ERK ischemic tissue [65, 70, 76, 77]. On the other hand, HGFpathway is required for cellular proliferation [33]. On regulates malignant behavior in various types of tumors,the other hand, the association and tyrosine phosphory- including invasion, metastasis, and tumor angiogenesislation of Gab-1, a docking protein that couples the Met [27, 29, 67]. Likewise, aberrant activation of the c-Metreceptor with multiple signaling proteins such as PI-3 receptor is associated with tumorigenesis and malignantkinase, PLC-g, Shp-2, and Crk-2, plays a definite role behavior of cancers [27, 29, 78]. The gain-of-functionin HGF-induced morphogenesis and cellular movement mutation in the c-met gene plays a determinant role in[36, 37]. The expression of Gab-1 in Madin-Darby canine hereditary and some sporadic papillary renal carcinomaskidney (MDCK) renal epithelial cells induces cell move- [79–81].ment and branching tubulogenesis [37]. The associationof Gab-1 with Shp-2 protein tyrosine phosphatase, but

ACTIONS OF HGF IN RENALnot PI-3 kinase and Crk-2, is essential for inducing epi-TISSUE ORGANIZATIONthelial branching tubulogenesis [36, 38]. Instead, activa-

Hepatocyte growth factor exerts mitogenic responsestion of PI-3 kinase and the Rho family GTPases (Rho,in renal epithelial cells derived from distinct regions andRac, Cdc42) plays a critical role in HGF-induced cellspecies, including rabbit and rat proximal tubular cellsmovement [39–42]. Rho GTPases regulate actin-related[82–84] and rat glomerular epithelial cells [84]. Figurecytoskeletal organization and contractive force for cell2A shows the effects of HGF and epidermal growthmovement and concomitant cell shape rearrangement.factor (EGF) on DNA synthesis of cultured rabbit proxi-Likewise, HGF induces tyrosine phosphorylation of fo-mal tubular cells [82]. HGF stimulates the proliferationcal adhesion kinase (FAK) [43, 44] and b-catenin [45, 46],

thereby enhancing the cell–ECM interaction and de- of renal epithelial cell lines, including a rat visceral glo-

Page 4: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems2026

merular cell line [85], proximal tubular cell lines [86],and a murine medullary collecting duct epithelial cell line[87]. Likewise, HGF exhibits mitogenic action on renalendothelial cells [88]. HGF has no apparent effect on ratand human mesangial cell proliferation [84, 88, 89] andis weakly mitogenic [89, 90], although mesangial cells doexpress the c-Met receptor [88–90]. HGF diminished ordecreased stress fibers, focal contacts, and extracellularfibronectin deposition in a murine mesangial cell culture,thereby suggesting a reduced cell–substratum interactionand enhanced cellular motility and remodeling in themesangial cell-related extracellular scaffold [89]. Like-wise, enhancement of cell motility in renal epithelial cellsclearly has been evidenced in MDCK renal epithelialcells [24, 25], and HGF stimulated Na,K-ATPase in arenal tubular cell line [86].

The indispensable role of tubular cells for renal func-tions depends on their multicellular architecture, that is,branching tubules, and formation of branching tubulo-genesis is involved in both renal regeneration and devel-opment. Montesano, Schaller, and Orci, using a model ofepithelial–mesenchymal interaction, first demonstratedthat MDCK renal epithelial cells undergo branching tu-bulogenesis in collagen gels in the presence of fibroblast-derived conditioned medium [91], and the fibroblast-derived epithelial morphogen proved to be HGF [92].Figure 2B shows the induction of branching tubulogen-esis by HGF in MDCK cells grown in a collagen gel.Other growth factors such as EGF, fibroblast growthfactor-1 (FGF-1), FGF-2, FGF-7, insulin-like growth fac-tor-I (IGF-I), IGF-II, and platelet-derived growth factor(PDGF) do not induce branching tubulogenesis, whereastransforming growth factor-b1 (TGF-b1) inhibits tubulo-genesis in MDCK cells [92, 93]. Further insight into theprocess of branching tubulogenesis reveals that branch-ing tubules formed by HGF in MDCK cells retain a well-defined apical-basolateral polarity [94] and that mem-brane type-matrix metalloproteinase plays an essentialrole in HGF-induced branching tubulogenesis [28]. Boweset al reported that HGF induces branching tubulogenesisof renal proximal tubular cells in primary culture [95].

Fig. 2. (A) Mitogenic action of HGF (d) and epidermal growth factorOther growth factors, including FGF-1, FGF-7, EGF, or (EGF; s) on cultured rabbit renal tubular cells. DNA synthesis of renal

tubular cells cultured in the absence or presence of HGF or EGF wasIGF-I, failed to induce this morphogenesis, however,measured. (B) Induction of branching tubulogenesis by HGF in MDCKcombinations of these growth factors induced branchingrenal epithelial cells grown in a collagen gel. Cells were grown in the

tubulogenesis [95]. These results indicate that morpho- absence or presence of 10 ng/mL HGF. (C) Potential roles of HGF inepithelial–stromal and endothelial–mesangial interactions in the kidney.genic signal transduction through HGF-Met is redun-

dantly achieved by a combination of multiple signals, co-operatively activated by sets of growth factors and theirreceptors. While HGF is not the sole molecule capable of

induces the ureteric bud to bud and branch, and theinducing branching tubulogenesis in renal tubular cells,ureteric bud, in turn, induces metanephric condensationit is definitely important for reconstructing the tubularand the mesenchymal to epithelial cell conversion [96, 97].architecture during renal development and regeneration.In the developing kidney, HGF is expressed in the meta-During renal development, a signal exchange betweennephric mesenchyme, and the Met receptor is expressedthe metanephric mesenchyme and the epithelial ureteric

bud plays a crucial role. The metanephric mesenchyme in the ureteric buds and in the epithelial cells of the

Page 5: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems 2027

Fig. 2. (Continued)

nephron [98–100]. Neutralizing antibody to HGF inhib- apoptotic action of Bcl-2/Bcl-xL, and Bag-1 expressionited metanephric growth and morphogenesis in an organ in cells promotes the cell survival action of HGF [106].culture of embryonic kidneys [99, 100]. In addition, HGF In tubular regions of the adult kidney, HGF is ex-enhances differentiation of metanephric mesenchymal pressed in interstitial cells, presumably endothelial cellscells into epithelial cells, whereas anti-HGF antibody and macrophages [107–109], while in the glomerulus, mes-inhibits it, thereby indicating that HGF mediates the angial cells and endothelial cells express HGF [84, 86, 89,mesenchymal to epithelial conversion of the metanephric 102]. Collectively, in the kidney, HGF seems to act onmesenchyme [100, 101]. renal epithelial, endothelial, and mesangial cells through

Hepatocyte growth factor protects renal epithelial cells autocrine- or paracrine-related pathways (Fig. 2C).from apoptotic cell death [102–105], and the anti-apoptoticeffect of HGF in renal epithelial cells is mediated by the

HGF IN RENOTROPIC SYSTEMSinduction of Bcl-xL expression and the phosphorylationIn addition to biological activities of HGF on renalof Bad, thereby inactivating this pro-apoptotic protein.

cells, involvement of HGF in renal regeneration has beenIt is noteworthy that Bag-1, a partner of Bcl-2/Bcl-xL, isproposed based on findings that renal and plasma HGFspecifically associated with the c-Met receptor [106]. The

association of Bag-1 with Bcl-2/Bcl-xL enhances the anti- levels and c-Met receptor expression are regulated in

Page 6: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems2028

Fig. 3. Possible pathways mediated by HGF for renal regeneration in response to acute renal injury. (A) Following renal injuries, HGF expressionis up-regulated in stromal cells in the kidney and in distant intact organs such as the lung, liver, and spleen. The right upper panel shows inductionof HGF mRNA in the lung following unilateral nephrectomy (UN) in rats. (B) Regulation of c-Met receptor activation in noninjured and injuredtissues. Change in cell–cell contact or junctional communication due to tissue injury may possibly regulate c-Met receptor function, such that HGFexerts its biological activities in the injured tissue-specific manner.

response to renal injuries. A unilateral nephrectomy the kidney acting through paracrine and an autocrinemechanisms and HGF produced in distant organs actingmodel has been used to study renotropic systems in com-

pensatory renal regeneration. HGF mRNA and protein through an endocrine mechanism (Fig. 3A).The induction of HGF expression in distant intactlevels increase in the remaining kidney after unilateral

nephrectomy [110, 111]. Rapid increases in HGF mRNA organs suggests that a circulating factor(s) responsiblefor induction of HGF may exist in the blood of animalsand/or protein levels in the kidney and plasma were seen

in various types of acute renal injuries induced by the with organ injury. Two studies demonstrated the pres-ence of HGF-inducing factor in plasma or serum follow-administration of nephrotoxins such as HgCl2 and folic

acid, glycerol administration, renal ischemia, vitamin E ing the onset of acute organ injury. Plasma obtainedfrom laboratory animals with hepatic or renal injurydeficiency, and ureteral obstruction [107, 112–115]. Like-

wise, increases in blood HGF levels were noted in patients stimulated HGF expression in vivo and in vitro, and theHGF inducer was shown to be proteinous factor [123].with acute renal failure [116, 117] and with acute renal

rejection after renal transplantation [118]. Similarly, HGF levels in sera were remarkably increasedin patients with acute renal failure, and sera from patientsAn induction of HGF mRNA in the kidney following

renal injuries indicates that the kidney is one source of with acute renal failure—but not sera from healthy vol-unteers—strongly stimulated HGF production in mesan-HGF. In the kidney, stromal cells such as macrophages

and endothelial and mesangial cells express HGF [107, gial cells in culture [95].Although the identification of HGF inducers in the109]; thus, HGF seems to act through paracrine- and

autocrine-related pathways. On the other hand, HGF plasma or serum remains to be addressed, several knownmolecules are involved in regulating HGF expression.mRNA expression is up-regulated in distant intact or-

gans such as the lung, liver, and spleen, as well as the Among inflammatory mediators, interleukin-1 (IL-1),interferon-g, tumor necrosis factor-a, and prostaglandininjured kidney following acute renal injury, including

unilateral nephrectomy (Fig. 3A), HgCl2 administration, E1 (PGE1) and PGE2 strongly activate gene expressionof HGF [124–126]. Likewise, FGF-1, FGF-2, PDGF, hep-and folic acid administration [114, 119–121]. It is note-

worthy that the induction of HGF mRNA expression in arin-binding EGF-like growth factor (HB-EGF), EGF,and TGF-a induce HGF expression [127–130], whereasdistant noninjured organs was seen in cases of hepatic

and heart injuries [119–122]. These observations suggest TGF-b1 and angiotensin II strongly suppress the geneexpression of HGF [131–133]. Norepinephrine up-regu-that HGF increases in the blood circulation may be de-

rived from noninjured organs, as well as injured organs. lates HGF expression [134], whereas glucocorticoid neg-atively regulates HGF expression [131]. The induction ofTherefore, renotropic systems supported by HGF may

involve two distinct pathways: HGF locally produced in HGF by inflammatory mediators implicates its coopera-

Page 7: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems 2029

tion with immune and regeneration/repair systems, while to two-chain HGF in injured organs. Since neutralizingantibody to HGF-activator inhibited processing to theregulation of HGF by adrenal hormones implicates its

involvement in neuroendocrine and regeneration systems. active HGF, this enzyme is likely to be involved in theinjured tissue-specific activation of HGF [139]. Takentogether, these biological actions of HGF seem to be

INJURED TISSUE-SPECIFIC ACTIVATION OFtightly regulated by distinct mechanisms involved in the

THE HGF–c-MET SYSTEMmodulation of receptor and ligand functions, such that

Given that HGF increases in the circulating blood, HGF exhibits biological actions in an injured tissue-spe-could it nonselectively target both injured and nonin- cific manner.jured tissues and cells? This may be an undesirable out-come, as there would be an overgrowth of noninjured

HGF IN CHRONIC RENAL FAILURE/tissues and cells. However, this is not the case. BindingRENAL FIBROSISof growth factors to their receptors induces down-regula-

tion of receptors because of endocytosis of the ligand- Progressive fibrosis in the kidney, liver, lung, heart,muscle, bone marrow, and skin is both a major cause ofreceptor complex. Thus, analysis of down-regulation

allows one to track ligand-dependent activation of growth suffering and death and an important contributor to thecost of health care, including hemodialysis. Dialysis mar-factor receptors. Analysis of the c-Met receptor in plasma

membranes of injured and noninjured tissues indicates ket estimates indicate that more than 400,000 patientsworldwide have undergone maintenance dialysis, andthat down-regulation of the c-Met receptor occurs in

injured but not in noninjured organs [135, 136]. Down- the number of patients with chronic renal disease is in-creasing.regulation of the c-Met receptor occurs in the kidney

(but not in other organs) after unilateral nephrectomy, Chronic renal disease, characterized by a progressiveloss of parenchyma cells and renal fibrosis, representswhile it occurs in only the liver after hepatic injury. While

the mechanisms by which HGF selectively activates the the morphological equivalent of end-stage chronic renaldisease and occurs independently of the primary under-c-Met receptor in injured tissue are unknown, we can

speculate about the possible mechanisms. In mature he- lying disorder. The intricate architecture and filtratingfunction of the kidney make it particularly vulnerablepatocytes in primary culture, HGF exerts mitogenic ac-

tion in a cell density-dependent manner. HGF potently to the consequences of fibrosis. Distinct lines of evidencepoint to a causal relation between overexpression ofenhances DNA synthesis of hepatocytes cultured at a

low cell density but does not do so in the case of a high TGF-b and the onset of glomerular and tubulointerstitialfibrosis [140–142]. Transgenic expression of TGF-b1 incell density [137]. Moreover, hepatocytes cultured at a

low cell density express a greater number of c-Met recep- mice leads to glomerulosclerosis and tubulointerstitialfibrosis [143–146]. Overexpression of TGF-b1 or TGF-b2tors than when cultured at a high cell density. The rate

of internalization and recycling of the c-Met receptors by intravenous injection or in vivo gene transfectionshowed that the kidney is highly susceptible to rapidon hepatocytes cultured at a low cell density was much

faster than that seen in the case of a high cell density fibrosis [147, 148]. Elevated expression of TGF-b wasnoted in animal models of renal fibrosis [108, 109, 149–[138]. These results suggest that cell–cell adhesion and

communication may regulate the function and expres- 152] and in patients with glomerulonephritis, diabeticnephropathy, and chronic allograft rejection [153–155].sion of the c-Met receptor, such that HGF exerts biologi-

cal activities in an injured tissue-specific manner (Fig. Biological neutralization of TGF-b1 with a proteoglycan(decorin) prevented the increased production of ECM3B). On the other hand, Liu et al found that c-Met

mRNA expression and c-Met protein were markedly in glomeruli [156].Biological actions of TGF-b are in accord with theincreased following injury to the kidney, where active

renal tissue repair/regeneration occurred, thereby sug- notion that overproduction of TGF-b is associated withthe onset of fibrotic disorders. TGF-b causes depositiongesting that local up-regulation of the c-Met receptor

may confer a specificity/susceptibility of HGF to renal of ECM proteins by simultaneously stimulating synthesisof most ECM proteins and the production of inhibitorstissue [114].

The other potential mechanism for injured tissue-spe- of matrix-degrading proteases [140]. For epithelial andendothelial cells, TGF-b induces growth arrest and in-cific activation of c-Met receptor may be associated with

proteolytic activation of single-chain pro-HGF to mature creases apoptotic cell death. TGF-b induces transdiffer-entiation of mesangial cells and renal tubular epithelialHGF, which occurs in an injured tissue-specific manner

[121]. Although HGF increases in the liver, kidney, lung, cells into a-smooth muscle actin (a-SMA)-positive myo-fibroblasts [157, 158]. Myofibroblasts play a definite roleand spleen in response to hepatic and renal injuries,

HGF remains as an inactive single chain in noninjured in the progression of renal fibrosis [159–161].Until recently, it remained unclear whether HGF isorgans, while a significant portion of HGF is converted

Page 8: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems2030

Fig. 4. Possible involvement of counterbalance between HGF and transforming growth factor-b (TGF-b) in the pathogenesis of chronic renalfailure/renal fibrosis. In the beginning of chronic injury, HGF expression is up-regulated, and the compensatory regenerative responses occur.Repetitive injury results in overexpression TGF-b, a potent fibrogenic cytokine that induces extracellular matrix (ECM) accumulation and epithelialand endothelial apoptosis. Since TGF-b suppresses HGF expression, HGF decreases in a reciprocal manner to the increase in TGF-b level duringthe progression of chronic renal failure.

associated with accelerating or preventing the progres- fibrosis and dysfunction. TGF-b expression and ECMaccumulation were increased concomitantly with in-sion of renal fibrosis. However, recent studies clearly

showed that HGF has a role in protecting the nephrotic creases in tubular apoptosis and with decreases in tubularproliferation. In contrast, the administration of HGFkidney from fibrosis [108, 109, 152, 162]. In the ICR

strain-derived glomerulonephritis (ICGN) mice, glomer- strongly suppressed TGF-b expression and preventedthe onset of renal fibrosis and chronic renal failure. Theseular injury is detectable from 3 weeks postnatally, while

renal dysfunction occurs from 12 to 14 weeks, a dysfunc- findings mean that the reciprocal balance between TGF-band HGF is closely associated with the pathogenesis andtion that coincides with tubular destruction and tubulo-

interstitial fibrosis. Because renal dysfunction in patients progression of renal fibrosis and that HGF plays a rolein protecting the kidney from fibrotic changes. In addi-with glomerular disease shows a better correlation with

tubulointerstitial fibrosis than with glomerulosclerosis, tion to the overexpression of TGF-b, a decrease in renalHGF expression may be responsible for the occurrencethe ICGN mouse offers an appropriate model for patho-

physiological studies on human chronic renal disease. of chronic renal disease (Fig. 4).It is noteworthy that a similar reciprocal change in theIn ICGN mice, the expression and the renal level of

TGF-b increase during progression of glomerular and expression of TGF-b and HGF was also noted during theonset of tubulointerstitial fibrosis caused by unilateraltubulointerstitial fibrosis. Renal TGF-b levels correlate

fairly well with accumulation of type I collagen and fi- ureter-ligated obstruction in mice [109]. Likewise, sup-pressive effects of endogenous HGF on chronic renalbronectin, and tubular apoptosis, whereas there is an in-

verse correlation with proliferation of tubular cells [108]. injury and renal fibrosis were demonstrated following5/6 nephrectomy in rats [162]. The administration ofOn the other hand, HGF levels in the kidney are initially

higher than normal at the early stage of glomerulone- neutralizing anti-HGF antibody decreased the glomeru-lar filtration rate and increased renal fibrosis, indicatingphritis, whereas there is a gradual decrease during pro-

gression of renal fibrosis, and the renal HGF level is that endogeneous HGF plays a role in protecting thekidney from chronic renal injury and renal fibrosis. Al-lower than that in normal kidney at advanced stages of

the disease (Fig. 4). Thus, the renal HGF level correlates though involvement of the TGF-b–versus–HGF counter-balance in the onset of chronic renal failure has yet towith a decrease in tubular proliferation and inversely

correlates with increase in the renal TGF-b level, ECM be noted in patients, this reciprocal expression patternwas noted in the livers of patients with a chronic hepaticaccumulation, and tubular apoptosis. To address the

involvement of HGF in progression of renal fibrosis, disease [163, 164]. The increase in blood HGF levels inpatients with “chronic” renal failure is much less thanneutralizing anti-HGF antibody or recombinant HGF

was administered to ICGN mice. Neutralization of en- that in patients with “acute” renal failure [116, 165].Given that a reciprocal balance between TGF-b anddogenous HGF strongly accelerated progression of renal

Page 9: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems 2031

Fig. 5. Preventive effect of HGF on acuterenal failure induced by administration of cis-platin (A) or HgCl2 (B). Acute renal failurewas induced by cisplatin (15 mg/kg bodyweight; A) or HgCl2 (7 mg/kg body weight;B). Mice were injected with HGF (250 mg/kgbody weight) at 0.5 hours prior to administra-tion of cisplatin or HgCl2 and 6, 12, 24, 36, 48and 72 hours or 6, 12, 24, 36, and 48 hoursafter the administration of cisplatin or HgCl2,respectively. BUN (j) and serum creatinine( ) at 96 (cisplatin) or 72 hours (HgCl2) weremeasured. Each value represents the mean 6SD [174].

HGF regulates the pathology of chronic renal disease, mice. In transgenic mice expressing HGF under controlhow this reciprocal balance is achieved and how supple- of the metallothionein promotor (MT-HGF transgenicmentation of exogenous HGF protects the kidney from mice), the mice developed tubular hyperplasia, glomeru-chronic renal fibrosis and dysfunction deserve attention. losclerosis, and polycystic disease [169]. The same miceFirst, TGF-b1 strongly suppresses expression of HGF also developed tumors of diverse origin, including themRNA [131–133], and this TGF-b1–induced suppression mammary gland, skin, and liver [170]. In contrast, inof HGF expression explains why the overexpression of transgenic mice in which the HGF gene is expressedTGF-b is associated with a decreased renal HGF expres- under control of a tissue-specific promoter, tumors didsion. In the promoter region of the HGF gene, there is not develop. Transgenic mice expressing HGF under ana TGF-b inhibitory element involved in TGF-b–induced albumin promoter (Alb-HGF transgenic mice) had atranscriptional inhibition [166, 167], and TGF-b inhibits greatly increased potential for liver regeneration aftertranscription of the HGF gene [168]. On the other hand, partial hepatectomy, but hepatocellular carcinoma didhow exogenous HGF suppresses renal TGF-b levels has not occur [171]. Keratinocyte-specific transgenic expres-yet to be determined. HGF might possibly inhibit tran- sion of HGF affects melanocyte development, leadingscription of the TGF-b gene directly through the c-Met to melanocytosis, but tumors did not occur, even fromreceptor, or the number of TGF-b–positive cells in the the skin [172]. Likewise, transgenic mice expressing HGFkidney may be decreased. TGF-b and HGF are counter- in pancreatic b cells did not develop tumors, and theacting in many multipotent biological actions. TGF-b mice were resistant to the diabetogenic effects of strepto-induces growth arrest and/or apoptosis in renal tubular zotocin [173]. Abundant expression of HGF in variouscells and endothelial cells, but HGF exhibits mitogenic tissues and greatly increased systemic HGF levels fromand anti-apoptotic activities in these cell types. HGF

early developmental stages in MT-HGF transgenic miceinduces branching tubulogenesis in renal epithelial cells,might affect differentiation, maturation, or epithelial re-while inhibition is seen with TGF-b. HGF stimulates ornewal, which results in renal pathology and tumor devel-induces proteases involved in the breakdown of ECMopment in various tissues. In contrast, in vivo studiesproteins in several types of cells, including MT1-MMP,analyzing the therapeutic potential of HGF in relevantuPA, and some matrix metalloproteases (MMPs) [26–29].models for various diseases found no side effects thatIn contrast, TGF-b strongly stimulates the synthesis ofwould limit the therapeutic usage of HGF or the HGFECM proteins and the production of inhibitors of prote-gene [48, 49, 56–76, 108, 109, 152, 174–177]. Therefore,ases involved in ECM breakdown [140]. Consistently,abundant expression of the HGF gene or abundant ad-HGF increased the expression of MMP-9, whereas HGFministration of HGF in which HGF levels reach muchdecreased the expression of tissue inhibitors of matrixhigher levels than are physiologically or pharmacologi-metalloptoteinase-1 (TIMP-1) and TIMP-2 in humancally effective for a long period may potentially cause aproximal renal tubular cells in culture [162].pathology, including renal pathology.

INVOLVEMENT OF ABERRANT HGFPOTENTIAL THERAPEUTIC APPLICATIONSEXPRESSION IN PATHOLOGY

Based on the renotropic and antifibrogenic actions ofInvolvement of an abundant expression of HGF inpathology has been implicated from studies on transgenic HGF, the potential application of HGF for the treatment

Page 10: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems2032

of renal diseases has been tested in various experimentalmodels. In a clinical setting, acute renal failure is oftencaused by nephrotoxic drug administration (for example,cisplatin, cyclosporine A, tacrolimus, and antibiotics)and renal ischemia. Administration of cisplatin causedacute renal failure in mice, whereas prophylactic admin-istration prevented histologic destruction of renal tu-bules and strongly suppressed the onset of acute renaldysfunction (Fig. 5) [174]. Similarly, HGF prevented thehistologic destruction of renal tissue and the onset ofacute renal dysfunction caused by HgCl2 administration,while it enhanced regeneration of tubular epithelial cells[174]. Preventive effects of HGF in cases of acute tubularinjury and renal dysfunction were also seen in case ofcyclosporine A and tacrolimus (FK506) [176, 177]. Like-wise, compared with control rats, rats administered HGFpostischemia had better renal function, reduced mortal-ity, and a lesser degree of histologic injury when renalischemia was induced in rats by bilateral artery occlusion[175]. All of these results suggest that HGF can be testedfor treatment of patients with acute renal failure or forprophylaxis of acute renal failure potentially caused byadministration of nephrotoxic drugs.

Preventive actions of HGF on acute renal injury couldbe potentially extended to chronic allograft nephropathyafter renal transplantation [178, 179]. Chronic allograftnephropathy, characterized by renal fibrosis and dys-function, often develops years after renal transplantationand is a major cause of graft loss. In an experimentalmodel of chronic allograft nephropathy in rats, HGF wasadministered daily for four weeks following engraftment.In control rats without HGF-treatment, allografts showedsevere structural damage within one week. After a periodof quiescence, macrophage infiltration, elevated TGF-b1expression, and myofibroblasts were seen at 16 weeks.

Fig. 6. Therapeutic effect of HGF on chronic renal failure/renal fibrosisAt 32 weeks, renal fibrosis and dysfunction occurred, and in ICGN mice. (A) Changes in TGF-b, collagen accumulation, tubularthe mortality rate was 50%. In contrast, there was little apoptosis, and tubular proliferation by HGF administration in kidneys

of ICGN mice, as determined by enzyme immunoassay (TGF-b) andearly injury and no later renal fibrosis or dysfunction, andimmunohistochemical analysis (collagen type I, apoptosis, and BrdU-all HGF-treated rats survived. Consistently, macrophage uptake). (B) Changes in BUN and serum creatinine (SCr) levels, without

infiltration and overexpression of TGF-b1 in the kidney (s) or with (d) HGF-treatment. When BUN levels reached 40 mg/dL,HGF (500 mg/kg) or saline alone was given daily for 28 days [108, 152].were suppressed in HGF-treated animals. Since acute

ischemic injury during renal transplantation is associatedwith the onset of chronic allograft nephropathy [180,181], the effects of HGF on ischemic injury might prevent therapeutic efficacy of HGF in murine models of chronicthe onset of chronic allograft nephropathy. renal disease and tubulointerstitial fibrosis was remark-

Understanding the molecular and cellular mechanisms able [108, 109, 152]. In ICGN mice, recombinant HGFleading to chronic renal disease has progressed; however, was administered for four weeks during middle- to end-many chronic renal diseases remain incurable. As de- stage chronic renal disease.scribed previously in this article, endogenous renal HGF In control mice not given HGF, molecular and cellularlevels decrease along with progression of renal fibrosis, events leading to end-stage chronic renal disease pro-and neutralization of endogenous HGF accelerates the gressed during this period. Renal TGF-b levels, the num-progression of renal fibrosis toward end-stage pathology. ber of a-SMA-positive myofibroblasts, type I collagenTherefore, supplemental administration of HGF to com- and fibronectin accumulation and the number of tubularpensate for decreased HGF levels may be therapeutic apoptosis increased, whereas the number of proliferating

tubular cells decreased. In contrast, in mice treated within cases of renal fibrosis and chronic renal failure. The

Page 11: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems 2033

Fig. 7. Putative mechanism for therapeutic effects of HGF on chronic renal failure/renal fibrosis. The supplement of HGF suppresses TGF-bexpression and enhances remodeling of renal tissues, including ECM degradation and epithelial and endothelial cell proliferation in fibrotic kidneys.HGF may prevent cell death in epithelial and endothelial cells.

HGF, expression of TGF-b and PDGF, ECM accumula- remarkably increased in the cirrhotic liver, whereas thetion, and the number of myofibroblasts and tubular apo- administration of HGF or the HGF gene induced theptosis decreased, whereas the number of regenerating remodeling of fibrotic/cirrhotic tissue toward a normaltubular cells increased (Fig. 6A). Consistent with these hepatic structure with an accompanying marked de-changes, the beneficial effects of HGF on the clinical crease in hepatic TGF-b expression. Although an associ-outcome were apparent as decreases in levels of serum ation of TGF-b–versus–HGF counterbalance in patho-creatinine, blood urea nitrogen (BUN), and urine albu- genesis and therapeutics of fibrotic disorders was firstmin, and as a diminution in histologic renal injury (Fig. deduced from a murine model of chronic renal disease/6B) [152]. It should be emphasized that serum creatinine, renal sclerosis, it is tempting to hypothesize that theBUN and urine albumin levels, and histologic renal in- counterbalance between TGF-b and HGF has a determi-jury in mice treated with HGF were less than such events nant role in the pathogenesis and therapeutics of a vari-seen at the start of HGF administration. These results ety of fibrosis-related diseases.indicate that treatment with HGF has therapeutic effectsin cases of chronic renal disease, rather than the HGF

CONCLUSION AND PERSPECTIVESsupplements merely retarding or inhibiting the progres-The renotropic system underlies the vital ability of thesion of chronic renal disease (Fig. 7).

kidney to regenerate in response to injury or partialIn addition to renal fibrosis in ICGN mice, the adminis-resection of the kidney. Several growth factors play rolestration of HGF attenuated tubulointerstitial fibrosis, de-in renotropic systems [104, 183]; however, the specificcreased TGF-b expression and tubular apoptosis, androle of each growth factor in renotropic systems has yetincreased tubular proliferation/regeneration followingto be fully clarified. HGF, a renotropic growth factor,unilateral ureter-ligated obstruction in mice [109]. Uni-mediates epithelial–stromal and endothelial–mesangiallateral ureter-ligated obstruction has been widely used asinteractions for regeneration and maintenance of thea model to study the pathogenesis of tubulointerstitial fi-kidney. In response to acute renal injury, the expressionbrosis. In addition, HGF inhibited the cyclosporine A- orof HGF increases and HGF exerts renotropic actions.tacrolimus-induced endothelin-1 release from culturedDuring progression of chronic renal failure/fibrosis, HGFrabbit proximal tubular cells [182]. Endothelin, which isexpression in the kidney decreases to a level much lowerinvolved in the pathogenesis of acute and chronic renalthan normal. The dominant expression of TGF-b overfailure, mediates the nephrotoxic side effects of cyclo-HGF is associated with the progression of chronic renalsporine A and tacrolimus. Moreover, it is noteworthyfailure. HGF has preventive and therapeutic effects inthat HGF plays a role in protecting the liver and lungscases of acute and chronic renal failure by enhancingfrom liver cirrhosis and lung fibrosis in laboratory ani-

mals [57, 59, 61, 63, 71, 74]. The expression of TGF-b1 the reorganization of renal tissue following injuries and

Page 12: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems2034

2. Austin H III, Goldin H, Preuss HG: Humoral regulation offibrosis. HGF may participate in remodeling and regen-renal growth. Nephron 27:163–170, 1981

erative processes of renal tissue not only by enhancing 3. Nakamura T, Nawa K, Ichihara A: Partial purification and char-acterization of hepatocyte growth factor from serum of hepatec-cell growth, but also by promoting the spatial arrange-tomized rats. Biochem Biophys Res Commun 122:1450–1459, 1984ment of parenchymal cells into organized tissue structures.

4. Russell WE, McGowan JA, Bucher NLR: Partial characteriza-To understand renotropic systems better, the ques- tion of hepatocyte growth factor from rat platelets. J Cell Physiol

119:183–192, 1984tions of how renal injury is recognized by renal cells and5. Nakamura T, Teramoto H, Ichihara A: Purification and charac-how the occurrence of injury leads to specific induction

terization of a growth factor from rat platelets for mature paren-of renotropic growth factors remain to be addressed. chymal hepatocytes in culture. Proc Natl Acad Sci USA 83:6489–

6493, 1986HGF increases in the blood circulation and in distant6. Nakamura T, Nawa K, Ichihara A, et al: Purification and subunitintact organs, as well as in the kidney, in response to

structure of hepatocyte growth factor from rat platelets. FEBSrenal injury. Molecules responsible for the induction of Lett 224:311–318, 1987

7. Gohda E, Tsubouchi H, Nakayama H, et al: Purification andHGF and how HGF is involved in regulation of otherpartial characterization of hepatocyte growth factor from plasmagrowth factors will also need further attention. Likewise,of a patient with fulminant hepatic failure. J Clin Invest 88:414–

the manner in which HGF exerts biological actions spe- 419, 19888. Zarnegar R, Michalopoulos GK: Purification and biologicalcifically in the injured kidney requires further study. Reg-

characterization of human hepatopoietin A, a polypeptide growthulation of the c-Met receptor function may be involved factor for hepatocytes. Cancer Res 49:3314–3320, 1989in this issue. Thus, renotropic systems could be achieved 9. Nakamura T, Nishizawa T, Hagiya M, et al: Molecular cloning

and expression of human hepatocyte growth factor. Nature 342:by time-dependent and cell type-specific activation of440–443, 1989ligand-receptor systems in growth factor networks that 10. Tashiro K, Hagiya M, Nishizawa T, et al: Deduced primary

orchestrate reconstruction processes of the injured kid- structure of rat hepatocyte growth factor and expression of themRNA in rat tissues. Proc Natl Acad Sci USA 87:3200–3204, 1990ney. A better comprehension of these issues will provide

11. Miyazawa K, Shimomura T, Kitamura A, et al: Molecular cloninga paradigm concerning tissue regeneration in biological and sequence analysis of the cDNA for a human serine proteasedefense systems. responsible for activation of hepatocyte growth factor. Structural

similarity of the protease precursor to blood coagulation factorPreclinical and clinical trials of HGF or HGF geneXII. J Biol Chem 268:10024–10028, 1993therapy for treatment of patients with renal diseases can 12. Naldini L, Vigna E, Bardelli A, et al: Biological activation of

be designed if a careful evaluation of the side effects is pro HGF (hepatocyte growth factor) by urokinase is controlledby a stoichiometric reaction. J Biol Chem 270:603–611, 1995made. The first clinical trials of HGF gene therapy for

13. Nakamura T: Structure and function of hepatocyte growth factor.patients with arteriosclerosis obliterans, myocardial in- Prog Growth Factor Res 3:67–85, 1991farction, and liver cirrhosis are in the planning stage. 14. Zarnegar R, Michalopoulos GK: The many faces of hepatocyte

growth factor: From hepatopoiesis to hematopoiesis. J Cell Biol129:1177–1180, 1995

ACKNOWLEDGMENTS 15. Matsumoto K, Nakamura T: Emerging multipotent aspects ofhepatocyte growth factor. J Biochem 119:591–600, 1996The work in the authors’ laboratory was supported by Research

16. Matsumoto K, Nakamura T: Hepatocyte growth factor (HGF)Grants from the Ministry of Education, Science, Sports, and Cultureas a tissue organizer for organogenesis and regeneration. Biochemof Japan, the Ministry of Health and Welfare of Japan, and by aBiophys Res Commun 239:639–644, 1997Research Grant from the Osaka Kidney Foundation. We are grateful

17. Kopp JB: Hepatocyte growth factor: Mesenchymal signal for epi-to M. Ohara for helpful comments.thelial homeostasis. Kidney Int 54:1392–1393, 1998

18. Balkovets DF, Lipschutz JH: Hepatocyte growth factor and theReprint requests to Toshikazu Nakamura, Ph.D., Biomedical Re-kidney: It is just not for the liver. Int Rev Cytol 186:225–260, 1999search Center, Osaka University Graduate School of Medicine, 2-2

19. Gustavo A, Hoeflich A, Jehle PM: Hepatocyte growth factorYamada-oka, Suita, Osaka 565-0871, Japan.in renal failure: Promise and reality. Kidney Int 57:1426–1436,E-mail: [email protected]

20. Furlong RA, Takehara T, Taylor WG, et al: Comparison ofbiological and immunochemical properties indicates that scatter

APPENDIX factor and hepatocyte growth factor are indistinguishable. J CellSci 100:173–177, 1991Abbreviations used in this article are: a-SMA, a-smooth muscle 21. Konishi T, Takehara T, Tsuji T, et al: Scatter factor from humanactin; BUN, blood urea nitrogen; ECM, extracellular matrix; EGF,embryonic fibroblasts is probably identical to hepatocyte growthepidermal growth factor; ERK, extracellular signal-regulated kinase; factor. Biochem Biophys Res Commun 180:765–773, 1991FAK, focal adhesion kinase; FGF-1, fibroblast growth factor-1; FK506, 22. Weidner KM, Arakaki N, Hartmann G, et al: Evidence for

tacrolimus; HB-EGF, heparin binding-epidermal growth factor; HGF, identity of human scatter factor and human hepatocyte growthhepatocyte growth factor; IGF, insulin-like growth factor; MDCK, factor. Proc Natl Acad Sci USA 88:7001–7005, 1991Madin-Darby canine kidney; MMP, matrix metalloprotease; PDGF, 23. Stoker M, Perryman M: An epithelial scatter factor released byplatelet-derived growth factor; PGE, prostaglandin E; PI-3, phosphati- embryo fibroblasts. J Cell Sci 77:209–223, 1985dylinositol-5-bisphosphate 3-kinase; PLC-g, phospholipase C-g; Ras- 24. Gherardi E, Gray J, Stoker M, et al: Purification of scatterGAP, Ras-GTPase activating protein; SH, src homology; TGF-b, trans- factor, a fibroblast-derived basic protein that modulates epithelialforming growth factor-b; uPA, urokinase-type plasminogen activator. interaction and movement. Proc Natl Acad Sci USA 86:5844–5848,

198925. Weidner KM, Behrens J, Vanderkerckhove J, Birchmeier W:REFERENCES

Scatter factor: Molecular characteristics and effects on the inva-1. Preuss HG, Goldin H: A renotropic system in rats. J Clin Invest siveness of epithelial cells. J Cell Biol 111:2097–2108, 1990

26. Pepper MS, Matsumoto K, Nakamura T, et al: Hepatocyte growth57:94–101, 1976

Page 13: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems 2035

factor induces urokinase-type plasminogen activator and plasmin- ies on the scattering of keratinocytes. Cell Struct Funct 18:117–124,1993ogen activator inhibitor-1 in MDCK epithelial cells. J Biol Chem

267:20493–20496, 1992 46. Shibamoto S, Hayakawa M, Takeuchi K, et al: Tyrosine phos-phorylation of b-catenin and plakoglobin enhanced by hepatocyte27. Vande Woude GF, Jeffers M, Cortner J, et al: Met-HGF/SF,

tumorigenesis, invasion and metastasis, in Plasminogen-Related growth factor and epidermal growth factor in human carcinomacells. Cell Adhes Commun 1:295–305, 1994Growth Factors, edited by Gherardi E, New York, John Wiley &

Sons, 1997, pp 119–132 47. Bowers DC, Fan S, Walter KA, et al: Scatter factor/hepatocytegrowth factor protects against cytotoxic death in human glioblas-28. Kadono Y, Shibahara K, Namiki M, et al: Membrane type-matrix

metalloproteinase is involved in the formation of hepatocyte toma via phosphatidylinositol 3-kinase- and Akt-dependent path-ways. Cancer Res 60:4277–4283, 2000growth factor/scatter factor-induced branching tubules in Mardin-

Darby canine kidney epithelial cells. Biochem Biophys Res Com- 48. Kosai K, Matsumoto K, Nagata S, et al: Hepatocyte growthfactor abrogates Fas-induced lethal liver apoptosis in mice. Bio-mun 251:681–687, 1998

29. Jiang WG, Hiscox S, Matsumoto K, Nakamura T: Hepatocyte chem Biophys Res Commun 244:683–690, 199849. Kosai K, Matsumoto K, Nakamura T: Hepatocyte growth factorgrowth factor/scatter factor, its molecular, cellular and clinical

implications in cancer. Crit Rev Oncol Hematol 29:209–248, 1999 prevents endotoxin-induced lethal hepatic failure in mice. Hepa-tology 30:151–159, 199930. Bottaro DP, Rubin JS, Faletto DL, et al: Identification of hepa-

tocyte growth factor receptor as the c-met proto-oncogene prod- 50. Schmidt C, Bladt F, Goedecke S, et al: Scatter factor/hepatocytegrowth factor is essential for liver development. Nature 373:699–uct. Science 251:802–804, 1991

31. Naldini L, Vigna E, Narsimhan RP, et al: Hepatocyte growth 702, 199551. Uehara Y, Minowa O, Mori C, et al: Placental defect and embry-factor (HGF) stimulates the tyrosine kinase activity of the recep-

tor encoded by the proto-oncogene c-MET. Oncogene 6:501–504, onic lethality in mice lacking hepatocyte growth factor/scatterfactor. Nature 373:702–705, 19951991

32. Park M, Dean M, Kaul K, et al: Sequence of MET protooncogene 52. Bladt F, Riethmacher D, Isenmann S, et al: Essential role forthe c-met receptor in the migration of myogenic precursor cellscDNA has features characteristic of the tyrosine kinase family

of growth factor receptors. Proc Natl Acad Sci USA 84:6379–6383, into the limb bud. Nature 376:768–771, 199553. Birchmeier C, Gherardi E: Developmental roles of HGF/SF and1987

33. Ponzetto C, Bardelli A, Zhen Z, et al: A multifunctional dock- its receptor, the c-Met tyrosine kinase. Trends Cell Biol 8:404–409,1998ing site mediates signaling and transformation by the hepatocyte

growth factor/scatter factor receptor family. Cell 77:261–271, 1994 54. Ebens A, Brose K, Leonard ED, et al: Hepatocyte growth factor/scatter factor is an axonal chemoattractant and neurotrophic fac-34. Zhu H, Naujoskas MA, Fixman ED, et al: Tyrosine 1356 in the

carboxyl-terminal tail of the HGF/SF receptor is essential for the tor for spinal motor neurons. Neuron 17:1157–1172, 199655. Matsumoto K, Nakamura T: Hepatocyte growth factor in renaltransduction of signals for cell motility and morphogenesis. J Biol

Chem 269:29943–29948, 1994 regeneration, renal disease and potential therapeutics. Curr OpinNephrol Hypertens 9:395–402, 200035. Weidner KM, Sachs M, Riethmacher D, Birchmeier W: Muta-

tion of juxtamembrane tyrosine residue 1001 suppresses loss-of- 56. Ishiki Y, Ohnishi H, Muto Y, et al: Direct evidence that hepato-cyte growth factor is a hepatotrophic factor for liver regenerationfunction mutations of the met receptor in epithelial cells. Proc

Natl Acad Sci USA 92:2597–2601, 1995 and for potent anti-hepatitis action in vivo. Hepatology 16:1227–1235, 199236. Schaeper U, Gehring NH, Fuchs KP, et al: Coupling of gab1 to

c-met, grb2, and shp2 mediates biological responses. J Cell Biol 57. Matsuda Y, Matsumoto K, Ichida T, Nakamura T: Hepatocytegrowth factor suppresses the onset of liver cirrhosis and abrogates149:1419–1432, 2000

37. Weidner KM, Di Cesare S, Sachs M, et al: Interaction between lethal hepatic dysfunction in rats. J Biochem 118:643–649, 199558. Roos F, Ryan AM, Chamow SM, et al: Induction of liver growthGab1 and the c-Met receptor tyrosine kinase is responsible for

epithelial morphogenesis. Nature 384:173–176, 1996 in normal mice by infusion of hepatocyte growth factor/scatterfactor. Am J Physiol 268:G380–G386, 199538. Maroun CR, Naujokas MA, Holgado-Madruga M, et al: The

tyrosine phosphatase SHP-2 is required for sustained activation 59. Yasuda H, Imai E, Shiota A, et al: Anti-fibrogenic effect of adeletion variant of hepatocyte growth factor on liver fibrosis inof extracellular signal-regulated kinase and epithelial morphogen-

esis downstream from the Met receptor tyrosine kinase. Mol Cell rats. Hepatology 24:636–642, 199660. Ohmichi H, Matsumoto K, Nakamura T: In vivo mitogenic actionBiol 20:8513–8525, 2000

39. Kotelevets L, Noe V, Bruyneel E, Myssiakine E, et al: Inhibi- of hepatocyte growth factor on lung epithelial cells: Pulmotrophicrole in lung regeneration. Am J Physiol 270:L1031–L1039, 1996tion by platelet-activating factor of Src- and hepatocyte growth

factor-dependent invasiveness of intestinal and kidney epithelial 61. Matsuda Y, Matsumoto K, Yamada A, et al: Preventive andtherapeutic effects in rats of hepatocyte growth factor infusioncells: Phosphatidylinositol 39-kinase is a critical mediator of tumor

invasion. J Biol Chem 273:14138–14145, 1998 on liver fibrosis/cirrhosis. Hepatology 26:81–89, 199762. Schmassmann A, Stettler C, Poulsom R, et al: Roles of hepato-40. Khwaja A, Lehmann K, Marte BM, Downward J: Phosphoino-

sitide 3-kinase induces scattering and tubulogenesis in epithelial cyte growth factor and receptor c-Met during gastric ulcer healingin rats. Gastroenterology 113:1858–1872, 1997cells through a novel pathway. J Biol Chem 273:18793–18801,

1998 63. Yaekashiwa M, Nakayama S, Ohnuma K, et al: Simultaneousor delayed administration of hepatocyte growth factor (HGF)41. Takaishi K, Sasaki T, Kato M, et al: Involvement of Rho p21

small GTP-binding protein and its regulator in the HGF-induced equally repress the fibrotic change in murine lung injury inducedby bleomycin: A morphologic study. Am J Respir Crit Care Medcell motility. Oncogene 9:273–279, 1994

42. Ridley AJ, Comoglio PM, Hall A: Regulation of scatter factor/ 156:1937–1944, 199764. Wakitani S, Imoto T, Kimura T, et al: Hepatocyte growth factorhepatocyte growth factor responses by Ras, Rac, and Rho in

MDCK cells. Mol Cell Biol 15:1110–1122, 1995 facilitates cartilage repair: Full thickness articular cartilage defectstudied in rabbit knee. Acta Orthop Scand 68:474–480, 199743. Matsumoto K, Matsumoto K, Nakamura T, Kramer RH: Hepa-

tocyte growth factor/scatter factor induces tyrosine phosphoryla- 65. Van Belle E, Witzenbichler B, Chen D, et al: Potential angio-genic effect of scatter factor/hepatocyte growth factor via induc-tion of focal adhesion kinase (p125FAK) and promotes migration

and invasion by oral squamous cell carcinoma cells. J Biol Chem tion of vascular endothelial growth factor: The case for paracrineamplification of angiogenesis. Circulation 97:381–390, 1998269:31807–31813, 1994

44. Lai JF, Kao SC, Jiang ST, et al: Involvement of focal adhesion 66. Miyazawa T, Matsumoto K, Ohmichi H, et al: Protection ofhippocampal neurons from ischemia-induced delayed neuronalkinase in hepatocyte growth factor-induced scatter of Madin-

Darby canine kidney cells. J Biol Chem 275:7474–7480, 2000 death by hepatocyte growth factor: A novel neurotrophic factor.J Cereb Blood Flow Metab 18:345–348, 199845. Watabe M, Matsumoto K, Nakamura T, Takeichi M: Coopera-

tive action of hepatocyte growth factor and anti-cadherin antibod- 67. Masunaga H, Fujise N, Shiota A, et al: Preventive effects of the

Page 14: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems2036

deleted form of hepatocyte growth factor against various liver 88. Yo Y, Morishita R, Yamamoto K, et al: Actions of hepatocytegrowth factor as a local modulator in the kidney: Potential roleinjuries. Eur J Pharmacol 342:267–279, 1998

68. Kaido T, Yoshikawa A, Seto S, et al: Portal branch ligation with in pathogenesis of renal disease. Kidney Int 53:50–58, 199889. Kolatsi-Joannou M, Woolf AS, Hardman P, et al: The hepato-a continuous hepatocyte growth factor supply makes extensive

hepatectomy possible in cirrhotic rats. Hepatology 28:756–760, cyte growth factor/scatter factor (HGF/SF) receptor, met trans-duces a morphogenetic signal in renal glomerular fibromuscular1998

69. Arisawa H, Yamashita Y, Ogawa H, et al: Deleted form of mesangial cells. J Cell Sci 108:3703–3714, 199590. Kallincos NC, Pollard AN, Couper JJ: Evidence for a func-hepatocyte growth factor ameliorates the mortality rate of severe

thermal injury in rats. Surgery 126:925–932, 1999 tional hepatocyte growth factor receptor in human mesangialcells. Regul Pept 74:137–142, 199870. Morishita R, Nakamura S, Hayashi S, et al: Therapeutic angio-

genesis induced by human recombinant hepatocyte growth factor 91. Montesano R, Schaller G, Orci L: Induction of epithelial mor-phogenesis in vitro by fibroblast-derived soluble factors. Cellin rabbit hind limb ischemia model as cytokine supplement ther-

apy. Hypertension 33:1379–1384, 1999 66:697–711, 199192. Montesano R, Matsumoto K, Nakamura T, Orci L: Identifica-71. Ueki T, Kaneda Y, Tsutsui H, et al: Hepatocyte growth factor

gene therapy of liver cirrhosis in rats. Nat Med 5:226–230, 1999 tion of a fibroblast-derived epithelial morphogen as hepatocytegrowth factor. Cell 67:901–908, 199172. Tahara M, Matsumoto K, Nukiwa T, Nakamura T: Hepatocyte

growth factor leads to recovery from alcohol-induced fatty liver 93. Santos OF, Nigam SK: HGF-induced tubulogenesis and branch-ing of epithelial cells is modulated by extracellular matrix andin rats. J Clin Invest 103:313–320, 1999

73. Yasuda S, Noguchi T, Gohda M, et al: Single low-dose adminis- TGF-b. Dev Biol 160:293–302, 199394. Santos OF, Moura LA, Rosen EM, Nigam SK: Modulation oftration of human recombinant hepatocyte growth factor attenu-

ates intimal hyperplasia in a balloon-injured rabbit iliac artery HGF-induced tubulogenesis and branching by multiple phosphor-ylation mechanisms. Dev Biol 159:535–548, 1993model. Circulation 101:2546–2549, 2000

74. Sato M, Kakubari M, Kawamura M, et al: The decrease in total 95. Bowes RC III, Lightfoot RT, Van De Water B, Stevens JL:Hepatocyte growth factor induces tubulogenesis of primary renalcollagen fibers in the liver by hepatocyte growth factor after

formation of cirrhosis induced by thioacetamide. Biochem Phar- proximal epithelial cells. J Cell Physiol 180:81–90, 199996. Grobstein C: Trans-filter induction of tubules in mouse meta-macol 59:681–690, 2000

75. Okura Y, Arimoto H, Tamura N, et al: Analysis of neurotrophic nephrogenic mesenchyme. Exp Cell Res 10:424–440, 195697. Saxen L, Lehtonen E: Embryonic kidney in organ culture. Differ-effects of hepatocyte growth factor in the adult hypoglossal nerve

axotomy model. Eur J Neurosci 11:4130–4144, 1999 entiation 36:2–11, 198798. Sonnenberg E, Meyer D, Weidner KM, Birchmeier C: Scatter76. Aoki M, Morishita R, Taniyama Y, et al: Angiogenesis induced

by hepatocyte growth factor in non-infarcted myocardium and factor/hepatocyte growth factor and its receptor, the c-met tyro-sine kinase, can mediate a signal exchange between mesenchymeinfarcted myocardium: Up-regulation of essential transcription

factor for angiogenesis, ets. Gene Ther 7:417–427, 2000 and epithelia during mouse development. J Cell Biol 123:223–235,199377. Bussolino F, Direnzo MF, Ziche M, et al: Hepatocyte growth

factor is a potent angiogenic factor which stimulates endothelial 99. Santos OF, Barros EJ, Yang XM, et al: Involvement of hepato-cyte growth factor in kidney development. Dev Biol 159:252–529,cell motility and growth. J Cell Biol 119:629–641, 1992

78. Matsumoto K, Date K, Ohmichi H, Nakamura T: HGF in lung 1994100. Woolf AS, Kolatsi-Joannou M, Hardman P, et al: Role ofmorphogenesis and tumor invasion: Roles as a mediator in epithe-

lial-mesenchymal and tumor–stromal interactions. Cancer Chemo- hepatocyte growth factor/scatter factor and the met receptor inthe early development of the metanephros. J Cell Biol 128:171–ther Pharmacol 38:S42–S47, 1996

79. Schmidt L, Duh F, Chan F, et al: Germline and somatic mutations 184, 1995101. Karp SL, Ortiz-Arduan A, Li S, Neilson EG: Epithelial differ-in the tyrosine kinase domain of the MET proto-oncogene in

papillary renal carcinomas. Nat Genet 16:68–73, 1997 entiation of metanephric mesenchymal cells after stimulation withhepatocyte growth factor or embryonic spinal cord. Proc Natl80. Schmidt L, Junker K, Weirich G, et al: Two North American

families with hereditary papillary renal carcinoma and identical Acad Sci USA 91:5286–5290, 1994102. Yo Y, Morishita R, Nakamura S, et al: Potential role of hepato-novel mutations in the MET proto-oncogene. Cancer Res 58:1719–

1722, 1998 cyte growth factor in the maintenance of renal structure: Anti-apoptotic action of HGF on epithelial cells. Kidney Int 54:1128–81. Jeffers M, Schmidt L, Nakagawa N, et al: Activating mutations

for the Met tyrosine kinase receptor in human cancer. Proc Natl 1138, 1998103. Liu Y, Sun AM, Dworkin LD: Hepatocyte growth factor protectsAcad Sci USA 14:11445–11450, 1997

82. Igawa T, Kanda S, Kanekake H, et al: Hepatocyte growth factor renal epithelial cells from apoptotic cell death. Biochem BiophysRes Commun 246:821–826, 1998is a potent mitogen for cultured rabbit renal tubular epithelial

cells. Biochem Biophys Res Commun 174:831–838, 1991 104. Hammerman MR: Growth factors and apoptosis in acute renalinjury. Curr Opin Nephrol Hypertens 7:419–424, 199883. Kan M, Zhang GH, Zarnegar R, et al: Hepatocyte growth factor/

hepatopoietin A stimulates the growth of rat kidney proximal 105. Liu Y: Hepatocyte growth factor promotes renal epithelial cellsurvival by dual mechanisms. Am J Physiol 277:F624–F633, 1999tubule epithelial cells (RPTE), rat nonparenchymal liver cells,

human melanoma cells, mouse keratinocytes and stimulates an- 106. Bardelli A, Longati P, Albero D, et al: HGF receptor associateswith the anti-apoptotic protein BAG-1 and prevents cell death.chorage-independent growth of SV-40 transformed RPTE. Bio-

chem Biophys Res Commun 174:331–337, 1991 EMBO J 15:6205–6212, 1996107. Igawa T, Matsumoto K, Kanda S, et al: Hepatocyte growth84. Harris RC, Burns KD, Alattar M, et al: Hepatocyte growth

factor stimulates phosphoinositide hydrolysis and mitogenesis in factor may function as a renotropic factor for regeneration in ratswith acute renal injury. Am J Physiol 265:F61–F69, 1993cultured renal epithelial cells. Life Sci 52:1091–1100, 1993

85. Kawaguchi M, Kawashima F, Ohshima K, et al: Hepatocyte 108. Mizuno S, Matsumoto K, Kurosawa T, et al: Reciprocal balanceof hepatocyte growth factor and transforming growth factor-b1growth factor is a potent promoter of mitogenesis in cultured rat

visceral glomerular epithelial cells. Cell Mol Biol 40:1103–1111, in renal fibrosis in mice. Kidney Int 57:937–948, 2000109. Mizuno S, Matsumoto K, Wen J, Nakamura T: Hepatocyte1994

86. Ishibashi K, Sasaki S, Sakamoto H, et al: Hepatocyte growth growth factor suppresses interstitial fibrosis in a mouse model ofobstructive nephropathy. Kidney Int 59:1304–1314, 2001factor is a paracrine factor for renal epithelial cells: Stimulation

of DNA synthesis and Na,K-ATPase activity. Biochem Biophys 110. Nagaike M, Hirao S, Tajima H, et al: Renotropic functions ofhepatocyte growth factor in renal regeneration after unilateralRes Commun 182:960–965, 1992

87. Cantley LG, Barros EJ, Gandhi M, et al: Regulation of mitogen- nephrectomy. J Biol Chem 266:22781–22784, 1991111. Ishibashi K, Sasaki S, Sakamoto H, et al: Expression of receptoresis, motogenesis, and tubulogenesis by hepatocyte growth factor

in renal collecting duct cells. Am J Physiol 267:F271–F280, 1994 gene for hepatocyte growth factor in kidney after unilateral ne-

Page 15: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems 2037

phrectomy and renal injury. Biochem Biophys Res Commun potent inhibitor of hepatocyte growth factor secretion by humanfibroblasts. Cell Biol Int Rep 16:917–926, 1992187:1454–1459, 1992

112. Goto T, Sugimura K, Harimoto K, et al: Hepatocyte growth 133. Nakano N, Morishita R, Moriguchi A, et al: Negative regulationof local hepatocyte growth factor (HGF) expression by angioten-factor in glycerol-induced acute renal failure. Nephron 77:440–

444, 1997 sin II and transforming growth factor-b in blood vessels: Potentialrole of HGF in cardiovascular disease. Hypertension 32:444–451,113. Yano T, Yano Y, Horikawa S, et al: Regenerative response in

acute renal failure due to vitamin E deficiency and gluthathione 1998134. Broten J, Michalopoulos G, Petersen B, Cruise J: Adrenergicdepletion in rats. Biochem Pharmacol 56:543–546, 1998

114. Liu Y, Tolbert EM, Lin L, et al: Up-regulation of hepatocyte stimulation of hepatocyte growth factor expression. Biochem Bio-phys Res Commun 262:76–79, 1999growth factor receptor: An amplification and targeting mechanism

for hepatocyte growth factor action in acute renal failure. Kidney 135. Tajima H, Higuchi O, Mizuno K, Nakamura T: Tissue distribu-tion of hepatocyte growth factor receptor and its exclusive down-Int 55:442–453, 1999

115. Oka A, Tanji N, Miyauchi Y, Yokoyama M: Expression of regulation in a regenerating organ after injury. J Biochem 111:401–406, 1992growth factors after the release of ureteral obstruction in the rat

kidney. Int J Urol 6:607–615, 1999 136. Yanagita K, Matsumoto K, Sekiguchi K, et al: Hepatocytegrowth factor may act as a pulmotrophic factor on lung regenera-116. Libetta C, Rampino T, Esposito C, et al: Stimulation of hepatocyte

growth factor in human acute renal failure. Nephron 80:41–45, tion after acute lung injury. J Biol Chem 268:21212–21217, 1993137. Takehara T, Matsumoto K, Nakamura T: Cell density-depen-1998

117. Nishida M, Kawamatsu H, Ishiwari K, et al: Serum hepatocyte dent regulation of albumin synthesis and DNA synthesis in rathepatocytes by hepatocyte growth factor. J Biochem 112:330–334,growth factor levels in patients with renal diseases. Am J Nephrol

19:509–512, 1999 1992138. Mizuno K, Higuchi O, Tajima H, et al: Cell density-dependent118. Takada S, Namiki M, Takahara S, et al: Serum HGF levels in

acute renal rejection after living-related renal transplantation. regulation of hepatocyte growth factor receptor on adult rat hepa-tocytes in primary culture. J Biochem 114:96–102, 1993Transplant Int 9:151–154, 1996

119. Yanagita K, Nagaike M, Ishibashi H, et al: Lung may have an 139. Miyazawa K, Shimomura T, Kitamura N: Activation of hepato-cyte growth factor in the injured tissues is mediated by hepatocyteendocrine function producing hepatocyte growth factor in re-

sponse to injury of distal organs. Biochem Biophys Res Commun growth factor activator. J Biol Chem 271:3615–3618, 1996140. Border WA, Noble NA: Transforming growth factor-b in tissue182:802–809, 1992

120. Kono S, Nagaike M, Matsumoto K, Nakamura T: Marked induc- fibrosis. N Engl J Med 331:1286–1292, 1994141. Peters H, Noble NA, Boder WA: Transforming growth factor btion of hepatocyte growth factor mRNA in intact kidney and

spleen in response to injury of distant organs. Biochem Biophys in human glomerular injury. Curr Opin Nephrol Hypertens 6:389–393, 1997Res Commun 186:991–998, 1992

121. Miyazawa K, Shimomura T, Naka D, Kitamura N: Proteolytic 142. Border WA, Noble NA: Interactions of transforming growthfactor-b and angiotensin II in renal fibrosis. Hypertension 31:181–activation of hepatocyte growth factor in response to tissue injury.

J Biol Chem 269:8966–8970, 1994 188, 1998143. Sanderson N, Factor V, Nagy P, et al: Hepatic expression of122. Ono K, Matsumori A, Shioi T, et al: Enhanced expression of

hepatocyte growth factor/c-Met by myocardial ischemia and re- mature transforming growth factor-b1 in transgenic mice resultsin multiple tissue lesions. Proc Natl Acad Sci USA 92:2572–2576,perfusion in a rat model. Circulation 95:2552–2558, 1997

123. Matsumoto K, Tajima H, Hamanoue M, et al: Identification and 1995144. Kopp JB, Factor VM, Mozes M, et al: Transgenic mice withcharacterization of “injurin,” an inducer of the gene expression of

hepatocyte growth factor. Proc Natl Acad Sci USA 89:3800–3804, increased plasma levels of TGF-b1 develop progressive renaldisease. Lab Invest 74:991–1003, 19961992

124. Matsumoto K, Okazaki H, Nakamura T: Up-regulation of hepa- 145. Clouthier DE, Comerford SA, Hammer RE: Hepatic fibrosis,glomerulosclerosis, and a lipodystrophy-like syndrome in PEPCK-tocyte growth factor mRNA by interleukin-1 in human skin fi-

broblasts. Biochem Biophys Res Commun 188:235–243, 1992 TGF-b1 transgenic mice. J Clin Invest 100:2697–2713, 1997146. Mozes MM, Bottinger EP, Jacot TA, Kopp JB: Renal expression125. Tamura M, Arakaki N, Tsubouchi H, et al: Enhancement of

human hepatocyte growth factor production by interleukin-1a of fibrotic matrix proteins and of transforming growth factor-b(TGF-b) isoforms in TGF-b transgenic mice. J Am Soc Nephroland -1b and tumor necrosis factor-a by fibroblasts in culture.

J Biol Chem 268:8140–8145, 1993 10:271–280, 1999147. Isaka Y, Fujiwara Y, Ueda N, et al: Glomerulosclerosis induced126. Matsumoto K, Okazaki H, Nakamura T: Novel function of pros-

taglandins as inducers of gene expression of HGF and putative by in vivo transfection of transforming growth factor-beta orplatelet-derived growth factor gene into the rat kidney. J Clinmediators of tissue regeneration. J Biochem 117:458–464, 1995

127. Seslar SP, Nakamura T, Byers SW: Regulation of fibroblast Invest 92:2597–2601, 1993148. Kelly FJ, Anderson S, Thompson MM, et al: Acute and chronichepatocyte growth factor/scatter factor expression by human

breast carcinoma cell lines and peptide growth factors. Cancer renal effects of recombinant human TGF-b2 in the rat. J Am SocNephrol 10:1264–1273, 1999Res 53:1233–1238, 1993

128. Gohda E, Matsunaga T, Kataoka H, et al: Induction of hepato- 149. Okuda S, Languino LR, Ruoslahti E, Border WA: Elevatedexpression of transforming growth factor-b and proteoglycan pro-cyte growth factor in human skin fibroblasts by epidermal growth

factor, platelet-derived growth factor and fibroblast growth factor. duction in experimental glomerulonephritis: Possible role inexpansion of the mesangial extracellular matrix. J Clin InvestCytokine 6:633–640, 1994

129. Nakamura T, Matsumoto K, Kiritoshi A, et al: Induction of 86:453–462, 1990150. Kaneto H, Morrissey J, Klahr S: Increased expression ofhepatocyte growth factor in fibroblasts by tumor-derived factors

affects invasive growth of tumor cells: In vitro analysis of tumor- TGF-b1 mRNA in the obstructed kidney of rats with unilateralureteral ligation. Kidney Int 44:313–321, 1993stromal interactions. Cancer Res 57:3305–3313, 1997

130. Kiso S, Kawata S, Tamura S, et al: Role of heparin-binding 151. Yamamoto T, Noble NA, Miller DE, Border WA: Sustainedexpression of TGF-b1 underlies development of progressive kid-epidermal growth factor-like growth factor as a hepatotrophic

factor in rat liver regeneration after partial hepatectomy. Hepatol- ney fibrosis. Kidney Int 45:916–927, 1994152. Mizuno S, Kurosawa T, Matsumoto K, et al: Hepatocyte growthogy 22:1584–1590, 1995

131. Matsumoto K, Tajima H, Okazaki H, Nakamura T: Negative factor prevents renal fibrosis and dysfunction in a mouse modelof chronic renal disease. J Clin Invest 101:1827–1834, 1998regulation of hepatocyte growth factor gene expression in human

lung fibroblasts and leukemic cells by transforming growth 153. Yoshioka K, Takemura T, Murakami K, et al: Transforminggrowth factor-b protein and mRNA in glomeruli in normal andfactor-b1 and glucocorticoids. J Biol Chem 267:24917–24920, 1992

132. Gohda E, Matsunaga T, Kataoka H, Yamamoto I: TGF-b is a diseased human kidneys. Lab Invest 68:154–163, 1993

Page 16: Hepatocyte growth factor: Renotropic role and potential therapeutics for renal diseases

Matsumoto and Nakamura: HGF in renotropic systems2038

154. Yamamoto T, Nakamura T, Noble NA, et al: Expression of trans- genic mice overexpressing hepatocyte growth factor/scatter fac-tor. Lab Invest 77:131–138, 1997forming growth factor-b is elevated in human and experimental

170. Takayama H, Larochelle WJ, Sharp R, et al: Diverse tumorigen-diabetic nephropathy. Proc Natl Acad Sci USA 90:1814–1818, 1993esis associated with aberrant development in mice overexpressing155. Cuhaci B, Kumar MS, Bloom RD, et al: Transforming growthhepatocyte growth factor/scatter factor. Proc Natl Acad Sci USAfactor-b levels in human allograft chronic fibrosis correlate with94:701–706, 1997rate of decline in renal function. Transplantation 68:785–790, 1999

171. Shiota G, Wang TC, Nakamura T, Schmidt EV: Hepatocyte156. Border WA, Border WA, Noble NA, et al: Natural inhibitorgrowth factor in transgenic mice: Effects on hepatocyte growth,of transforming growth factor-beta protects against scarring inliver regeneration and gene expression. Hepatology 19:962–972,experimental kidney disease. Nature 360:361–364, 19921994157. el Nahas AM, Muchaneta-Kubara EC, Essawy M, Soylemez-

172. Kunisada T, Yamazaki H, Hirobe T, et al: Keratinocyte expres-oglu O: Renal fibrosis: Insights into pathogenesis and treatment.sion of transgenic hepatocyte growth factor affects melanocyteInt J Biochem Cell Biol 29:55–62, 1997development, leading to dermal melanocytosis. Mech Dev 94:67–158. Fan JM, Ng YY, Hill PA, et al: Transforming growth factor-b78, 2000regulates tubular epithelial-myofibroblast transdifferentiation in

173. Garcia-Ocana A, Takane KK, Syed MA, et al: Hepatocytevitro. Kidney Int 56:1455–1467, 1999 growth factor overexpression in the islet of transgenic mice in-159. Boukhalfa G, Desmouliere A, Rondeau E, et al: Relationship creases beta cell proliferation, enhanced islet mass, and induces

between alpha-smooth muscle actin expression and fibrotic changes mild hypoglycemia. J Biol Chem 275:1226–1232, 2000in human kidney. Exp Nephrol 4:241–247, 1996 174. Kawaida K, Matsumoto K, Shimazu H, Nakamura T: Hepato-

160. Muchaneta-Kubara EC, el Nahas AM: Myofibroblast pheno- cyte growth factor prevents acute renal failure and acceleratestypes expression in experimental renal scarring. Nephrol Dial renal regeneration in mice. Proc Natl Acad Sci USA 91:4357–4361,Transplant 12:904–915, 1997 1994

161. Pedagogos E, Hewitson T, Fraser I, et al: Myofibroblasts and 175. Miller SB, Martin DR, Kissane J, Hammerman MR: Hepato-arteriolar sclerosis in human diabetic nephropathy. Am J Kidney cyte growth factor accelerates recovery from acute ischemic renalDis 29:912–918, 1997 injury in rats. Am J Physiol 266:F129–F134, 1994

162. Liu Y, Rajur K, Tolbert E, Dworkin LD: Endogenous hepato- 176. Amaike H, Matsumoto K, Oka T, Nakamura T: Preventive effectof hepatocyte growth factor on acute side effects of cyclosporincyte growth factor ameliorates chronic renal injury by activatingA in mice. Cytokine 8:387–394, 1996matrix degradation pathways. Kidney Int 58:2028–2043, 2000

177. Takada S, Takahara S, Nishimura K, et al: Effect of hepatocyte163. Fang JW, Bird GL, Nakamura T, et al: Hepatocyte proliferationgrowth factor on tacrolimus-induced nephrotoxicity in spontane-as an indicator of outcome in acute alcoholic hepatitis. Lancetously hypertensive rats. Transplant Int 12:27–32, 1999343:820–823, 1994

178. Azuma H, Takahara S, Kitamura M, et al: Effect of hepatocyte164. Masuhara M, Yasunaga M, Tanigawa K, et al: Expression ofgrowth factor on chronic rejection in rat renal allografts. Trans-hepatocyte growth factor, transforming growth factor-a, andplant Proc 31:854–855, 1999transforming growth factor-b1 messenger RNA in various human

179. Azuma H, Takahara S, Matsumoto K, et al: Hepatocyte growthliver diseases and correlation with hepatocyte proliferation. Hepa-factor prevents development of chronic allograft nephropathy intology 24:323–329, 1996an experimental rat transplant model. J Am Soc Nephrol (in165. Sugimura K, Kim T, Goto T, et al: Serum hepatocyte growthpress)factor levels in patients with chronic renal failure. Nephron 70: 180. Azuma H, Nadeau K, Takada M, et al: Cellular and molecular

324–328, 1995 predictors of chronic renal dysfunction after initial ischemia/re-166. Seki T, Hagiya M, Shimonishi O, et al: Organization of the human perfusion injury of a single kidney. Transplantation 64:190–197,

hepatocyte growth factor-encoding gene. Gene 102:213–219, 1991 1997167. Miyazawa K, Kitamura A, Kitamura N: Structural organization 181. Takada M, Chandraker A, Nadean K, et al: The role of the B7

and the transcriptional initiation site of the human hepatocyte co-stimulatory pathway in experimental cold ischemia/reperfu-growth factor gene. Biochemistry 30:9170–9176, 1991 sion injury. J Clin Invest 100:1190–1203, 1997

168. Okajima A, Miyazawa K, Kitamura N: Characterization of the 182. Haug C, Grill C, Schmid-Kotsas A, et al: Endothelin releasepromotor region of the rat hepatocyte growth factor/scatter factor by rabbit proximal tubule cells: Modulatory effects of cyclosporinegene. Eur J Biochem 213:113–119, 1993 A, tacrolimus, HGF and EGF. Kidney Int 54:1626–1636, 1998

169. Takayama H, Larochelle WJ, Sabnis SG, et al: Renal tubular 183. Schena FP: Role of growth factors in acute renal failure. KidneyInt 53(Suppl 66):S11–S15, 1998hyperplasia, polycystic disease, and glomerulosclerosis in trans-