19
[Frontiers in Bioscience 3, d25-43, January 1, 1998] 25 HOW DO INTRACELLULAR PROTEOLYTIC SYSTEMS CHANGE WITH AGE? Ana Maria Cuervo and J. Fred Dice Department of Physiology Tufts University School of Medicine, Boston, USA Received 12/5/97 Accepted 12/9/97 TABLE OF CONTENTS 1. Abstract 2. Introduction: what happens to senescent cells? 3. Model systems for the study of aging 4. Intracellular proteolytic systems and aging 4.1. Cytosolic proteolytic systems: Proteasomes and calpains 4.1.1. Ubiquitin-proteasome system 4.1.2. Calpains 4.2. Lysosomal proteolytic pathways 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport of cytosolic proteins through the lysosomal membrane 5. Tissue-specific changes in proteolytic systems with aging 5.1. Central nervous system 5.2. Liver and cultured fibroblasts 5.3. Skeletal muscle 5.4. Eye lens 6. Conclusions and perspectives 7. Acknowledgments 8. References 1. ABSTRACT One of the common features of cells from senescent tissues is the accumulation of abnormal proteins. Several hypotheses have been proposed to explain the origin of those abnormal proteins. A defect in proteolytic systems usually responsible for the elimination of altered proteins from the cells could clearly contribute to such accumulation. Here we describe the effect of age on the major proteolytic systems within cells: the ubiquitin-proteasome pathway, the calcium- activated calpain pathways, and multiple lysosomal pathways. Our group has contributed to the characterization of a selective pathway of degradation of cytosolic proteins in lysosomes that is activated under conditions of nutrient deprivation. In this lysosomal pathway of proteolysis proteins are transported through the lysosomal membrane assisted by cytosolic and lysosomal molecular chaperones and a receptor protein in the lysosomal membrane. The activity of this pathway significantly decreases with age, and this decrease might account for the cytosolic accumulation of aberrant substrate proteins in senescent cells. The cellular consequences of the decline of this lysosomal pathway together with possible methods to restore the reduced function are also addressed in this review. 2. INTRODUCTION: WHAT HAPPENS TO SENESCENT CELLS? Aging is associated with a progressive reduction in almost all physiological functions. Senescent organisms show an impaired responsiveness to environmental stimuli and to physiological stress (1). A deterioration of the immune system in aging has also been described, and this immune system impairment is believed to contribute to increased mortality from infections, autoimmune diseases, and cancer in the elderly (reviewed in ref. 2). At the cellular level, aging results in the inability of cells to proliferate. Senescent cells resemble cells that are arrested at the G1-S boundary of the cell cycle (3). A blockage in the expression of the c-fos gene, which leads to changes in the AP-1(active gene regulatory protein), along with changes in CREBP (cyclic AMP response element binding protein) and CTF (CAAAT transcription factor) transcription factor complexes, are believed to contribute to the loss of cellular proliferation in senescent cells (4, 5). Age-related changes in the activity of several cell cycle regulatory components such as Rb (retinoblastoma protein), Cdk2 (cyclin-dependent kinase 2), p21 (inhibitor of cyclin-dependent kinases), and p53 (tumor suppresser protein) seem also to participate in the mechanism of proliferation arrest in aging (reviewed in ref. 6). Genetic studies have shown that the senescent phenotype is dominant, and at least four separate genes are involved in the suppression of cellular proliferation (7). Senescent cells remain metabolically active for long periods of time, but there are progressive changes in cell structure and function. For example, cells accumulate somatic mutations in their DNA and lipofuscin-containing dense bodies in their cytoplasm (8).

[Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

[Frontiers in Bioscience 3, d25-43, January 1, 1998]

25

HOW DO INTRACELLULAR PROTEOLYTIC SYSTEMS CHANGE WITH AGE?

Ana Maria Cuervo and J. Fred Dice

Department of Physiology Tufts University School of Medicine, Boston, USA

Received 12/5/97 Accepted 12/9/97

TABLE OF CONTENTS

1. Abstract2. Introduction: what happens to senescent cells?3. Model systems for the study of aging4. Intracellular proteolytic systems and aging

4.1. Cytosolic proteolytic systems: Proteasomes and calpains4.1.1. Ubiquitin-proteasome system4.1.2. Calpains

4.2. Lysosomal proteolytic pathways4.2.1. Fusion of lysosomes with vesicular compartments4.2.2. Macroautophagy and microautophagy4.2.3. Direct transport of cytosolic proteins through the lysosomal membrane

5. Tissue-specific changes in proteolytic systems with aging5.1. Central nervous system5.2. Liver and cultured fibroblasts5.3. Skeletal muscle5.4. Eye lens

6. Conclusions and perspectives7. Acknowledgments8. References

1. ABSTRACT

One of the common features of cells from senescenttissues is the accumulation of abnormal proteins. Severalhypotheses have been proposed to explain the origin of thoseabnormal proteins. A defect in proteolytic systems usuallyresponsible for the elimination of altered proteins from thecells could clearly contribute to such accumulation. Here wedescribe the effect of age on the major proteolytic systemswithin cells: the ubiquitin-proteasome pathway, the calcium-activated calpain pathways, and multiple lysosomal pathways.

Our group has contributed to the characterization ofa selective pathway of degradation of cytosolic proteins inlysosomes that is activated under conditions of nutrientdeprivation. In this lysosomal pathway of proteolysis proteinsare transported through the lysosomal membrane assisted bycytosolic and lysosomal molecular chaperones and a receptorprotein in the lysosomal membrane. The activity of thispathway significantly decreases with age, and this decreasemight account for the cytosolic accumulation of aberrantsubstrate proteins in senescent cells. The cellular consequencesof the decline of this lysosomal pathway together with possiblemethods to restore the reduced function are also addressed inthis review.

2. INTRODUCTION: WHAT HAPPENS TOSENESCENT CELLS?

Aging is associated with a progressive reduction inalmost all physiological functions. Senescent organisms

show an impaired responsiveness to environmental stimuli andto physiological stress (1). A deterioration of the immunesystem in aging has also been described, and this immunesystem impairment is believed to contribute to increasedmortality from infections, autoimmune diseases, and cancer inthe elderly (reviewed in ref. 2).

At the cellular level, aging results in the inability ofcells to proliferate. Senescent cells resemble cells that arearrested at the G1-S boundary of the cell cycle (3). A blockagein the expression of the c-fos gene, which leads to changes inthe AP-1(active gene regulatory protein), along with changesin CREBP (cyclic AMP response element binding protein) andCTF (CAAAT transcription factor) transcription factorcomplexes, are believed to contribute to the loss of cellularproliferation in senescent cells (4, 5). Age-related changes inthe activity of several cell cycle regulatory components such asRb (retinoblastoma protein), Cdk2 (cyclin-dependent kinase2), p21 (inhibitor of cyclin-dependent kinases), and p53 (tumorsuppresser protein) seem also to participate in the mechanismof proliferation arrest in aging (reviewed in ref. 6). Geneticstudies have shown that the senescent phenotype is dominant,and at least four separate genes are involved in the suppressionof cellular proliferation (7). Senescent cells remainmetabolically active for long periods of time, but there areprogressive changes in cell structure and function. Forexample, cells accumulate somatic mutations in their DNAand lipofuscin-containing dense bodies in their cytoplasm(8).

Page 2: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

26

Protein degradation together with proteinsynthesis take place continuously in all cells (9). Smallmodifications in the balance between these processes allowcells to rapidly adapt to changes in the extracellularenvironment (10). In addition to this continuous turnover ofproteins inside cells, abnormally synthesized proteins orproteins incorrectly modified are also eliminated from thecells by the proteolytic systems (11). Intracellular proteasesalso participate in many other fundamental cell processesincluding cell differentiation, cell cycle progression,antigen presentation, and intracellular traffic of proteins(reviewed in ref. 12).

Because of these many cellular functions inwhich the proteolytic systems participate, the consequencesof a reduction in intracellular protein degradation arewidespread. Along with a severe difficulty in adapting toenvironmental changes, cells with impaired proteindegradation are less able to eliminate abnormal or damagedproteins. Accumulation of those altered proteins may causea variety of further problems within cells. In addition,defects in the processes of intercellular communication (i.e.antigen presentation, hormone and peptide secretion) willmake those cells more vulnerable to the attack ofexogenous agents and unable to develop a coordinatedsystemic response. These changes may contribute tocommon phenotypes of senescent cells, including theinability to progress through the cell cycle (reviewed in ref.13).

Several hypotheses have been proposed about themechanisms responsible for aging. A detailed descriptionof each of these theories is beyond the scope of this review(for review see ref. 13). However, in general, these theoriescan be separated into catastrophic (aging is the result ofdamage accumulation throughout the cells' lifespan) andgenetic (aging is due to a genetic clock that causes age-related phenotypes in cells). Lately, a multifaceted originfor senescence is becoming generally accepted (14-16).Thus, along with a genetic program, the effect ofextracellular agents during the cellular lifespan modulatesthe severity of the age-related alterations. It is in thatmodulation that proteolytic systems play an important role.In many different tissues including liver, cardiac andskeletal muscle, and brain, a decline in protein degradationwith age has been described (17-22).

Several proteolytic systems participate inintracellular protein degradation (reviewed in ref. 23). Theymainly differ in their intracellular localization and in theregulation of their activities. We will consider theubiquitin-proteasome proteolytic pathway located in thecytosol and nucleus, the calpains located in cytosol andassociated with the cytoskeleton, and several differentpathways of lysosomal proteolysis. Although all of thesedegradation pathways are present in all cells, the activity ofeach of these systems varies from tissue to tissue and alsodepends on environmental conditions. Specific groups ofsubstrate proteins have been assigned to each of theseproteolytic pathways, but it is becoming apparent that someproteins can be substrates for multiple proteolytic systems.Thus, single proteins have been described to follow one or

another proteolytic pathway depending on the cellularconditions (i.e. c-fos and c-jun (24); IkB (25); membranereceptors (26), and components of one proteolytic systeminfluence the activities of other proteolytic systems (27-30). In the present work, we have first addressed what isknown about the effect of age on the main intracellularproteolytic systems, followed by several examples of howthose systems are differently affected by age dependingon the tissue analyzed.

3. MODEL SYSTEMS FOR THE STUDY OF AGING

There are many different model systems usedfor the study of aging. For some studies, it is possible towork with whole animals of different ages, andgenetically defined strains of mice and rats are commonlyutilized (31, 32; reviewed in ref. 33). A mouse model ofaccelerated senescence has also been established (34), andsenescence biomarkers on those animals have been verywell characterized (35-37; for review see ref. 38).Recently, transgenic mice have also proved to be useful inthe analysis of spontaneous mutations accumulatingduring aging (39). In addition, the insertional mutation ofa gene in one of those transgenic mice has allowed theidentification of a new gene, klotho, that is involved inthe suppression of several aging phenotypes (40).

In order to simplify the study of aging inorganisms, animal species with shorter lifespans thanmammals have also been used. Thus, the nematodeCaenorhabditis elegans, with a lifespan of 20 days and1% of the mammalian genome, has allowed an extensiveanalysis of the genetics of senescence (reviewed in ref.41). Some insects, such as Drosophila melanogaster, witha relatively short lifespan (1-3 months) but with moregenetically heterogeneous populations, have been utilizedfor the analysis of genetic influences on lifespan (41).

Some unicellular organisms, such as the yeastSaccharomyces cerevisiae, have considerably simplifiedstudies in aging. The highly developed molecular biologyof yeast and the ability to select for mutants in thismicroorganism have allowed the identification of severalgenes that show altered expression with age. Still, thesenescence of budding yeast is a process that differs inseveral respects from the senescence in multicellularorganisms (41).

The study of some cellular processes isdifficult to perform in whole aged animals, sincecompensatory mechanisms are frequently activated.Several in vitro models of aging have also beenestablished (reviewed in ref. 42). Specific cell typessuch as human diploid fibroblasts, endothelial cells,keratinocytes and lymphocytes have been shown to havefinite replicative lifespans in culture. DNA synthesisand cell division cease, but metabolic activity andcellular viability may be maintained for an extendedperiod of time (43). The loss of proliferation in these invitro systems is interpreted as an expression of aging atthe cellular level. The more relevant facts of thosesenescent cultures and their correlations with

Page 3: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

27

Table 1. Senescent fibroblasts as a model of agingCHARACTERISTICS OF SENESCENT CELLS IN CULTURE REFERENCE

q The proliferative potential of cells depends on the number of cell doublings undergone and is independent of thechronological age of the cells

44

q There is an inverse relationship between the age of a donor and the proliferative potential of their fibroblasts invitro

45

q There is a correlation between the proliferative potential of cells from different species and the average life spanof that species

46

q Cells from patients with premature aging disorders show reduced proliferative potential 47

q Senescent fibroblasts in culture display enlarged, flattened morphology and fail to replicate their DNA inresponse to normal growth stimuli

48

aging in complete organisms are summarized in table 1.Whether or not aged organisms contain cells that havereached the end of their proliferative potential has been acontroversial topic. However, the analysis of variant beta-galactosidase activities has recently been successfullyapplied to identify senescent cells in aging skin in vivo(49).

4. INTRACELLULAR PROTEOLYTIC SYSTEMSAND AGING

Senescent human fibroblasts in culture have beenvery helpful in identifying the defective proteolytic systemswith age. General techniques for the study of intracellularprotein degradation in cultured cells, such as metabolicradiolabeling of intracellular proteins or microinjection ofradiolabeled proteins, and further analysis of theirintracellular breakdown have demonstrated that totalprotein degradation was considerably slowed in oldfibroblasts when compared with young cells (19, 50-53).The addition of specific protease inhibitors to the culturemedium and/or the analysis of protein breakdown underspecific cellular conditions has been used to implicate theprotease or proteases responsible for the decrease in proteindegradation rates with age.

In recent years, several cell-free models havebeen developed to analyze independently the function ofsome of the major proteolytic systems of cells. Thus,ubiquitination of proteins can be accomplished in vitro withrabbit reticulocyte or Xenopus egg extracts (54, 55), the20S proteasome has been isolated from several differentorganisms and its proteolytic activities carefullycharacterized (56, 57), endosomal/ lysosomal fusion can bereproduced with isolated vesicles in vitro (58), anddirect transport of proteins through the lysosomalmembrane can be followed in isolated lysosomes (59-61).

4.1. Cytosolic proteolytic systems: Proteasomes andcalpains4.1.1. Ubiquitin-proteasome system

Several proteases have been described in thecellular cytosol. The 20S proteasome, a multicatalyticprotease complex, is without doubt one of the moststudied and best characterized cytosolic proteases. The20S proteasome is normally found associated withseveral heterogeneous proteins that form one or two 19S

caps at the ends of the 20S structure, that regulate itsproteolytic activity (62). This complex is called the 26Sproteasome (reviewed in ref. 63). The nature of many ofthese regulatory subunits remains unclear, but ATPase,ubiquitin-binding, and ubiquitin isopeptidase activities havebeen detected (64).

The 26S proteasome has been consideredmainly responsible for the degradation of short-livedand abnormal proteins in the cytosol and nucleus(reviewed in ref. 65). However, recent evidence for theparticipation of this proteolytic system in basal proteinturnover of cytosolic proteins has also been presented(66). In addition, not only cytosolic proteins, but alsopreviously compartmentalized proteins, can beredirected to the cytosol where they are degraded by the26S proteasome (67). Degradation of proteins by theproteasome usually requires a previous covalentmodification of the substrate protein in order to berecognized by the proteolytic complex. Thismodification, known as ubiquitination, consists of thecovalent attachment of a 8.6 kDa protein (ubiquitin) tothe epsilon-amino group of a lysine residue of thesubstrate protein (reviewed in ref. 68). Covalentattachment of ubiquitin to the protein substrate ismediated by a complex group of enzymes (activatingenzyme (E1), carrier/conjugating enzymes (E2) andconjugating enzymes (E3)) (see figure 1) (69). Ubiquitincan also be covalently attached to itself to form aubiquitin chain on a substrate protein that is thentargeted for degradation by the 26S proteasome (70).Degradation of polyubiquitinated proteins seems to befacilitated by the direct interaction of some subunits ofthe proteasome with the multiubiquitin chain (71, 72).Other enzymes are able to remove ubiquitin fromproteins (deubiquitination), allowing it to recycle (73,74). Ubiquitination is required for degradation of manyimportant regulatory proteins such as cyclins, c-fos, andp53 (68).

Although ubiquitination is the mostgeneralized target signal for protein degradation by the26S proteasome, ubiquitin-independent degradation ofsome proteins by the proteasome has also been reported(i.e. ornithine decarboxylase) (75). On the other hand,monoubiquitination of some membrane proteins in theircytosol-exposed region targets them for degradation bythe vacuolar (lysosomal) system in yeast (76, 77).

Page 4: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

28

Figure 1. The ubiquitin-proteasome system. Most short-livedand abnormal proteins in the cell cytosol are degraded by the26S proteasome, directly or after a covalent modificationknown as ubiquitination. The multiple enzymatic steps andseveral enzymes required for the ubiquitination of proteins aresummarized in this figure. See the text for details.

Regarding the proteolytic core of the proteasome,at least three different neutral peptidase activities have beendescribed (78). Recently, specific inhibitors for thismulticatalytic complex have been developed (79-81). Thephysiological activators described for the 20S proteasomeare located at the 19S ATPase complex. In addition, in thepresence of detergent, fatty acids, or polylysine, the activityof the proteasome significantly increases (82), but thephysiological significance of such regulation is not known.The proteasome normally exists in a latent state in the cell(65) which may be important in preventing uncontrolledproteolysis (78). Lately, several authors have demonstratedthat the subunit composition of the proteasome changesdepending on specific stimuli, and that these alteredsubunits result in changes in the various proteolyticactivities (83).

A decrease in a non-acidic protease activity withage has been reported in several types of cells (84, 85).The 26S proteasome is one possible candidate for thatproteolytic activity. However, no clear evidence for suchage-related changes in proteasome activity has beenfound. A detailed analysis of each of the peptidase andproteolytic functions of the 20S proteasome along with itsintracellular levels in senescent cells has been performedby Shibatani and Ward (86). These authors, working withrat cytosolic extracts, found no differences with age in theintracellular levels or the total proteolytic activity of the20S proteasome (tested with casein, a well establishedsubstrate). Only after SDS stimulation was an increase inthe chymotrypsin and trypsin-like activities found in theaged hepatocytes. Under those conditions thepeptidylglutamyl peptide-hydrolyzing activity of the 20Sproteasome was clearly decreased, but that has beenshown to be of minor importance compared with the otherproteasome activities, at least in yeast (87, 88). Therefore,Shibatini and Ward (86) concluded that the reportedchanges in 20S proteasome activity with age are not

sufficient to account for the decreased proteindegradation in old cells.

Similar results were obtained by Sahakian etal. (89) working with purified rat liver 20S proteasomeinstead of cellular extracts. In addition, the decrease innon-acidic protease activity with age, first described inrat heart and liver, seems to be restricted to someorganisms and tissues since recent studies have shownthat this activity is not modified in houseflies or in ratbrain with age (90).

In the absence of changes in the proteasomeactivity, a reduction in protein ubiquitination could alsoresult in reduced intracellular protein breakdown.Several changes in intracellular protein ubiquitinationwith age have been reported. Studies in senescenthuman fibroblasts revealed that aged cells have less freeubiquitin and more ubiquitin-protein conjugates thanyoung cells (91). Similar changes have also beenobserved in lens (92) and in brain (93). However, levelsof ubiquitin mRNA do not change with age, and there isno difference in the ability of senescent fibroblasts todegrade ubiquitinated proteins (91). The fact thatchanges in protein ubiquitination do not always indicatechanges in protein degradation rates may reflect theparticipation of ubiquitin in intracellular processes otherthan protein degradation (94-96). Although these resultsindicate that a reduction in the ubiquitin-proteasomepathway is not a common finding in aging, thisproteolytic pathway may decline in certain tissues underparticular conditions. For example, as described in detailbelow, under oxidizing conditions, a failure in theubiquitination process in senescent eye lenses seems tocontribute to the accumulation of abnormal proteins(97).

4.1.2. Calpains Calpains, or calcium-dependent proteases,constitute the other major cytosolic proteolytic system(reviewed in ref. 98). The activity of these neutral, thiolproteases is tightly regulated by intracellular calciumlevels. Micromolar calcium concentrations activatemicro-calpain, but millimolar calcium concentrationsare required for milli-calpain activation (99). Where inthe cell millimolar calcium concentrations may beachieved remains an unsolved problem. Translocation ofcalpains to the cell membrane and their limited autolysisonce there result in calpain activation (100, 101) (figure2). Calpains partially degrade membrane andcytoskeletal proteins and several membrane-associatedenzymes (102-104). The limited proteolysis of certainmembrane proteins by calpains has been shown to benecessary for the process of membrane fusion (105).Two of the best characterized substrates for the calpainsystem are the band 3 protein (an anion exchanger) inthe erythrocyte membrane (99) and protein kinase C(106).

An increase in proteolysis of band 3 protein bycalpains in erythrocytes of old individuals has beendescribed (107). This increased degradation of band 3

Page 5: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

29

Figure 2. Calcium-dependent proteases. Changes inintracellular calcium levels modulate the activity of twocytosolic thiol proteases (calpains). Those proteases areusually present in an inactive form associated with calpastatin,an intracellular inhibitor. After translocation to the cellmembrane and limited autolysis, calpains are activated.Calpains are responsible for the degradation of certainmembrane and cytoskeletal proteins. See the text for details.

Fig. 3. Lysosomal pathways of protein degradation. Intra- andextracellular proteins can reach the lysosomal matrix bydifferent mechanisms: vesicular fusion (endocytosis (1) andcrinophagy (2)), macroautophagy (3), microautophagy (4),and direct transport through the lysosomal membrane (5). Seethe text for details.

protein correlates with a higher translocation of calpainsfrom the cytosol to the cell membrane. A diminution ofthiols in senescent cells, as a consequence of free radical

reactions and oxidative damage (108), may cause enhancedcalpain translocation to the cell membrane (100). Theincrease in degradation of band 3, and probably othermembrane proteins, modifies the stability of the erythrocytemembrane and reduces erythrocyte life span in agedanimals and humans (109). It is likely that this increase indegradation of membrane proteins described inerythrocytes is also common for most cell types. Forexample, as described below, the abnormal proteolysis andneuronal degeneration in Alzheimer's disease may be, inpart, a consequence of calpain activation (110).

The age-related changes in the calpain systemseem not to be related with changes in the proteolyticactivity of calpains themselves, because when calpainsfrom young individuals were added to membranes from olderythrocytes an increase in membrane protein degradationwas also observed (109). Those authors suggest theexistence of specific changes in cellular membrane proteinswith age which result in an increased affinity of thosemembranes for calpains thereby bringing the protease closeto its potential substrate proteins. Age-related changes inthe substrate susceptibility to proteases should also beconsidered. Several protein modifications make proteinsmore susceptible to proteolytic attack. Those susceptibilitychanges have been extensively studied in the brain, and wewill review them in more detail in the section of proteindegradation in the central nervous system. Finally, possiblemodifications in the activity of calpastatin, thephysiological inhibitor of calpains, with age need also to beconsidered (101).

4.2. Lysosomal function in senescent cellsLysosomes are organelles that contain a powerful

mixture of proteases, peptidases, and other hydrolasescapable of degrading most intracellular and extracellularmacromolecules (reviewed in ref. 111). The proteasesresponsible for the degradation of proteins insidelysosomes are called cathepsins (reviewed in ref. 112). Ingeneral, lysosomes have been considered to be responsiblefor the continuous basal turnover of most intracellularproteins (mainly long-lived proteins) in liver, kidney andcertain other tissues.

Several types of evidence have identifiedlysosomes as one of the major proteolytic systems affectedby age: (i) predominantly, degradation of long-livedproteins is retarded in senescent fibroblasts (13, 53) and inlivers of aged rats (113); (ii) in most cells there is an age-related increase in number and size of lysosomes (114);(iii) storage bodies similar to those described in senescentcells become evident in young cells after inhibitinglysosomal proteolysis with cathepsin inhibitors (115).

When considering the age-related reduction inlysosomal function, it is necessary to separately analyze thedifferent mechanisms by which proteins can be transportedinto lysosomes. The best characterized of thesemechanisms are: vesicular fusion (endocytosis andcrinophagy), macroautophagy, microautophagy and thedirect protein transport through the lysosomal membrane(reviewed in ref. 23) (figure 3).

Page 6: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

30

4.2.1. Fusion of lysosomes with vesicularcompartments

Primary lysosomes are able to completelydegrade the protein content of several kinds ofintracellular vesicles after membrane fusion. Vesiclescontaining secretory proteins originate at the Golgiapparatus, and they normally release their proteincontent to the extracellular medium after fusion of thevesicles with the plasma membrane. Under specificconditions, when a decrease in protein secretion isrequired, 20-70% of those vesicles directly fuse withlysosomes in a process called crinophagy, after whichsecretory proteins are completely degraded (116, 117).In spite of the several methods developed to analyze thisvesicular traffic (reviewed in ref. 118), the intrinsicmechanisms regulating this fusion process are stillunclear.

Fusion of lysosomes with endosomes, vesiclescontaining several extracellular and plasma membraneproteins, is fairly well-characterized (for review see ref.119). Internalization of several membrane receptorsafter ligand binding takes place by receptor-mediatedendocytosis into clathrin-coated vesicles and subsequenttraffic of those vesicles through theendosomal/lysosomal pathway. In that processmembrane receptors may be recycled to the cell surfacewhile the ligand is completely degraded after fusion oflate endosomes with lysosomes. In other cases thereceptor is not recycled and is delivered to lysosomesfor degradation along with the ligand. Endocytosis isalso required for uptake of extracellular nutrients,maintenance of cell polarity, and presentation ofantigenic peptides. Extensive reviews of the movementof membrane receptors through different intracellularcompartments and the signals that trigger receptorinternalization and receptor recycling have recently beenpublished (119, 120).

A decrease in levels of some serum proteins(121) and plasma membrane proteins (122) with age hasbeen described. Those preliminary reports suggestedsome alterations in the endocytic system responsible forthe internalization and degradation of those proteins.However, more direct studies of the endocytic system insenescent cells suggest little change in this process withage. An increase in the internalization of latex beads insenescent fibroblasts has been found (123), and adecrease in the endocytic activity of aged fibroblastsand Kupffer cells has also been reported (124, 125). Incontrast, internalization and degradation of low densitylipoprotein and epidermal growth factor are unmodifiedin senescent fibroblasts (126, 127). These discrepantresults may be related to the different analyses of theexperimental data performed or in cell type analyzed ineach study. Thus, Gurley and Dice (128) showed thatendocytosis values should be expressed per microgramof cell protein instead of normalized per number ofcultured cells, since old cells are much larger thanyoung cells. Under those circumstances there was nodifference in endocytosis rates for young and oldfibroblasts. In addition, rates of fluid-phase and

absorptive endocytosis were not modified with age.Thus, there is not a general failure in lysosomal functionduring senescence.

4.2.2. Macroautophagy and microautophagyA large number of intracellular proteins can be

degraded in lysosomes by a process called macroautophagy(reviewed in ref. 129). In this process, complete regions ofthe cytoplasm, including cytosolic proteins as well as entireorganelles, are surrounded by a membrane to becomedouble-membraned structures known as autophagosomes orautophagic vacuoles (figure 3). The proteins insideautophagosomes are then completely degraded after fusionwith lysosomes. In organs such as the liver,macroautophagy accounts for most intracellular proteindegradation, especially during early starvation (130).Factors inhibiting this process include amino acids, insulin,cell swelling, and disruption of the cytoskeleton (131, 132).Among the mechanisms controlling macroautophagy,protein phosphorylation plays an important role (133).Although proteins in the sequestered cytoplasm appear tobe taken up nonselectively, under certain conditionsselectivity in this degradative process has beendemonstrated. For example, peroxisomal autophagy can bepreferentially activated under conditions whereperoxisomes are no longer needed (134, 135). In addition,selective degradation of specific cytosolic proteins in theyeast vacuole following a novel vesicular pathway has beenrecently described (136). The fusion of lysosomes with theautophagosome follows similar mechanisms as thosepreviously described for the endocytic and secretorypathways, and evidence of interaction between bothprocesses has been presented (137).

During senescence, severe changes in theautophagosome/lysosomal system have been described.Degradation of several proteins microinjected into thecytoplasm of senescent human fibroblasts is significantlylower than in young fibroblasts (20). For some of thoseproteins (RNase S protein, lysozyme), their degradation isbelieved to follow the autophagosome/lysosomal pathway.The half-lives of some of those microinjected proteins aresummarized in table 2. A decrease in autophagosomeformation as well as a decrease in the degradative activityof lysosomes would result in the slower degradation ofsubstrate proteins. Since autophagic vacuole formationrates decrease in senescent cells (138), and rates ofelimination of autophagic vacuoles also decrease with age(139), reductions in autophagosome formation and fusionwith lysosomes both contribute to the age-related decline inmacroautophagy.

A second process of intracellular proteinsequestration by lysosomes has been calledmicroautophagy. During microautophagy only smallportions of cytoplasm are engulfed by small invaginationsin the surface of lysosomes. After internalization of thosevesicles and breakage of their surrounding membrane, thecytoplasmic proteins are degraded inside lysosomes (140).In contrast with macroautophagy that is mainly activatedunder conditions of nutrient deprivation, microautophagy isresponsible for the continuous basal degradation of long-

Page 7: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

31

Table 2. Age-related changes in the half-lives of severalcytosolic proteins

T½ (h)Protein Young Old Model System Reference

Aspartateaminotransferase

55 90 Human fibroblast 152

Lysozyme 28 80 Human fibroblast 20

Aldolase 175 520 Human fibroblast 20RNase S-protein 88 120 Human fibroblast 20RNase A 100 185 Human fibroblast 20Poly-Glu:Tyr:Ala 110 375 Human fibroblast 20

Ornithinedecarboxylase

0.15 0.5 Mouse Liver 152

Aldolase 26 37 Mouse Liver 152

Aldolase 35 170 Nematode 152Enolase 58 161 Nematode 152Triosephosphateisomerase

64 120 Nematode 152

Ovalbumin 106 164 Rat hepatocyte 195Horseradishperoxidase

50 75 Rat hepatocyte 195

Figure 4. The hsc73-mediated pathway of cytosolic proteindegradation. The main components of this system of directtransport of proteins through the lysosomal membrane are: acytosolic chaperone of 73 kDa (hsc73), a receptor protein atthe lysosomal membrane (lgp96) and an intralysosomal formof the cytosolic chaperone (lys-hsc73). The substrate proteinafter interaction with hsc73 (1) is directed to lysosomes whereafter binding (2) and transport through the lysosomalmembrane (3), it is completely degraded in the lysosomalmatrix (4).

lived proteins (141). No information is availableconcerning rates of microautophagy in aging.

4.2.3. Direct transport of cytosolic proteins through thelysosomal membrane.

A third mechanism of lysosomal degradation ofcertain cytosolic proteins occurs after direct transportthrough the lysosomal membrane (reviewed in ref. 23).This protein transport resembles in many aspects thetransport of proteins into other cellular compartments

(142). At least two molecular chaperones and a receptorprotein, elements also described for other transportsystems, are also present in this lysosomal pathway. Thesequence of events that directs specific cytosoliclysosomes to their lysosomal degradation by this pathwayis summarized in figure 4. The substrate proteins containin their sequence a consensus peptide motif biochemicallyrelated to the pentapeptide, KFERQ (143, 144), that isrecognized by a cytosolic chaperone of 73 kDa (hsc73)(143, 145). The binding of hsc73 to that region of thesubstrate protein directs the complex toward thelysosomal membrane (59). A lysosomal membraneglycoprotein of 96 kDa (lgp96 in rats or lamp2 in humansand mice) acts as a receptor for the substrate protein(146). Assisted by a form of the cytosolic hsc73 in thelysosomal matrix (147, 148), the substrate protein reachesthe lysosomal matrix where it is completely degraded.Binding to the lysosomal membrane and uptake into thelysosomal matrix are the rate-limiting steps in this processand are directly related with lysosomal levels of lgp96and hsc73, respectively (146, 148).

This hsc73-mediated lysosomal pathway wasfirst described in human fibroblasts in culture (51, 149,150) and then characterized in rat liver (60, 151). Thedirect transport of proteins into lysosomes is activated byserum deprivation of confluent cultured cells (152) orprolonged starvation of intact animals (153, 61).Approximately 30% of the cytosolic proteins aredegraded by this pathway (153). Several of the substratesalready identified for this pathway are: ribonuclease A(150), glyceraldehyde-3-phosphate dehydrogenase (60),aldolase (60), some subunits of the 20S proteasome (154),transcription factors such as c-fos (155) and IkB (Cuervo,A.M., Hing, M., Lim, B. and Dice, J.F., unpublishedresults), and alpha-2-microglobulin, a secretory proteinthat is also located in the cytosol of hepatocytes andkidney proximal tubular cells (Cuervo, A.M., Hildebrand,H., Bomhard, H. and Dice, J.F., unpublished results).

Analysis of the degradation of proteinsmicroinjected into senescent fibroblasts revealed adecrease in the degradation of ribonuclease A, one ofthe described substrates for the direct lysosomalpathway (152) (figure 5A). In addition, differencesbetween young and old cells in total protein degradationrates were especially evident in confluent cells afterserum withdrawal, a condition under which the hsc73-mediated transport of proteins into lysosomes isactivated.

Working with lysosomes isolated from old ratswe have also observed a decrease in their ability todirectly take up substrate proteins for this pathway(Cuervo and Dice, unpublished results) (figure 5B). Allthose results suggest that the direct transport ofcytosolic proteins through the lysosomal membrane isimpaired in old cells, and this reduction is at leastpartially responsible for the age-related decrease in totalprotein degradation. The components of the hsc73-mediated pathway that are affected by age are currentlybeing studied in our laboratory.

Page 8: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

32

Table 3. Changes in several protein degradation pathways in specific cell types with ageLYSOSOMAL

________________________________________________Tissue Ubiquitin/ Proteasome Calpains Macroautophagy Microautophagy Hsc73-mediated

Central nervous systemNo change

IncreaseDecrease No data No data No data

Skeletal muscle No change No change No data No data No change

Liver No change No change Decrease No data DecreaseEye lens Decrease No change No data No data Decrease

Figure 5. Evidence for reduction of the hsc73-mediatedpathway with age. Decrease in the degradation ofmicroinjected RNase A in old fibroblast when compared withyoung fibroblasts, especially after serum deprivation (A), anda reduced ability for the direct uptake of this protein byisolated liver lysosomes from old rats (B) strongly support areduction of the hsc73-mediated pathway of proteindegradation with age.

In conclusion, the analysis of the effects ofsenescence on the lysosomal function reveals that, ratherthan a generalized failure in the lysosomal system with age,specific degradative mechanisms are impaired in senescent

cells. The important role of lysosomes in bulk proteinturnover in liver, kidney, brain and fibroblasts make themmainly responsible for the age-related decrease in proteindegradation in those organs.

5. TISSUE-SPECIFIC CHANGES IN PROTEOLYTICSYSTEMS WITH AGE

We discuss here four representative examples ofchanges in protein turnover that are organ-dependent. The age-related modifications in the activity of several proteindegradation pathways in those tissues are summarized in table 3.

5.1. Central nervous systemThe central nervous system is one of the tissues

that shows the most dramatic changes in proteindegradation with age. Age-related morphological andneurochemical changes in brain cells have been very well-characterized, and many of them can be reproduced afterexposure to different protease inhibitors (156, 157).Consequently, reduced protease activities or enhancedprotease inhibitor activities might be involved in brainaging.

Several age-related changes in the activity of brainproteases have been reported (see table 4). In addition tochanges in levels and activity of brain proteases with age,alterations in their substrate sensitivity to proteolytic attackneed to be considered. For example, there are many reports ofincreased levels and activities of cathepsin D and calpain in theaged brain, but such changes do not correlate with an increasedprotein degradation, since proteolysis declines with age (169).In the same way, the accumulation of lipofuscin-pigment inlysosomes is not due to a decrease in cathepsin B activity withage (170). These discrepancies and others summarized in table4 may be due to the different substrate proteins used (reviewedin ref. 169).

The lysosomal/endosomal and calpain systemsseem to be the most affected by age in neuronal tissues.Thus, as described above, lysosomes undergo markedmorphological changes in senescent cells and participate inneurodegenerative processes. As a result of aging ormetabolic and oxidative stress, indigestible or incompletelydegraded material accumulates within the endosomal-lysosomal system and produces a heterogeneous group ofresidual bodies containing lipofuscin (171-173). Althoughthose aggregates can also be caused by treatment withlysosomotropic inhibitors (174), their accumulation in

Page 9: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

33

Table 4. Age-related changes in protein degradation in the nervous systemCHANGES IN PROTEASE ACTIVITY REFERENCEq Increase of calpain activity in spinal cord 158q Increase in calpastatin levels in brain 159q Increase in calpain and cathepsin D activity in brain 160q Decrease in calpain levels but increase in calpain activity in Alzheimer’s disease patients 110,161q Increase in levels of cathepsin D in brain 162q Increase of brain calpain activity depends on the cerebral area analyzed 163q Increase in cathepsin D, E, and B activities and decrease in cathepsin L activity in brain 162

CHANGES IN SUBSTRATE SUSCEPTIBILITY TO PROTEASESq Microtubule-associated proteins in brain are more susceptible to cathepsin D-like proteases 164

q Phosphorylation and carbonyl modification of neurofilament proteins reduce their proteolytic susceptibility 161, 165, 166q Age-related modifications of synaptosomal membrane proteins inhibits their proteolysis 167q Binding of aluminun to neurofilament proteins inhibits their proteolysis 161q Peroxidation of myelin membrane proteins increases their susceptibility to proteolysis 168

aging does not appear to be due to reduced protease activitybut to changes in proteolytic susceptibility of the substrateproteins. An increased expression of some lysosomalproteases (cathepsin D, B and E) (175) together with adecreased stability of the lysosomal membrane (176) havealso been described in senescent brain.

In Alzheimer's disease, one of the most commonneurodegenerative diseases, proteins of the cell membraneand cytoskeleton are abnormally processed and accumulatein the brain. It is now believed that lysosomal alterationsare important in the pathogenesis of this disease and in theaccumulation of amyloidogenic peptides (177-179). Acidhydrolases accumulate in atrophic and degeneratingneurons, and their release to the extracellular space maycontribute to senile plaque formation (179). The typicalamyloid deposits in Alzheimer's disease are normallyproduced in an acidic compartment by noncysteineproteases, and then they are eliminated by lysosomalcysteine proteases (mainly cathepsins B and L) (177, 180-182). Treatment with general cysteine proteinase inhibitorscause accumulation of potentially amyloidogenic precursorprotein fragments (183, 184).

5.2. Liver and cultured fibroblastsIn spite of the many differences between

hepatocytes and fibroblasts, the parallelism of most of theresults regarding age-related changes in intracellularprotein degradation lead us to consider them together inthis section. Many of the age-related changes in thedegradative systems described in section 4.2. were obtainedusing rat liver or human fibroblasts in culture, and theseresults are summarized in table 5.

The liver is one of the organs in which a decreasein total protein degradation with age has been well-established (113). The most affected proteolytic system inliver from old animals is the lysosomal system. Lysosomesare responsible for more than 80 % of protein degradationin liver (196, 197). Characteristic age-related changes in themorphology and function of the macroautophagy/lysosomalsystem have been described in hepatic cells: there is anincrease in number and size of lysosomes in liver cells with

age (192); the activity of lysosomal enzymes increases withage in parenchymal and non-parenchymal cells of the liver(193); there is a decrease in the formation of autophagicvacuoles and in the degradation of their content (141); thehalf-lives of proteins microinjected in hepatic cells increasewith the age of the donor animal (table 2; 195); and theselective uptake of cytosolic proteins by lysosomes from ratliver decreases with age (figure 5B; Cuervo and Dice,unpublished results). This decrease in the activity of thehsc73-mediated lysosomal pathway in rat liver with age isconsidered to be one of the major proteolytic pathwaysresponsible for the decrease in total protein breakdown inliver.

As described previously, human fibroblasts inculture have been extensively used for studies of aging (seesection 3). In spite of the controversy in acceptingsenescence in culture as a good model of aging in vivo,fibroblasts in culture show very similar alterations inprotein degradation pathways as does liver from aged rats.table 5 summarizes these changes in proteolytic pathways.

5.3. Skeletal muscleIn contrast to the nervous system, liver, and

fibroblasts, where lysosomes appear to play a key role inalterations of protein degradation associated with aging,the ubiquitin-dependent system is mainly responsible forprotein degradation in skeletal muscle, especially in manymuscle-wasting conditions (198, 199). During aging thereis a progressive loss of muscle mass (200) that originatesbecause of an imbalance between synthesis anddegradation of proteins (reviewed in ref. 201). However,when basal rates of skeletal muscle protein turnover areanalyzed, there is a significant reduction with age in bothsynthesis and degradation. Studies analyzing rates ofprotein synthesis by [14C]tyrosine incorporation intomuscle, and degradation by urinary excretion of 3-methylhistidine, related to myofibrillar proteinbreakdown, have demonstrated that both processessignificantly decrease in skeletal muscle of rats with age(202). The protein synthesis/degradation imbalance insenescence is only evident after induction of a catabolicstate such as in starvation, denervation atrophy,

Page 10: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

34

Table 5. Comparison of the age-related changes in protein degradation in rat liver and cultured human fibroblastsPROTEIN DEGRADATION LIVER FIBROBLASTS

CHANGE REFERENCE CHANGE REFERENCE

q Total protein degradation Decrease 113 Decrease 185

q Proteolytic susceptibility Increase 186 Increase 187q Oxidized and modified proteins Increase 188 Increase 189

PROTEOLYTIC SYSTEMSCytosolicq Proteasome

• Proteasome levels No change 86 No change 190

• Degradation ofubiquitinated proteins

No change 191 No change 91

q Calpain activity No data Increase 110

Lysosomalq Elimination of autophagicq vacuoles

Decrease 141 Decrease 140

q Half-life of microinjectedq proteins

Increase 195 Increase 20

q Hsc73-mediated Decrease Cuervo & Dice,unpublished

Decrease 20

q Number and sizes ofq lysosomes

Increase 192 Increase 114

q Activity of cathepsins Increase 193 Increase 194

cancer, acidosis or trauma, and reflect a failure to rapidlyrestore muscle protein after those circumstances (203).Those authors also demonstrated that muscle wastingresulted from increased proteolysis in normal rats, but it iscaused by a decrease in protein synthesis in senescent rats.Under catabolic conditions only a small increase ofcathepsin D and milli-calpain have been observed, but itdoes not modify total protein breakdown (204).

5.4. Eye lensAnother tissue where age-related changes in

the activity of proteolytic systems has been studied isthe eye lens. In the lens the proteolytic removal ofdamaged proteins may play an important role inmaintaining the lens transparency (90, 205-207).Normal lenses have the ability to increase ubiquitinconjugation activity in response to oxidative stress, butthat response is impaired in senescent lenses (92, 97).No changes in levels of ubiquitin conjugating enzymesin lenses were detected with age, but changes in theirability to respond to oxidative stress were found (97).The age-related decrease in the ability to mount aubiquitin-dependent response upon oxidation maycontribute to the accumulation of damaged proteins inthe old lenses.

Other proteolytic systems also decline withage in lens. Studies in cultured epithelial lens cellsrevealed that, after successive passages, there was adecrease in the proteolytic response to serum removal.This degradative defect mainly affects proteolysis oflong-lived proteins suggesting that the degradativedefect may be related with the hsc73-mediatedlysosomal pathway of protein degradation (208).

6. CONCLUSIONS AND PERSPECTIVES

Many of the changes that happen in senescentcells can be explained based on reduced rates ofintracellular protein degradation. The decrease in totalprotein breakdown with age has been extensively reported,and the specific intracellular proteolytic systemsresponsible for this decrease are now becoming clear.Dietary restriction extends the lifespan of rats, and thistreatment also delays the age-related decline in liver proteindegradation (195), consistent with reduced proteindegradation being an important parameter in aging.

We have presented a general review of age-related changes in different proteolytic systems. However,possible modifications in the proteolytic susceptibility ofsubstrate proteins with age should also be considered,because both a decrease in the activity of proteolyticpathways and also a decrease in the proteolyticsusceptibility of substrate proteins could result inaccumulation of proteins in senescent cells. For example,certain oxidized proteins and cross-linked proteins are poorsubstrates for proteases and are slowly degraded by cells(188). This stabilization may contribute to the observedaccumulation and damaging actions of oxidized proteins

during aging (188). However, these reductions inproteolytic susceptibility with age do not seem to apply tothe majority of protein modifications (for review see ref.90). In fact, there is an increase in susceptibility todegradation by exogenous proteases of cytosolic proteinsfrom liver of old rats (186), and from senescent fibroblasts(187), and it has been proposed that different age-specific

Page 11: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

35

protein modifications must contribute to the increasedsusceptibility to proteolytic attack (186).

There are still many aspects of intracellularprotein degradation that need to be addressed in order toidentify the primary defects with age. The discovery ofproteolytic activities in some cellular compartments untilrecently unknown (i.e. endosomes and mitochondria) hasalready expanded the roles of the protein breakdown insidecells. Identification of other new proteases might alsoexplain the decrease in non-acidic protease activity in somemodels of senescence.

As described above, one of the principalpathways contributing to the age-related degradation failureis the hsc73-mediated lysosomal pathway. Severalmechanistic aspects of this specific lysosomal pathway areunclear; the exact mechanism underlying cytosolic hsc73stimulation of substrate binding to the lysosomal membranereceptor; the role of other cytosolic regulators of the hsc73function; the existence of other lysosomal membrane andmatrix protein(s) that participate in the transit of substratesinto the lysosomal matrix. New substrates for this pathwayneed to be identified, and the contribution of thisproteolytic system to the overall intracellular degradation indifferent organs and under different conditions must befurther established.

The identification of proteolytic defects with agewill also allow the development of different correctivemethods to eliminate the cytosolic accumulation ofabnormal proteins. One approach is to reduce proteindamage (209). However, the use of protective enzymes islimited because cells are exposed to a large number ofpotential injurious agents. A more general solution for thisproblem will be to facilitate the elimination of theaccumulated proteins.

We have previously demonstrated that theoverexpression of the receptor protein for the hsc73-mediated lysosomal pathway in normal cultured cellsresults in an increase in the activity of this pathway (146).Regulated gene expression systems, such as the Tet-Offand Tet-On system regulated by tetracycline, are nowavailable. Those systems allow a high-level expression ofcloned genes in response to varying concentrations oftetracycline (210). The regulated expression of lgp96 infibroblasts may allow us to increase the activity of thehsc73-mediated lysosomal pathway at different times ofsenescence. Thus, we could determined the reversibilityof the age-related decrease in the activity of the selectivelysosomal pathway, along with the effect of its activationin the elimination of the protein deposits in the cell. Thatprocedure may result in a recovery of other normalcellular functions.

7. ACKNOWLEDGMENTS

We thank Elizabeth Frutiger for the critical readingof this manuscript. Work in the authors' laboratory wassupported by NIH grant DK07542 (AMC) and NIH grantAG06116 (JFD).

8. REFERENCES

1. DA Jurivich, L Qiu & JF Welk: Attenuated stressresponses in young and old human lymphocytes. MechAgeing Develop 94, 233-249, (1997)

2. G Pawelec & R Solana: Immunosenescence. TrendsImmunol 18, 514-516, (1997)

3. R Yanishevsky, ML Mendelsohn, BH Mayall & VJCristofalo: Proliferative capacity and DNA content of aginghuman diploid cells in culture: A cytophotometric andautoradiographic analysis. J Cell Physiol 84, 165-170,(1974)

4. T Seshadri & J Campisi: Repression of c-fostranscription and an altered genetic program in senescenthuman fibroblasts. Science 247, 205-209, (1990)

5. GP Dimri & J Campisi: Altered profile of transcriptionfactor-binding activities in senescent human fibroblasts.Exp Cell Res 212, 132-140, (1994)

6. CA Afshari & JC Barrett: Molecular genetics of in vitrocellular senescence. In: Cellular Aging and Cell Death.Eds: Holbrook NJ, Martin GR, Lockshin, RA.Wiley-Liss,Inc. NY. 109-121, (1996)

7. Y Ning, JL Weber, AM Killary, DH Ledbetter, JR Smith& OM Pereira-Smith: Genetic Analysis of indefinitedivision in human cells: Evidence for a senescence-relatedgene(s) on human chromosome 4. Proc Natl Acad SciUSA 88, 5635-5639, (1991)

8. CF van Bezooijen, FR Leeuw-Israel & CF Hollander: Onthe role of hepatic cell ploidy in changes in liver functionwith age and following partial hepatectomy. Mech AgeingDevelop 1, 351-356, (1973)

9. R Schoenheimer: Dynamic State of Body Constituents.Harvard University Press, Cambridge, (1942)

10. RT Schimke: Regulation of protein degradation inmammalian tissues. In: Mammalian Protein Metabolism.Ed: Munro, HN. Academic Press, NY, 177-277, (1970)

11. AL Goldberg & JF Dice: Intracellular proteindegradation in mammalian and bacterial cells. Annu RevBiochem 34, 835-869, (1974)

12. FJ Doherthy & RJ Mayer: Intracellular ProteinDegradation. Oxford University Press, New York, (1992)

13. JF Dice: Cellular and molecular mechanisms ofaging. Physiol Rev 73, 149-159, (1993)

14. J Papaconstantinou: Unifying model of theprogrammed (intrinsic) and stochastic (extrinsic)theories of aging. The stress response genes, signaltransduction-redox pathways and aging. Ann New YorkAcad Sci 719, 195-211, (1994)

Page 12: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

36

15. JA Knight: The process and theories of aging. AnnClinic Lab Sci 25, 1-12, (1995)

16. J Vijg & JY Wei: Understanding the biology of aging:the key to prevention and therapy. J Am Geriat Soc 43,426-434, (1995)

17. S Makrides: Protein synthesis and degradation duringaging and senescence. Biol Rev 83, 393-422, (1983)

18. DF Goldspink & FJ Kelly: Protein turnover and growthin the whole body, liver, and kidney of the rat from foetusto senility. Biochem J 217, 507-516, (1984)

19. JF Dice: Altered intracellular protein degradation inaging: A possible cause of proliferative arrest. ExpGerontol 24, 451-459, (1989)

20. JF Dice: Altered degradation of proteins microinjectedinto senescent human fibroblasts. J Biol Chem 257, 14624-14627, (1982)

21. RW Gracy, KU Yuksel, ML Chapman, JK Cini, MJahani, HS Lu, B Gray & JM Talent: Impaired proteindegradation may account for the accumulation of abnormalproteins in aging cells. In: Modifications of Proteins duringAging. Eds: Adelman RC, Dekker, EE. Alan R. Liss, NY,1-18, (1985)

22. A Taylor & KJ Davies: Protein oxidation and loss ofprotease activity may cause cataract formation in the agedlens. Free Radic Biol Med 3, 371-377, (1987)

23. AM Cuervo, SA Hayes & JF Dice: Molecularchaperones and intracellular protein degradation withemphasis on a selective lysosomal pathway of proteolysis.In: Molecular Chaperones in the Life Cycle of Proteins.Eds: Fink, AL and Goto, Y. Marcel Dekker, Inc. NY, 491-510, (1997)

24. I Jariel-Encontre, C Salvat, AM Steff, M Pariat, CAcquaviva, O Furstoss & M Piechaczyk: Complexmechanisms for c-fos and c-jun degradation. Mol Biol Rep24, 51-56, (1997)

25. F Chen, Y Lu, DC Kuhn, M Maki, X Shi, SC Sun &LM Demers: Calpain contributes to silica-induced I kappaB-alpha degradation and nuclear factor-kappa B activation.Arch Biochem Biophys 342, 383-388, (1997)

26. TJ Jensen, MA Loo, S Pind, DB Williams, ALGoldberg & JR Riordan: Multiple proteolytic systems,including the proteasome, contribute to CFTR processing.Cell 83, 129-135, (1995)

27. AL Schwartz, JS Trausch, A Ciechanover, JW Slot & HGeuze: Immunoelectron microscopic localization of theubiquitin-activating enzyme E1 in HepG2 cells. Proc NatlAcad Sci USA 89, 5542-5546, (1992)

28. P Low, FJ Doherty, E Fellinger, M Sass, RJ Mayer & LLaszlo: Related organelles of the endosome-lysosome

system contain a different repertoire of ubiquitinatedproteins in sf9 insect cells. FEBS Lett 368, 125-131, (1995)

29. RJ Mayer, C Tipler, J Arnold, L Laszlo, A Al-Khedhairy, J Lowe & M Landon: Endosome-lysosomes,ubiquitin and neurodegeneration. Adv Exp Med Biol389:261-269, (1996)

30. EF Blommaart, JJ Luiken & AJ Meijer: Autophagicproteolysis: control and specificity. Histochem J 29, 365-385, (1997)

31. JD Burek: Pathology of Aging Rats. CRC Press, Inc.,Boca Raton, FL (1978)

32. BJ Cohen, MR Anver, DH Ringler & RC Adelman:Age-associated pathological changes in male rats. FedProc37, 2848-2850, (1978)

33. RL Sprott: Mouse and rat genotype choices. ExpGerontol. 32, 79-86, (1997)

34. T Takeda, M Hosokawa, S Takeshita, M Irino, KHiguchi, T Matsushita, Y Tomita, K Yasuhira, HHamamoto, K Shimizu, M Ishii & T Yamamuro: A newmurine model of accelerated senescence. Mech AgeingDevelop 17,183-194, (1981)

35. B Sass, LS Rabstein, R Madison, RM Nims, RLPeters & GJ Kelloff: Incidence of spontaneous neoplasmsin F344 rats throughout the natural life span. J NatlCancer Inst 54, 1449-1456, (1975)

36. GL Coleman, W Barthold, GW Osbaldiston, SJ Foster& AM Jonas: Pathologic changes during aging in barrier-reared Fischer 344 male rats. J Gerontol 32, 258-278,(1977)

37. JM Ward & H Reznik-Schuller: Morphological andhistochemical characteristics of pigments in aging F344rats. Vet Pathol 17, 678-685, (1980)

38. JB Nold & GA Parker: Diseases and Neoplasms ofthe Aging Rat. 42nd Annual Pathology of LaboratoryAnimals Course, (1995). http://vetpath1.afip.mil/POLA95

39. HJ Martus, ME Dolle, JA Gossen, ME Boerrigter &J Vijg: Use of transgenic mouse models for studyingsomatic mutations in aging. Mut Res 338, 203-213,(1995)

40. M Kuro, Y Matsumura, H Aizawa, h Kawaguchi, TSuga, T Utsug, Y Ohyama, M Kurabayashi, T Kaname,E Kume, H Iwasaki, A Iida, T Shiraki-Iida, SNishikawa, R Nagai & YI Nabeshima: Mutation of themouse klotho gene leads to a syndrome resemblingageing. Nature 390, 45-51, (1997)

41. CE Finch: Longevity, Senescence and the Genome.The University of Chicago Press, Chicago, IL.(1990)

Page 13: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

37

42. VJ Cristofalo & RJ Pignolo: Molecular markers ofsenescence in fibroblast-like cultures. Exp Gerontol 31,111-123, (1996)

43. L Hayflick & PS Moorhead: The serial cultivation ofhuman diploid cell strains. Exp Cell Res 25, 585-621,(1961)

44. L Hayflick: The cell biology of human aging. New EnglJ Med 295, 1302-1308, (1976)

45. EL Schneider & Y Mitsui: The relationship between invitro cellular aging and in vivo human age. Proc Natl AcadSci USA 73, 3584-3588, (1976)

46. S Goldstein: Aging in vitro. Growth of cultured cellsfrom the Galapagos turtle. Exp Cell Res 83, 297-302,(1974)

47. S Goldstein, S Murano, H Benes, EJ Moerman, RAJones, R Thweatt, RJ Shmookler- Reis & BH Howard:Studies on the molecular genetic basis of replicativesenescence in Werner syndrome and normal fibroblasts.Exp Gerontol 24, 461-468, (1989)

48. VJ Cristofalo & RJ Pignolo: Replicative senescence ofhuman fibroblast-like cells in culture. Physiol Rev 73, 617-638, (1993)

49. GP Dimri, X Lee, G Basile, M Acosta, G Scott, CRoskelley, EE Medrano, M Linskens, I Rubelj, O Pereira-Smith & M Peacocke: A biomarker that identifies senescenthuman cells in culture and in aging skin in vivo. Proc NatlAcad Sci USA 92, 9363-9367, (1995)

50. SM Russell, CJ Wilde, DA White, HR Hasan & RJMayer: Characteristics of protein degradation in liver andmammary gland. Acta Biologic Medic Germ 40, 1397-1406, (1981)

51. MA McElligott & JF Dice: Erythrocyte-mediatedmicroinjection, a technique to study protein degradation inmuscle cells. Biochem J 216, 559-566, (1983)

52. PO Hasselgren, P Pedersen, HC Sax, BW Warner & JEFischer: Methods for studying protein synthesis anddegradation in liver and skeletal muscle. J Surg Res 45,389-415,

53. A Okada & JF Dice: Altered degradation ofintracellular proteins in aging human fibroblasts. MechAgeing Dev 26, 341-356, (1984)

54. T Tamura, K Tanaka, N Tanahashi & A Ichihara:Improved method for preparation of ubiquitin-ligatedlysozyme as substrate of ATP-dependent proteolysis.FEBS Lett 292, 154-158, (1991)

55. SK Kong & PB Chock: Protein ubiquitination isregulated by phosphorylation. An in vitro study. J BiolChem 267, 14189-14192, (1992)

56. S Ugai, T Tamura, N Tanahashi, S Takai, N Komi,CH Chung, K Tanaka & A Ichihara: Purification andcharacterization of the 26S proteasome complexcatalyzing ATP-dependent breakdown of ubiquitin-ligatedproteins from rat liver. J Biochem 113, 754-768, (1993)

57. T Shibatani & WF Ward: Sodium dodecyl sulfate(SDS) activation of the 20S proteasome in rat liver. ArchBiochem & Biophys 321, 160-166, (1995)

58. Y Goda & SR Pfeffer: Cell-free systems to studyvesicular transport along the secretory and endocyticpathways. FASEB J 3, 2488-2495, (1989)

59. SR Terlecky & JF Dice: Polypeptide import anddegradation by isolated lysosomes. J Biol Chem 268,23490-23495, (1993)

60. F Aniento, E Roche, AM Cuervo & E Knecht: Uptakeand degradation of glyceraldehyde-3-phosphatedehydrogenase by rat liver lysosomes. J Biol Chem 268,10463-10470, (1993)

61. AM Cuervo, E Knecht, SR Terlecky & JF Dice:Activation of a selective pathway of lysosomal proteolysisin rat liver by prolonged starvation Am J Physiol 269,C1200-C1208, (1995)

62. JM Peters, Z Cejka, JR Harris, JA Kleinschmidt & WBaumeister: Structural features of the 26 S proteasomecomplex. J Mol Biol 234, 932-937, (1993)

63. A Lupas, HM Flanagan, T Tamura & W Baumeister:Self-compartmentalizing proteases. Trends Biochem Sci 22,399-404, (1997)

64. O Coux, K Tanaka & AL Goldberg: Structure andfunctions of the 20S and 26S proteaseomes. Annu RevBiochem 65, 801-847, (1996)

65. M Orlowski: The mutlticatalytic proteinase complex,a major extralysosomal proteolytic system. Biochemistry29, 10289-10297, (1990)

66. D Krappmann, FG Wulczyn & C Scheidereit:Different mechanisms control signal-induceddegradation and basal turnover of the NF-kappaBinhibitor IkappaB alpha in vivo. EMBO J 15, 6716-6726,(1996)

67. EJ Wiertz, D Tortorella, M Bogyo, J Yu, W Mothes,TR Jones, TA Rapoport & HL Ploegh: Sec61-mediatedtransfer of a membrane protein from the endoplasmicreticulum to the proteasome for destruction. Nature 384,432-438, (1996)

68. A Varshavsky: The ubiquitin system. TrendsBiochem Sci 22, 383-387, (1997)

69. M Hochstrasser: Ubiquitin-dependent proteindegradation Annu Rev Genet 30, 405-439, (1996)

Page 14: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

38

70. V Chau, JW Tobias, A Bachmair, D Marriott, DJ Ecker,DK Gonda & A Varshavsky: A multiubiquitin chain isconfined to a specific lysine in a targeted short-livedprotein. Science 243, 1576-1583, (1989)

71. Q Deveraux, V Ustrell, C Pickart & M Rechsteiner: A26 S protease subunit that binds ubiquitin conjugates. J BiolChem 269, 7059-7061, (1994)

72. S van Nocker, S Sadis, DM Rubin, M Glickman, H Fu,O Coux, I Wefes, D Finley & RD Vierstra: Themultiubiquitin-chain-binding protein Mcb1 is a componentof the 26S proteasome in Saccharomyces cerevisiae andplays a nonessential, substrate-specific role in proteinturnover. Mol Cell Biol 16, 6020-6028, (1996)

73. RT Baker, JW Tobias & A Varshavsky: Ubiquitin-specific proteases of Saccharomyces cerevisiae. Cloning ofUBP3, and functional analysis of the UBP gene family. JBiol Chem 267, 23364-23375, (1992)

74. KD Wilkinson, VL Tashayev, LB O'Connor, CNLarsen, E Kasperek & CM Pickart: Metabolism of thepolyubiquitin degradation signal: structure, mechanism,and role of isopeptidase T. Biochemistry 34, 14535-14546,(1995)

75. F Tokunaga, T Goto, T Koide, Y Murakami, S Hayashi,T Tamura, K Tanaka & A Ichihara: ATP-and antizyme-dependent endoproteolysis of ornithine decarboxylase tooligopeptides by the 26 S proteasome. J Biol Chem 269,17382-17385, (1994)

76. L Hicke & H Riezman: Ubiquitination of a yeastplasma membrane receptor signals its ligand-stimulatedendocytosis. Cell 84, 277-287, (1996)

77. J Horak & DH Wolf: Catabolite inactivation of thegalactose transporter in the yeast Saccharomycescerevisiae: ubiquitination, endocytosis, and degradation inthe vacuole. J Bacteriol 179, 1541-1549, (1997)

78. K Tanaka, K Ii, A Ichihara, L Waxman, & ALGoldberg: A high molecular weight protease in thecytosol of rat ) liver. I. Purification, enzymologicalproperties, and tissue distribution. J Biol Chem 261,15197-15203, (1986

79. CV Harding, J France, R Song, JM Farah, SChatterjee, M Iqbal & R Siman: Novel dipeptidealdehydes are proteasome inhibitors and block the MHC-Iantigen-processing pathway. J Immunol 155, 1767-1775,(1995)

80. DH Lee & AL Goldberg: Selective inhibitors of theproteasome-dependent and vacuolar pathway of proteindegradation in Saccharomyces cerevisiae. J Biol Chem271, 27280-27284, (1996)

81. L Ditzel, D Stock & J Lowe: Structural investigationof proteasome inhibition. Biol Chem 378, 239-247, (1997)

82. H Djaballah, AJ Rowe, SE Harding & AJ Rivett:The multicatalytic proteinase complex (proteasome):structure conformational changes associated withchanges in proteolytic activity. Biochem J 292, 857-862,(1993)

83. M Groettrup, A Soza, M Eggers, L Kuehn, TP Dick, HSchild, HG Rammensee, UH Koszinowski & PM Kloetzel:A role for the proteasome regulator PA28alpha in antigenpresentation. Nature, 381, 166-168, (1996)

84. CD Smith, JM Carney, PE Starke-Reed, CN Oliver, ERStadtman, RA Floyd & WR Markesbery: Excess brainprotein oxidation and enzyme dysfunction in normal agingand in Alzheimer's disease. Proc Natl Acad Sci USA 88,10540-10543, (1991)

85. PE Starke-Reed & CN Oliver: Protein oxidation andproteolysis during aging and oxidative stress. ArchBiochem Biophys 275, 559-567, (1989)

86. T Shibatani & WF Ward: Effect of age and foodrestriction on alkaline protease activity in rat liver. JGerontol 51, B316-B322, (1996)

87. W Heinemeyer, JA Kleinschmidt, J Saidowsky, CEscher & DH Wolf: Proteinase yscE, the yeastproteasome/multicatalytic-multifunctional proteinase:mutants unravel its function in stress induced proteolysisand uncover its necessity for cell survival. EMBO J 10,555-562, (1991)

88. W Hilt, C Enenkel, A Gruhler, T Singer & DH Wolf:The PRE4 gene codes for a subunit of the yeast proteasomenecessary for peptidylglutamyl-peptide-hydrolyzingactivity. Mutations link the proteasome to stress-andubiquitin-dependent proteolysis. J Biol Chem 268, 3479-3486, (1993)

89. JA Sahakian, LI Szweda, B Friguet, K Kitani, & RLLevine: Aging of the liver: Proteolysis of oxidativelymodified glutamine synthetase. Arch Biochem Biophys 318,411-417, (1995)

90. S Agarwal & RS Sohal: Age and proteolysis ofoxidized proteins. Arch Biochem Biophys 309, 24-28,(1994)

91. JX Pan, SR Short, SA Goff & JF Dice: Ubiquitin pools,ubiquitin mRNA levels, and ubiquitin-mediated proteolysisin aging human fibroblasts. Exp Gerontol 28, 39-49, (1993)

92. JH Jahngen, RD Lipman, DA Eisenhauer, EG Jahngen& A Taylor: Aging and cellular maturation cause changesin ubiquitin-eye lens conjugates. Arch Biochem Biophys276, 32-37, (1990)

93. GO Ivy, K Kitani & Y Ihara: Anomalousaccumulation of tau and ubiquitin immunoreactivities inrat brain caused by protease inhibition and by normalaging: a clue to PHF pathogenesis?. Brain Research 498,

Page 15: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

39

360-365, (1989)

94. D Finley & V Chau: Ubiquitination. Annu Rev CellBiol 7, 25-69, (1991)

95. M Hochstrasser: Ubiquitin, proteasomes, and theregulation of intracellular protein degradation. Curr OpinCell Biol 7, 215-223, (1995)

96. H Kawahara & K Tanaka: Proteasomes: The proteindeath machinery. In: Molecular Chaperones in the LifeCycle of Proteins. Eds: Fink, AL and Goto, Y. MarcelDekker, Inc. NY, 511-531, (1997)

97. F Shang, X Gong, HJ Palmer, TR Nowell & A Taylor:Age-related decline in ubiquitin conjugation in response tooxidative stress in the lens. Exp Eye Res 64, 21-30, (1997)

98. RL Mellgren & T Murachi: Intracellular Calcium-Dependent Proteolysis. CRC Press, Inc., Boca Raton, FL.(1990)

99. T Murachi: Calcium-dependent proteinases and specificinhibitors: calpain and calpastatin. Biochem Soc Symp 49,149-167, (1984)

100. P Cottin, S Poussard, JG Desmazes, D Georgescauld& A Ducastaing: Free calcium and calpain I activity.Biochim Biophys Acta 1079, 139-145, (1991)

101. RD Lane, DM Allan & RL Mellgren: A comparison ofthe intracellular distribution of m-calpain, m-calpain, andcalpastatin in proliferating human A431 cells. Exp Cell Res203, 5-16, (1992)

102. RL Mellgren: Calcium-dependent proteases: anenzyme system active at cellular membranes?. FASEB J 1,110-115, (1987)

103. AH Schmaier, HN Bradford, D Lundberg, A Farber &RW Colman: Membrane expression of platelet calpain.Blood 75, 1273-1281, (1990)

104. DE Croall & GN DeMartino: Calcium-activatedneutral protease (calpain) system: structure, function, andregulation, Physiol Rev 71, 813-847, (1991)

105. T Glaser & NS Kosower: Calpain-calpastatin andfusion. Fusability of erythrocytes is determined by aprotease-protease inhibitor [calpain-calpastatin] balance.FEBS Lett 206, 115-120, (1986)

106. A Kishimoto, K Mikawa, K Hashimoto, I Yasuda, STanaka, M Tominaga, T Kuroda & Y Nishizuka: Limitedproteolysis of protein kinase C subspecies by calcium-dependent neutral protease (calpain). J Biol Chem 264,4088-4092, (1989)

107. N Schwarz-Ben Meir, T Glaser & NS Kosower:Band 3 protein degradation by calpain is enhanced inerythrocytes of old people. Biochem J 275, 47-52, (1991)

108. D Harman: The aging process: major risk factor fordisease and death. Proc Natl Acad Sci USA 88, 5360-5363, (1991)

109. T Glaser, N Schwarz-Ben Meir, S Barnoy, SBarak, Z Eshhar & NS Kosower: Calpain (Ca+2-dependent thiol protease) in erythrocytes of young andold individuals. Proc Natl Acad Sci USA 91, 7879-7883, (1994)

110. K Saito, JS Elce, JE Hamos & RA Nixon:Widespread activation of calcium-activated neutralproteinase (calpain) in the brain in Alzheimer'sdisease: a potential molecular basis for neuronaldegeneration. Proc Natl Acad Sci USA 90, 2628-2632,(1993)

111. GG Sahagian & PM Novikoff: Lysosomes. In:Liver: Biology and Pathobiology. Ed: Arias, IM.Raven Press, NY. 275-291, (1994)

112. P Bohley & PO Seglen: Proteases and proteolysisin the lysosome. Experientia 48, 151-157, (1992)

113. L Lavie, AZ Reznick & D Gershon: Decreasedprotein and puromycinyl-peptide degradation in liversof senescent mice. Biochem J 202, 47-51, (1982)

114. L Hayflick: Recent advances in the cell biology ofaging. Mech Ageing Develop 14, 59-79, 1980

115. GO Ivy, F Schottler, J Wenzel, M Baudry & GLynch: Inhibitors of lysosomal enzymes: accumulationof lipofuscin-like dense bodies in the brain. Science226, 985-987, (1984)

116. H Glaumann: Crinophagy as a means fordegrading excess secretory proteins in rat liver. In:Current Trends in the Study of Intracellular ProteinDegradation. Eds: Knecht E, Grisolia, S. SpringerInternational, Vizcaya, Spain, 97-110, (1989)

117. SE Lenk, DL Fisher & WA Dunn Jr: Regulationof protein secretion by crinophagy in perfused ratliver. Eur J Cell Biol 56, 201-209, (1991)

118. JK Angleson & WJ Betz: Monitoring secretion inreal time: capacitance, amperometry and fluorescencecompared. Trends Neurosci 20, 281-287, (1997)

119. S Mukherjee, RN Ghosh & FR Maxfield:Endocytosis. Physiol Rev 77, 759-803, (1997)

120. JA Koenig & JM Edwardson: Endocytosis andrecycling of G protein-coupled receptors. TrendsPharmacol Sci 18, 276-287, (1997)

121. GJ Horbach, R Van Leeuwen, SH Yap & CF vanBezooijen: Changes in fluid-phase endocytosis in therat with age and their relation to total albuminelimination. Mech Ageing Dev 33, 305-312, (1986)

Page 16: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

40

122. A Brouwer, RJ Barelds & DL Knook: Age-relatedchanges in the endocytic capacity of rat liver Kupffer andendothelial cells. Hepatology 5, 362-366, (1985)

123. RP Edwards, HI Georgescu & CH Evans: Enhancedendocytosis by aging chondrocytes and fibroblasts. ExpGerontol 19, 185-190, (1984)

124. J Michl, M Tolar, V Spurna & D Rezacova: Growth-promoting alpha-globulin and ageing. In: Cell Impairmentin Aging and Development. Eds: Cristofalo, V andHoleckova, E. Plenum Press, NY, 137-146, (1975)

125. A Brouwer, MA Horan, RJ Barelds & DL Knook:Cellular aging of the reticuloendothelial system. ArchGerontol Geriatr 5, 317-324, (1986)

126. HC Lee, MA Paz & PM Gallop: Low densitylipoprotein receptor binding in aging human diploidfibroblasts in culture. J Biol Chem 257, 8912-8918, (1982)

127. P Phillips & V Cristofalo: A review of recent researchon cellular aging in culture. In: Review of BiologicalResearch in Aging. Ed: Rothstein, M. Vol. 3 Alan R. Liss,NY, 385-415, (1987)

128. R Gurley & JF Dice: Degradation of endocytosedproteins is unaltered in senescent human fibroblasts. CellBiol Int Rep 12, 885-894, (1988)

129. GE Mortimore, G Miotto, R Venerando & MKadowaki: Autophagy. Biochemistry 27, 93-135, (1996)

130. B Grinde: Autophagy and lysosomal proteolysis in theliver. Experientia 41, 1089-1095, (1985)

131. E Bergamini, M Bombara, A Del Roso, Z Gori, PMasiello, M Masini, M Pollera & S Vittorini: Theregulation of liver protein degradation by amino acids invivo. Effect of glutamine and leucine Arch Physiol Biochem103, 512-515, (1995)

132. G Rez, E Fellinger, O Oliva, Z Palfia, J Csak, LLaszlo, AL Kovacs, J Kovacs & AP Karpati: Mechanismand dynamics of macroautophagy in murine exocrinepancreatic cells. A review of vinblastine-induced changes.Acta Biol Hungarica 42, 57-86, (1991)

133. EF Blommaart, JJ Luiken, PJ Blommaart, GM vanWoerkom & AJ Meijer: Phosphorylation of ribosomalprotein S6 is inhibitory for autophagy in isolated rathepatocytes. J Biol Chem 270, 2320-2326, (1995)

134. DL Tuttle, AS Lewin & WA Dunn Jr: Selectiveautophagy of peroxisomes in methylotrophic yeasts. Eur JCell Biol 60, 283-290, (1993)

135. VI Titorenko, I Keizer, W Harder & M Veenhuis:Isolation and characterization of mutants impaired in theselective degradation of peroxisomes in the yeastHansenula polymorpha. J Bacteriol 177, 357-363, (1995)

136. HL Chiang, R Schekman & S Hamamoto: Selectiveuptake of cytosolic, peroxisomal and plasma membraneproteins into the yeast lysosome for degradation. J BiolChem 271, 9934-9941, (1996)

137. W Liou, HJ Geuze, MJ Geelen & JW Slot: Theautophagic and endocytic pathways converge at the nascentautophagic vacuoles. J Cell Biol 136, 61-70, (1997)

138. AS Stupina, AK Terman, TI Kvitnitskaia-Ryzhova,NA Mezhiborskaia & VA Zherebitskii: The age-relatedcharacteristics of autophagocytosis in different tissues oflaboratory animals. Tsitologiia Genetika 28, 15-20, (1994)

139. A Terman: The effect of age on formation andelimination of autophagic vacuoles in mouse hepatocyte.Gerontology 41, 319-325, (1995)

140. J Ahlberg & H Glaumann: Uptake--microautophagy--and degradation of exogenous proteins by isolated rat liverlysosomes. Effects of pH, ATP, and inhibitors ofproteolysis. Exp Mol Pathol 42, 78-88, (1985)

141. GE Mortimore, BR Lardeux & CE Adams: Regulationof microautophagy and basal protein turnover in rat liver.Effects of short-term starvation. J Biol Chem 263, 2506-2512, (1988)

142. G Schatz & B Dobberstein: Common principles ofprotein translocation across membranes. Science 271, 1519-1526, (1996)

143. HL Chiang & JF Dice: Peptide sequences that targetproteins for enhanced degradation during serumwithdrawal. J Biol Chem 263, 6797-6803, (1988)

144. JF Dice: Peptide sequences that target cytosolicproteins for lysosomal proteolysis. Trends Biochem Sci 15,305-309, (1990)

145. HL Chiang, SR Terlecky, CP Plant & JF Dice: A rolefor a 70 kDa heat shock protein in lysosomal degradation ofintracellular protein. Science 246, 382-385, (1989)

146. AM Cuervo & JF Dice: A receptor for the selectiveuptake and degradation of proteins by lysosomes. Science273, 501-503, (1996)

147. FA Agarraberes, SR Terlecky & JF Dice: Anintralysosomal hsp70 is required for a selective pathway oflysosomal protein degradation. J Cell Biol 137, 825-834,(1997)

148. AM Cuervo, JF Dice & E Knecht: A lysosomalpopulation responsible for the hsc73-mediated degradationof cytosolic proteins in lysosomes. J Biol Chem 272, 5606-5615, (1997)

149. NT Neff, L Bourret, P Miao & JF Dice: Degradationof proteins microinjected into IMR-90 human diploidfibroblasts. J Cell Biol 91, 184-194, (1981)

Page 17: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

41

150. JM Backer, L Bourret & JF Dice: Regulation ofcatabolism of microinjected ribonuclease A requires theamino-terminal 20 amino acids. Proc Natl Acad Sci USA80, 2166-2170, (1983)

151. AM Cuervo, SR Terlecky, JF Dice & E Knecht:Selective binding and uptake of ribonuclease A andglyceraldehyde-3-phosphate dehydrogenase by isolated ratliver lysosomes. J Biol Chem 269, 26374-26380, (1994)

152. JF Dice & SA Goff: Error catastrophe theory. In:Modern Biological Theories of Aging. Eds: Warner R,Butler RN, Sprott RL, Schneider EL. Raven Press, NY,155-168, (1987)

153. SS Wing, HL Chiang, AL Goldberg & JF Dice:Proteins containing peptide sequences related to KFERQare selectively depleted in liver and heart, but not skeletalmuscle, of fasted rats. Biochem J 275, 165-169, (1991)

154. AM Cuervo, A Palmer, AJ Rivett & E Knecht:Degradation of proteasomes by lysosomes in rat liver. EurJ Biochem 227: 792-800, (1995)

155. F Aniento, AG Papavassiliou, E Knecht & E Roche:Selective uptake and degradation of c-fos and v-fos by ratliver lysosomes. FEBS Lett 390, 47-49, (1996)

156. K Kuki, K Maeda, S Takauchi, T Kakigi, S Maeda &C Tanaka: Neurochemical and pathological alterationsfollowing infusion of leupeptin, a protease inhibitor, intothe rat brain. Dementia 7, 233-238, (1996)

157. E Bednarski, CE Ribak & G Lynch: Suppression ofcathepsins B and L causes a proliferation of lysosomes andthe formation of meganeurites in hippocampus. J Neurosci17, 4006-4021, (1997)

158. R Siman, C Gall, LS Perlmutter, C Christian, MBaudry & G Lynch: Distribution of calpain I an enzymeassociated with degenerative activity in rat brain. Brain Res347, 399-403, (1985)

159. K Blomgren & JO Karlsson: Developmental changesof calpain and calpastatin in rabbit brain. Nuerochem Res14, 1149-1152, (1989)

160. A Kenessey, M Banay-Schwartz, T DeGuzman & ALajtha: Increase in cathepsin D activity in rat brain inaging. J Nuerosci Res 23, 454-456, (1989)

161. RA Nixon, KI Saito, F Grynspan, WR Griffin, SKatayama, T Honda, PS Mohan, TB Shea & M Beermann:Calcium-activated neutral proteinase (calpain) system inaging and Alzheimer's disease. Ann NY Acad Sci 747, 77-91, (1994)

162. H Nakanishi, K Tominaga, T Amano, I Hirotsu, TInoue & K Yamamoto: Age-related changes in activitiesand localization of cathepsins D, E, B, and L in the ratbrain tissues. Exp Neurol 126, 119-128, (1994)

163. M Banay-Schwartz, T DeGuzman, M Palkovits & ALajtha: Calpain activity in adult and aged human brainregions. Neurochem Res 19, 563-567, (1994)

164. A Matus & GD Green: Age-related increase incathepsin D-like protease that degrades brain microtubule-associated proteins. Biochemistry 26, 8083-8086, (1987)

165. HC Pant: Dephosphorylation of neurofilamentproteins enhances their susceptibility to degradation bycalpain. Biochem J 256, 665-668, (1988)

166. JA Greenwood, JC Troncoso, AC Costello & GVJohnson: Phosphorylation modulates calpain-mediatedproteolysis and calmodulin binding of the 200-kDa and160-kDa neurofilament proteins. J Neurochem 61, 191-199,(1993)

167. N Ragusa, RF Villa, G Magri, F Ingrao, A Gorini, LTurpeenoja & AM Giuffrida-Stella: Modifications ofsynaptosomal plasma membrane protein composition invarious brain regions during aging. Int J Develop Neurosci10, 265-273, (1992)

168. ER Bongarzone, EF Soto & JM Pasquini: Increasedsusceptibility to degradation by trypsin and subtilisin of invitro peroxidized myelin proteins. Neurochem Res 20, 421-426, (1995)

169. M Benuck, M Banay-Schwartz, T DeGuzman & ALajtha: Changes in brain protease activity in aging. JNeurochem 67, 2019-2029, (1996)

170. E Porta, S Llesuy, AJ Monserrat, S Benavides & MTravacio: Changes in cathepsin B and lipofuscin duringdevelopment and aging in rat brain and heart.Gerontology 41, 81-89, (1995)

171. KR Braze, B Kaack & PM Klara: Lipofuscin, intra-and extraneuronal accumulation and regional distribution.In: Advances in Behavioral Biology. Eds: Ordy JM &Brizzee KR, Plenum Press, NY, 463-481,(1975)

172. AN Siakotos & D Armstrong: Age pigment, abiochemical indicator of intracellular aging. In: Advancesin Behavioral Biology. Eds: Ordy, JM & Brizzee, KR.Plenum. NY. 369-397, (1975)

173. H Nakanishi, T Amano, DF Sastradipura, YYoshimine, T Tsukuba, K Tanabe, I Hirotsu, T Ohono &K Yamamoto: Increased expression of cathepsins E and Din neurons of the aged rat brain and their colocalizationwith lipofuscin and carboxy-terminal fragments ofAlzheimer amyloid precursor protein. J Neurochem 68,739-749, (1997)

174. A Nunomura & T Miyagishi: Ultrastructuralobservations on neuronal lipofuscin (age pigment) anddense bodies induced by a proteinase inhibitor, leupeptin,in rat hippocampus Acta Neuropathol 86, 319-328, (1993)

Page 18: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

42

175. M Banay-Schwartz, T DeGuzman, A Kenessey, MPalkovits & A Lajtha: The distribution of cathepsin Dactivity in adult and aging human brain regions. JNeurochem 50, 2207-2211, (1992)

176. PL Sawant, ID Desai & AL Tappel: Factors affectingthe lysosomal membrane and availability of enzymes. ArchBiochem Biophys 105, 247-253, (1964)

177. LI Benowitz,W Rodriguez, P Paskevich, EJ Mufson,D Schenk & RL Neve: The amyloid precursor protein isconcentrated in neuronal lysosomes in normal andAlzheimer's disease subjects. Exp Neurol 106, 237-250,(1989)

178. JG Beeson, ER Shelton, HW Chan & FH Gage: Ageand damage induced changes in amyloid protein precursorimmunohistochemistry in the rat brain. J Com Neurol 342,69-77, (1994)

179. AM Cataldo, JL Barnett, SA Berman, J Li, S Quarless,S Bursztajn, C Lippa & RA Nixon: Gene expression andcellular content of cathepsin D in human neocortex:evidence for early upregulation of the endosomal-lysosomal system in pyramidal neurons in Alzheimer'sdisease. Neuron 14, 1-20, (1995)

180. C Haass, EH Koo, A Mellon, AY Hung & DJ Selkoe:Targeting of cell-surface beta-amyloid precursor protein tolysosomes: alternative processing to amyloid-bearingfragments. Nature 357, 500-503, (1992)

181. TE Golde, S Estus, LH Younkin, DJ Selkoe & SGYounkin: Processing of the amyloid protein precursor topotentially amyloidogenic derivatives. Science 255, 728-730, (1992)

182. R Siman, S Mistretta, JT Durkin, MJ Savage, T Loh, STrusko & RW Scott: Processing of the beta-amyloidprecursor; multiple proteases generate and degradepotentially amyloidogenic fragments. J Biol Chem 268,16602-16609, (1993)

183. BA Bahr, B Abai, CM Gall, PW Vanderklish, KBHoffman & G Lynch: Induction of beta-amyloid-containingpolypeptides in hippocampus: evidence for a concomitantloss of synaptic proteins and interactions with anexocitotoxin. Exp Neurol 129, 81-94, (1994)

184. I Hajimohammadreza, VE Anderson, JB Cavanagh,MP Seville, CC Nolan, BH Anderton & PN Leigh:Amyloid precursor protein fragments and lysosomal densebodies are found in rat brain neurons after ventricularinfusion of leupeptin. Brain Res 640, 25-32, (1994)

185. S Goldstein, D Stotland & RA Cordeiro: Decreasedproteolysis and increased amino acid efflux in aging humanfibroblasts. Mech Ageing Dev 5, 221-233, (1976)

186. SA Sukharev, OV Pleshakova, AB Moshnikova, VBSadovnikov & AI Gaziev: Age- and radiation-dependentchanges in carbonyl content, susceptibility to proteolysis,

and antigenicity of soluble rat liver proteins. Comp BiochemPhysiol 116, 333-338, (1997)

187. MO Bradley, JF Dice, L Hayflick & RT Schimke: Proteinalterations in aging W138 cells as determined by proteolyticsusceptibility. Exp Cell Res 96, 103-112, (1975)

188. RT Dean, S Fu, R Stocker & MJ Davies: Biochemistry andpathology of radical-mediated protein oxidation. Biochem J 324,1-18, (1997)

189. CN Oliver, BW Ahn, EJ Moerman, S Goldstein & ERStadtman: Age-related changes in oxidized proteins. J BiolChem 262, 5488-5491, (1987)

190. BR DiPaolo, RJ Pignolo & VJ Cristofalo: Identification ofproteins differentially expressed in quiescent and proliferativesenescent fibroblasts cultures. Exp Cell Res 220, 178-185, (1995)

191. RC Costa & U Del Monte: ATP-stimulated proteolysis bysoluble extracts of liver of aged rats. In: Modern AgingResearch. Protein Metabolism in Aging. Ed: Segal, HL,Rothstein, M and Bergamini, E. Wiley-Liss, NY. 151-158,(1990)

192. H Tauchi, M Hananouchi & T Sato: Accumulation oflipofuscin pigment in human hepatic cells from different racesand in different environmental conditions. Mech Ageing Dev 12,183-195, (1980)

193. G Ferland, A Perea, M Audet & B Tuchweber:Characterization of liver lysosomal enzyme activity inhepatocytes, Kupffer and endothelial cells during aging: effect ofdietary restriction. Mech Ageing Dev 56, 143-154 (1990)

194. BR DiPaolo, RJ Pignolo & VJ Cristofalo: Overexpressionof the two-chain form of cathepsin B in senescent WI-38 cells.Exp Cell Res 201, 500-505, (1992)

195. A Ishigami & S Goto: Age-related change in thedegradation rate of ovalbumin microinjected into mouse liverparenchymal cells. Arch Biochem Biophys 277, 189-195, (1990)

196. J Ahlberg, A Berkenstam, F Henell & H Glaumann:Degradation of short- and long-lived proteins in isolated rat liverlysosomes: effects of pH, temperature, and proteolytic inhibitors.J Biol Chem 260, 5847-5854, (1985)

197. G Mortimore: Mechanism and regulation of induced andbasal protein degradation in liver. In: Lysosomes: Their Role inProtein Breakdown. Eds: Glaumann H, Ballard FJ. AcademicPress, NY, 415-444, (1987)

198. K Furuno, MN Goodman & AL Goldberg: Role ofdifferent proteolytic systems in the degradation of muscleproteins during denervation atrophy. J Biol Chem 265, 8550-8557, (1990)

199. R Medina, SS Wing & AL Goldberg: Increase in levels ofpolyubiquitin and proteasome mRNA in skeletal muscle duringstarvation and denervation atrophy. Biochem J 307, 631-637,(1995)

Page 19: [Frontiers in Bioscience 3, d25-43, January 1, 1998] HOW ... · 4.2.1. Fusion of lysosomes with vesicular compartments 4.2.2. Macroautophagy and microautophagy 4.2.3. Direct transport

Effect of senescence on protein degradation

43

200. H Klitgaard, A Brunet, B Maton, C Lamaziere, C Lesty &H Monod: Morphological and biochemical changes in old ratmuscles: effect of increased use. J Appl Physiol 67, 1409-1417,(1989)

201. F Yoshizawa, T Nagasawa, N Nishizawa & R Funabiki:Protein synthesis and degradation change rapidly in response tofood intake in muscle of food-deprived mice. J Nutrition 127,1156-1159, (1997)

202. G Fruhbeck, B Muguerza, I Castilla-Cortazar & SSantidrian: Effect of aging on the rate of muscle protein turnoverin rat. Rev Esp Fisiol 52, 207-214, (1996)

203. D Dardevet, C Sornet, D Taillandier, I Savary, D Attaix & JGrizard: Sensitivity and protein turnover response toglucocorticoids are different in skeletal muscle from adult andold rats. J Clin Invest 96, 2113-2119, (1995)

204. D Attaix, D Taillandier, S Temparis, D Larbaud, EAurousseau, L Combaret & L Voisin: Regulation of ATP-ubiquitin-dependent proteolysis in muscle wasting. Reprod NutrDev 34, 583-597, (1994)

205 JJ Harding: Changes in lens proteins in cataract. In:Molecular and Cellular Biology of the Eye Lens. Ed:Bloemendal H John Wiley & Sons.,NY, 327-365, (1981)

206. A Taylor, PF Jacques & CK Dorey: Oxidants and aging:impact on vision. In: Antioxidants: Chemical, Physiological,Nutritional and Toxicological Aspects. Ed: Williams GM.Princeton Science Publishing Co., Princeton, NJ, 349-371,(1993)

207. J Blondin & A Taylor: Measures of leucine aminopeptidasecan be used to anticipate UV-induced age-related damage to lensproteins L ascorbate can delay this damage. Mech Age Dev 41,39-46, (1987)

208. A Taylor, JJ Berger, J Reddan & A Zuliani: Effects of agingin vitro on intracellular proteolysis in cultured rabbit lensepithelial cells in the presence and absence of serum. In vitroCell Develop Biol 27A, 287-292, (1991)

209. J Farber: Endogenous defenses against the cytotoxicity ofhydrogen peroxide in cultured rat hepatocytes. J Biol Chem 260,86-92, (1985)

210. DX Yin & RT Schimke: Tetracycline-controlled geneexpression system achieves high-level and quantitative control ofgene expression. Anal Biochem 235, 195-201, (1996)

Key words: Aging; Proteases, Molecular chaperones;Proteasome, Lysosomes, Calpains________________________________________________Send correspondence to: J. Fred Dice, PhD, Department ofPhysiology,Tufts University, School of Medicine,136Harrison Avenue, Boston MA 02111, Tel: (617) 636- 6707Fax: (617) 636-0445 E-mail: jdice@ opal.tufts.edu