Proteostenosis and plasma cell pathophysiology

June 7, 2017 | Autor: Eelco Van Anken | Categoria: Humans, Animals, Proteins, Neoplasms, Physiological Stress Markers, Biochemistry and cell biology
Share Embed


Descrição do Produto

This article appeared in a journal published by Elsevier. The attached copy is furnished to the author for internal non-commercial research and education use, including for instruction at the authors institution and sharing with colleagues. Other uses, including reproduction and distribution, or selling or licensing copies, or posting to personal, institutional or third party websites are prohibited. In most cases authors are permitted to post their version of the article (e.g. in Word or Tex form) to their personal website or institutional repository. Authors requiring further information regarding Elsevier’s archiving and manuscript policies are encouraged to visit: http://www.elsevier.com/copyright

Author's personal copy Available online at www.sciencedirect.com

Proteostenosis and plasma cell pathophysiology Simone Cenci, Eelco van Anken and Roberto Sitia Plasma cells differentiate from B lymphocytes to sustain antibody production. As professional secretors, they allow dissecting proteostasis in the exocytic compartment, the stresses that protein production entails and their possible roles in signaling. Most plasma cells are short-lived to limit antibody responses. After a few days of intense immunoglobulin production, they undergo apoptosis, offering a unique model of cellular senescence. Recent observations reveal that proteotoxic stresses physiologically contribute to regulate their biogenesis, function and lifespan, explaining partly the sensitivity of multiple myeloma cells to proteasome inhibitors. This essay summarizes these plasma cell lessons, and their general implications for the regulation of proteostasis, cell senescence and cancer therapeutics. Address Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Universita` Vita-Salute San Raffaele, Milano, Italy Corresponding author: Sitia, Roberto ([email protected])

Current Opinion in Cell Biology 2011, 23:216–222 This review comes from a themed issue on Cell regulation Edited by Laurie Glimcher and Claudio Hetz Available online 18th December 2010 0955-0674/$ – see front matter # 2010 Elsevier Ltd. All rights reserved. DOI 10.1016/j.ceb.2010.11.004

Introduction Cells of the B lineage have been goldmines for cell and molecular biologists, providing numerous novel paradigms of general significance [1,2!!,3]. Plasma cells, specialists in antibody (Ab) production, have yielded key mechanistic insights on the biogenesis of the endoplasmic reticulum (ER) and secretory pathway, and its astonishing efficiency and precision [4–6,7!!]. But they probably have more surprises up their sleeves (Box 1). Most plasma cells are short-lived, while some gain lifelong survival in bone marrow niches, contributing to shape immunological memory [8]. Over the past decade, a milestone observation in plasma cell pathophysiology came from the clinics, with the discovery that proteasome inhibitors (bortezomib, Velcade) are particularly effective against multiple myeloma (MM) [9!!,10,11!!], the malignant transformation of plasma cells. The sensitivity to proteasome inhibitors is a general feature of Ig-secreting plasma cells, as Ab responses to both T-dependent and Current Opinion in Cell Biology 2011, 23:216–222

independent antigens are blunted by bortezomib [12!], which offers an avenue for reversing autoimmune disease through shipwrecking autoreactive plasma cells [13!]. For many MM patients, bortezomib heralds higher chances for successful treatment. For cell biologists, it opened up novel questions and perspectives on the mechanisms that coordinate differentiation, proliferation and death of plasma cells, a powerful model for cellular senescence and lifespan control. How is their demise controlled? Is it linked to Ab synthesis and secretion, and if so how? Why are certain myelomas particularly sensitive to proteasome inhibitors? The nature of the molecular timers or counters setting apoptotic programs in normal and malignant plasma cells holds great immunological and biomedical interest [5,14]. This essay focuses on the role of proteostasis as a signaling device and therapeutic target in plasma cell pathophysiology.

How do (most) plasma cells die? As they differentiate from small B lymphocytes, plasma cells meticulously prepare for large scale synthesis, folding and secretion of Ig chains by enlarging their ER and secretory organelles [7!!]. After a few days of intense labor, in which each of them releases thousands of Ab per second, most eventually die (Figure 1). The B to plasma cell metamorphosis is orchestrated via diverse pathways. Some are lineage specific, like the transcription factor Blimp-1, whose expression is necessary and sufficient to launch the differentiation program [15]. B cell differentiation hijacks also general homeostatic pathways, amongst which a primary role is reserved for the unfolded protein response (UPR), a multidimensional signaling cascade aimed at adjusting ER capacity according to need during stress or differentiation [16,17]. Plasma cell development is blocked in the absence of the key UPR transcription factor, XBP-1 [18!!], which Box 1 The many virtues of the plasma cell model. The differentiation of B lymphocytes into plasma cells can be readily induced with mitogen or antigen, either in vitro (with inducible B lymphomas or with primary B lymphocytes obtained from different animal models, especially genetically modified mice) or in vivo, by immunization or polyclonal activation. After a proliferative burst, cell cycle arrests, differentiation allows exuberant antibody secretion and eventually massive apoptosis ensues after 3 to 6 days. The dramatic morphological and functional changes allow investigating the complex relationship between signaling via developmental and stress pathways, cell cycle, cell function, proteostasis, and lifespan control.

www.sciencedirect.com

Author's personal copy

Proteostenosis and plasma cell pathophysiology Cenci, van Anken and Sitia 217

Figure 1

Antibody secreting cells Differentiation Homing to bone marrow

Antibody titers

Apoptosis Ag Proliferation Affinity maturation

Long-lived Plasma cell

B cell Time Days

Months Current Opinion in Cell Biology

Terminal B to plasma cell differentiation. Upon encounter with antigen (Ag) in a suitable context, resting B cells rapidly proliferate, some undergoing affinity maturation and class switch to become memory cells. For the remainder, B cells rapidly differentiate into antibody secreting plasma cells that sustain massive antibody production, folding, assembly and secretion, causing a sharp rise in the titer of specific antibodies. After a few days of intense secretion, most antibody secreting cells undergo apoptosis, which explains the decrease in serum Ab titers. Some plasma cells return to the bone marrow, and home in niches where they survive for long periods, releasing antibodies at a level that confers immunological memory and protection.

promotes expansion of the secretory pathway [19!] and ER associated degradation [20!,21!,22!,23]. Increased expression of XBP-1 and its target genes also accompanies plasma cell differentiation from memory B cells [24]. While Blimp-1 promotes XBP-1 transcription [15], the mechanisms that in plasma cells activate the Ire1-dependent splicing of XBP-1 transcripts—an essential event for signal propagation [25,26]—are not fully understood. Ire1 is also required for early B cell development, independent of XBP-1 [27!], which is also dispensable for generation of memory B cells [28]. Intriguingly, another UPR pathway, propagated via PERK, is essential for pancreatic b cell survival [29!], but dispensable for B cell differentiation [14,27!,30,31]. Plasma cells may hence prove valuable to address how different branches of the UPR, once considered a monolithic response, are selectively activated in tissues. The propensity of plasma cells to undergo apoptosis is probably linked to their stressful life, following the adagium: ‘those who live fast, die young’. In view of their full gear secretory occupation, the UPR—which like most adaptive responses can lead to apoptosis if excessive or prolonged—is a primary suspect for sealing plasma cells’ fate. Yet, their development and lifespan are essentially normal in the absence of Chop, a UPR factor generally involved in maladaptive responses and cell death [14]. It remains to be elucidated whether other pro-apoptotic www.sciencedirect.com

branches of the UPR [16] are specifically engaged in plasma cells. Besides elements of the UPR, plasma cell differentiation also entails oxidative stress, which could play an important role especially considering that H2O2 is produced during Ero1-driven disulfide bond formation [32!,33!,34], and that a single plasma cell can form over 105 disulfides per second to sustain Ig folding and assembly. Plasma cells lacking peroxiredoxin IV (PrxIV), an efficient H2O2 scavenger localized in the secretory pathway [35], however, do not display a significantly different lifespan [36]. As surprisingly, plasma cells lacking both Ero1a and b display only some slowdown in Ig assembly [37!!]. Thus, additional pathways probably operate in the ER to maintain redox homeostasis [38]. Also Ca2+ signaling may contribute to plasma cell development and lifespan control, as the contact sites between mitochondria and ER—key hubs for Ca2+ and redox signal integration [39!,40!,41]—probably undergo profound reshaping during B to plasma cell differentiation. The lack of success in pinpointing whether any of these stress pathways specifically provokes plasma cell death may reflect a multifactorial and perhaps synergic control.

Challenged proteostasis as a signal Our knowledge of the biology of protein degradation advanced greatly in the past decade. Milestone discovCurrent Opinion in Cell Biology 2011, 23:216–222

Author's personal copy

218 Cell regulation

Figure 2

secretory machinery antibody production ERAD load ubiquitinated proteins proteasomal capacity days

quiescent phase

anticipatory phase

full-gear secretory phase

proteostenotic apoptosis phase Current Opinion in Cell Biology

Schematic representation of changes in proteostasis during B to plasma cell differentiation. As soon as B cells have been awakened from quiescence by Ag stimulation, they undergo a complete rearrangement, most notably expanding the secretory capacity, in anticipation of bulk antibody secretion. The complexity of antibody folding and assembly at massive loads then exceedingly burdens the folding-quality control systems with excess or aberrant antibody subunits being retrotranslocated to the cytosol via ER associated degradation (ERAD) to be disposed of by proteasomes. Paradoxically, the abundance of proteasomes decreases in differentiating cells. As a consequence poly-ubiquitinated proteins accumulate and antibody-secreting cells become proteostenotic. To cope with proteotoxicity, detergent insoluble proteins that accumulate in the lumen of the early secretory compartment (ESC) are often sorted in defined sub-regions, such as Russell Bodies [79]. Finally, the full-gear antibody production demands its toll and the cells perish, probably as a result of the accumulated proteotoxic waste.

eries include the following: a substantial pool of newly synthesized proteins is rapidly degraded by proteasomes in a variety of cell lines and tissues, hence the concept of rapidly degraded polypeptides (RDP) [42!!]; cells are equipped with excess idle proteasomes, probably as a functional reserve for the rapid clearance of aberrant proteins under stress conditions [43!,44,45]; proteasome biogenesis is tuned to demand via a proteasome stress response [46]. Owing to the intrinsic complexity of Ab molecules and their high rate of synthesis, plasma cells face a great demand for protein degradation. Like many substrates of ER quality control, unassembled Ig subunits are degraded by proteasomes [6]. Moreover, the reshaping of B cells into plasma cells increases RDP production [47!,48!!], even before Ig synthesis (Cenci et al., unpublished). Paradoxically, while the demand for proteasomal degradation increases exponentially in activated B cells, proteasome abundance and activity decrease [48!!]. As a consequence, the ubiquitin proteasome system (UPS) is overloaded (Figure 2): ubiquitinated proteins accumulate, free ubiquitin diminishes, and certain proteasome targets—including pro-apoptotic factors—are stabilized [12!,48!!]. The decrease of the proteasome pool following Current Opinion in Cell Biology 2011, 23:216–222

B cell activation is a challenging contradiction of the proteasome stress response [46]. An unfavorable proteasome load vs. capacity ratio correlates with the accumulation of Bax, Bim and IkBa and may hence sensitize plasma cells to spontaneous and pharmacological apoptosis [12!,48!!]. Indeed, such imbalance coincides with increased sensitivity to proteasome inhibitors, shedding light on their anti-myeloma toxicity [47!] (see below). The coup de grace may then be offered jointly or in a redundant fashion, by other imbalances experienced by plasma cells, such as prolonged UPR, oxidative stress, or altered Ca2+ homeostasis. From the perspective of cell biology, we are left wondering what could be the advantage for disabling the proteasome stress response in terminal B cell differentiation. A strict control of plasma cells lifespan is important to limit humoral immune response. The reduced proteostatic capacity ( proteostenosis) may provide an inbuilt counter for secreted molecules. By engineering a bottleneck in an otherwise efficient quality control strategy devoted to Ig folding and assembly, plasma cells may keep track of the work accomplished (Ab secretion), and limit the humoral response accordingly. A key issue that remains to be addressed is what decreases the proteasome complement during plasma cell differenwww.sciencedirect.com

Author's personal copy

Proteostenosis and plasma cell pathophysiology Cenci, van Anken and Sitia 219

tiation, despite the demand increases. Apart from the effects of interferon-g on the three b-peptidases that hallmark immunoproteasomes [49], and of antioxidant responses involving Nrf2 and Nrf1 [50,51!], the mechanisms regulating proteasome biogenesis in metazoans are unclear. The plasma cell paradox can provide a powerful system to address this central issue in molecular cell biology.

Proteostenosis, senescence and cell death

By linking cell death to protein degradation [48!!], the plasma cell paradigm provides a unique model for cellular aging. In yeasts and bacteria, mother cells retain damaged proteins, preventing replicative senescence [52!,53]. In metazoans, asymmetric division may have been co-opted to enable cell fate diversification, lineage commitment and morphogenesis [54]. At the cellular level, aging is characterized by a progressive reduction of the capacity to adapt to stress and maintain homeostasis [55]. Protein degradation is key to enable rapid proteome adaptations and minimize protein damage [56]. Senescent cells and tissues accumulate oxidized and damaged proteins, with increased propensity to aggregate, further challenging proteostasis [57–59]. An age-associated decline in proteasomal degradation may explain why aging predisposes to many diseases [60]. A generalized collapse of the proteostatic capacity precedes tissue damage and may be causal to the aging process in C. elegans [61!]. An integral role for proteostenosis in the aging process is also suggested by the cross-talk between signaling pathways that control lifespan and proteostasis. For example, inhi-

bition of insulin growth factor (IGF) signaling has beneficial effects in animal models of Alzheimer’s disease [62,63] and prolongs lifespan in worms, flies, and mammals [64!!,65,66]. Although the mechanism is not fully understood, reduced IGF signaling may decrease the buildup of aggregation-prone proteins and the ensuing proteotoxicity by reinforcing the cellular protein folding capacity [67,68]. By offering a cellular model of progressive proteostenosis linked to accelerated senescence, plasma cells enable the dissection of the molecular mechanisms of how proteostasis, cellular senescence and lifespan control intertwine, likely to unveil novel strategies against age-related diseases, in primis cancer.

Flipping the coin: exploiting proteotoxicity against multiple myeloma and other cancers If an unbalanced proteasomal load vs. capacity ratio limits the lifespan of normal plasma cells, we can flip the coin and ask whether we can manipulate it against MM, a condition in which cells deemed to die no longer do. The wealth of clinical reports on the efficacy of proteasome inhibitors against MM [10,11!!,69,70] generates great promise, although a substantial proportion of patients fail to respond [10,71]. The findings obtained with non-malignant plasma cells have provided clues for the diverse susceptibility of MM cells to proteasome inhibitors. In accordance with the load vs. capacity model (Figure 3), bortezomib sensitivity was found to correlate with the burden on the UPS because of either increased levels of Ig synthesis [72], high RDP

Figure 3

resistant

sensitive Current Opinion in Cell Biology

Schematical representation of the load vs. capacity model as a determinant for sensitivity to proteasomal inhibitors of MM cells. Multiple myeloma cell lines as well as primary CD138+ cells from patients display different sensitivity to proteasome inhibitors. In both cell lines and patient derived cells, sensitivity was found to correlate with the proteasomal load vs. capacity ratio. In resistant cells, the proteasomal capacity (depicted in orange) exceeds the load of poly-ubiquitinated proteins (depicted in gray). Conversely, in sensitive cells, more proteins are degraded by fewer proteasomes, generating long queues of poly-ubiquitinated proteins. The free ubiquitin pool (depicted as white spheres) decreases, and many proteins that otherwise would be proteasome substrates are no longer ubiquitinated. These proteins (depicted in dark blue) are stabilized, because they are no longer ‘visible’ to the proteasome. Such stabilization of proteins may contribute to predisposing cells to apoptosis, as has been shown for Ig-H, Bim, Bax and IkBa. www.sciencedirect.com

Current Opinion in Cell Biology 2011, 23:216–222

Author's personal copy

220 Cell regulation

production, or reduced proteasome pools [47!]. As a result, the highest vulnerability was found in MM populations hallmarked by reduced proteasome capacity and accumulation of poly-ubiquitinated proteins, clear symptoms of proteostenosis. We could therefore extrapolate the stress threshold paradigm and attempt to further manipulate stress levels against resistant MM and other malignancies. While novel inhibitors are being tested that target the proteasome differently [73], overall stress levels may be further manipulated targeting a variety of homeostatic control pathways. For instance, proteostenotic MM lines fail to increase de novo proteasome expression in response to proteasome stress, while less sensitive cells respond to proteasome stress with increased proteasome biogenesis, acquiring resistance [47!]. Hence, the mechanisms that control (immuno)proteasome biogenesis could unveil important targets for MM treatment. The emerging complementarity of UPS and autophagy [74] predicts that the latter pathway may serve an important role in plasma cells, sustaining viability and secretory function. Our preliminary evidence supports this prediction both in normal (N Pengo et al., unpublished) and malignant (F Fontana et al., unpublished) plasma cells, providing valuable models to dissect the molecular circuits linking these two degradative systems. The paradoxical discrepancy between proteasomal load and capacity observed in plasma cells opens the possibility to design, test, and exploit pro-apoptotic synergies for therapeutic intervention against MM and possibly other cancers. In particular, treatments that further compromise proteolysis or increase the degradative burden, for example, by manipulating the UPR or redox stress, may help tip the balance in favor of increased susceptibility to proteasome inhibitors.

Conclusions Studies on the B-lymphocytic lineage continue to offer novel paradigms in cell biology. The metamorphosis of long-lived B cells into short-lived Ab secreting cells provides a powerful system to recapitulate and dissect mechanistically senescence and death. Indeed, the features revealed by normal and neoplastic plasma cells support the general, novel idea that cytotoxic stress can be exploited against cancer [75–78]. Most stress responses entail apoptotic pathways that can be activated if stress duration or intensity increase, turning these responses maladaptive. In cancer, these responses are generally intact, with lower apoptotic thresholds, while physiological apoptotic responses to genotoxic stress are often disabled. Owing to deregulated growth, cancer cells generally experience more cytotoxic stress than normal counterparts (e.g. hypoxia, nutrient deprivation, pH changes, oxidative stress), and adaptive responses are often activated to higher levels, providing therapeutic specificity. Thus, strategies exploiting cytotoxic stress Current Opinion in Cell Biology 2011, 23:216–222

hold therapeutic promise against cancer, and preliminary results generate some optimism.

Acknowledgements We thank Anna Mondino for critical reading of the manuscript and helpful suggestions, Niccolo` Pengo and Francesca Fontana for providing novel results and discussions, Ana Fella and Raffaella Brambati for secretarial assistance, and AIRC, Cariplo, Ministero della Salute, MIUR, Telethon, and the Armenise-Harvard Foundation for generously supporting our work.

References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: ! of special interest !! of outstanding interest 1. Milstein C: From antibody structure to immunological diversification of immune response. Science 1986, 231:1261-1268. Turner CA Jr, Mack DH, Davis MM: Blimp-1, a novel zinc fingercontaining protein that can drive the maturation of B lymphocytes into immunoglobulin-secreting cells. Cell 1994, 77:297-306. This paper identifies Blimp-1 as a master gene dictating plasma cell differentiation.

2. !!

3.

Radbruch A, Muehlinghaus G, Luger EO, Inamine A, Smith KGC, Dorner T, Hiepe F: Competence and competition: the challenge of becoming a long-lived plasma cell. Nat Rev Immunol 2006, 6:741-750.

4.

Brewer JW, Hendershot LM: Building an antibody factory: a job for the unfolded protein response. Nat Immunol 2005, 6:23-29.

5.

Shapiro-Shelef M, Calame K: Regulation of plasma-cell development. Nat Rev Immunol 2005, 5:230-242.

6.

Sitia R, Braakman I: Quality control in the endoplasmic reticulum protein factory. Nature 2003, 426:891-894.

7. !!

van Anken E, Romijn EP, Maggioni C, Mezghrani A, Sitia R, Braakman I, Heck AJ: Sequential waves of functionally related proteins are expressed when B cells prepare for antibody secretion. Immunity 2003, 18:243-253. This paper describes the proteome changes that sustain the differentiation of antibody secreting cells, showing activation of UPR genes before massive Ig synthesis. 8.

Tokoyoda K, Hauser AE, Nakayama T, Radbruch A: Organization of immunological memory by bone marrow stroma. Nat Rev Immunol 2010, 10:193-200.

9. !!

Hideshima T, Richardson P, Chauhan D, Palombella VJ, Elliott PJ, Adams J, Anderson KC: The proteasome inhibitor PS-341 inhibits growth, induces apoptosis, and overcomes drug resistance in human multiple myeloma cells. Cancer Res 2001, 61:3071-3076. The first report on the potent in vitro effects of proteasome inhibition on human MM cell lines and freshly isolated patient MM cells. 10. Adams J: The proteasome: a suitable antineoplastic target. Nat Rev Cancer 2004, 4:349-360.

11. Richardson PG, Sonneveld P, Schuster MW, Irwin D, !! Stadtmauer EA, Facon T, Harousseau J-L, Ben-Yehuda D, Lonial S, Goldschmidt H et al.: Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Eng J Med 2005, 352:2487-2498. The first report on the clinical efficacy of the first-in-class proteasome inhibitor bortezomib against relapsed multiple myeloma. 12. Cascio P, Oliva L, Cerruti F, Mariani E, Pasqualetto E, Cenci S, ! Sitia R: Dampening Ab responses using proteasome inhibitors following in vivo B cell activation. Eur J Immunol 2008, 38:658-667. The first report on the capacity of proteasome inhibitors to dampen antibody responses in vivo in T-dependent and independent mouse immunization models. 13. Neubert K, Meister S, Moser K, Weisel F, Maseda D, Amann K, ! Wiethe C, Winkler TH, Kalden JR, Manz RA et al.: The www.sciencedirect.com

Author's personal copy

Proteostenosis and plasma cell pathophysiology Cenci, van Anken and Sitia 221

proteasome inhibitor bortezomib depletes plasma cells and protects mice with lupus-like disease from nephritis. Nat Med 2008, 14:748-755. This paper shows the efficacy of proteasome inhibitors to treat a mouse model of lupus, a paradigmatic autoAb-mediated autoimmune diseases.

28. Todd DJ, McHeyzer-Williams LJ, Kowal C, Lee A-H, Volpe BT, Diamond B, McHeyzer-Williams MG, Glimcher LH: XBP1 governs late events in plasma cell differentiation and is not required for antigen-specific memory B cell development. J Exp Med 2009, 206:2151-2159.

14. Masciarelli S, Fra AM, Pengo N, Bertolotti M, Cenci S, Fagioli C, Ron D, Hendershot LM, Sitia R: CHOP-independent apoptosis and pathway-selective induction of the UPR in developing plasma cells. Mol Immunol 2010, 47:1356-1365.

29. Harding HP, Zeng H, Zhang Y, Jungries R, Chung P, Plesken H, ! Sabatini DD, Ron D: Diabetes mellitus and exocrine pancreatic dysfunction in perk-/- mice reveals a role for translational control in secretory cell survival. Mol Cell 2001, 7:1153-1163. This paper shows that the Perk pathway is indispensable for the function of pancreatic beta cells, an established paradigm of protein folding and secretion.

15. Sciammas R, Davis MM: Blimp-1; immunoglobulin secretion and the switch to plasma cells. Curr Top Microbiol Immunol 2005, 290:201-224. 16. Ron D, Walter P: Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol 2007, 8:519-529. 17. Schroder M, Kaufman RJ: The mammalian unfolded protein response. Annu Rev Biochem 2005, 74:739-789. 18. Reimold AM, Iwakoshi NN, Manis J, Vallabhajosyula P, !! Szomolanyi-Tsuda E, Gravallese EM, Friend D, Grusby MJ, Alt F, Glimcher LH: Plasma cell differentiation requires the transcription factor XBP-1. Nature 2001, 412:300-307. This paper demonstrates that XBP-1 is essential for plasma cell differentiation. 19. Sriburi R, Jackowski S, Mori K, Brewer JW: XBP1: a link between ! the unfolded protein response, lipid biosynthesis, and biogenesis of the endoplasmic reticulum. J Cell Biol 2004, 167:35-41. This study shows that XBP-1 is sufficient to induce biosynthesis of membrane phospholipids to sustain ER biogenesis. 20. Yoshida H, Matsui T, Hosokawa N, Kaufman RJ, Nagata K, Mori K: ! A time-dependent phase shift in the mammalian unfolded protein response. Dev Cell 2003, 4:265-271. This study shows that EDEM, required for degradation of misfolded glycoproteins in the ER, is transactivated by the Ire1/XBP-1 pathway, linking ER folding capacity to ERAD. As XBP-1 is produced after ATF6 activation, the study also reveals a time-dependent transition in the mammalian UPR. 21. Friedlander R, Jarosch E, Urban J, Volkwein C, Sommer T: A ! regulatory link between ER-associated protein degradation and the unfolded-protein response. Nat Cell Biol 2000, 2:379-384. See annotation to Ref [22!]. 22. Travers KJ, Patil CK, Wodicka L, Lockhart DJ, Weissman JS, ! Walter P: Functional and genomic analyses reveal an essential coordination between the unfolded protein response and ERassociated degradation. Cell 2000, 101:249-258. This paper, and the one by Friedlander et al. [21!], shows that ERAD is intimately associated to the UPR.

30. Brewer JW, Diehl JA: PERK mediates cell-cycle exit during the mammalian unfolded protein response. Proc Natl Acad Sci USA 2000, 97:12625-12630. 31. Gass JN, Jiang H-Y, Wek RC, Brewer JW: The unfolded protein response of B-lymphocytes: PERK-independent development of antibody-secreting cells. Mol Immunol 2008, 45:1035-1043. 32. Tu BP, Weissman JS: The FAD- and O(2)-dependent reaction ! cycle of Ero1-mediated oxidative protein folding in the endoplasmic reticulum. Mol Cell 2002, 10:983-994. This paper first describes the generation of hydrogen peroxide during Ero1 dependent oxidative folding. 33. Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD, Calfon M, Sadri N, ! Yun C, Popko B, Paules R et al.: An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell 2003, 11:619-633. This paper describes the intimate links between ER stress, metabolism and redox homeostasis. 34. Wang L, Li S-J, Sidhu A, Zhu L, Liang Y, Freedman RB, Wang C-C: Reconstitution of human Ero1-Lalpha/protein-disulfide isomerase oxidative folding pathway in vitro, Positiondependent differences in role between the a and a’ domains of protein-disulfide isomerase. J Biol Chem 2009, 284:199-206. 35. Tavender TJ, Bulleid NJ: Peroxiredoxin IV protects cells from oxidative stress by removing H2O2 produced during disulfide formation. J Cell Sci 2010, 123:2672-2679. 36. Bertolotti M, Yim SH, Garcia-Manteiga JM, Masciarelli S, Kim YJ, Kang MH, Iuchi Y, Fujii J, Vene` R, Rubartelli A et al.: B- to plasmacell terminal differentiation entails oxidative stress and profound reshaping of the antioxidant responses. Antioxid Redox Signal 2010, 13:1133-1144. 37. Zito E, Chin K-T, Blais J, Harding HP, Ron D: ERO1-beta, a !! pancreas-specific disulfide oxidase, promotes insulin biogenesis and glucose homeostasis. J Cell Biol 2010, 188:821-832. This paper shows that unexpectedly mice lacking both Ero1a and b are viable and fertile, and the two oxidases have non-redundant functions.

23. Eriksson KK, Vago R, Calanca V, Galli C, Paganetti P, Molinari M: EDEM contributes to maintenance of protein folding efficiency and secretory capacity. J Biol Chem 2004, 279:44600-44605.

38. Margittai E, Sitia R: Oxidative protein folding in the secretory pathway and redox-signaling across compartments and cells. Traffic 2010.

24. Jourdan M, Caraux A, De Vos J, Fiol G, Larroque M, Cognot C, Bret C, Duperray C, Hose D, Klein B: An in vitro model of differentiation of memory B cells into plasmablasts and plasma cells including detailed phenotypic and molecular characterization. Blood 2009, 114:5173-5181.

39. Gilady SY, Bui M, Lynes EM, Benson MD, Watts R, Vance JE, ! Simmen T: Ero1alpha requires oxidizing and normoxic conditions to localize to the mitochondria-associated membrane (MAM). Cell Stress Chaperones 2010, 15:619-629. This paper reports the redox-dependent localization of Ero1a in MAM.

25. Yoshida H, Matsui T, Yamamoto A, Okada T, Mori K: XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 2001, 107:881-891.

40. Hayashi T, Su T-P: Sigma-1 receptor chaperones at the ER! mitochondrion interface regulate Ca(2+) signaling and cell survival. Cell 2007, 131:596-610. This study identifies Sigma1R as a key regulator of calcium signaling at MAMs.

26. Calfon M, Zeng H, Urano F, Till JH, Hubbard SR, Harding HP, Clark SG, Ron D: IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature 2002, 415:92-96.

41. de Brito OM, Scorrano L: An intimate liaison: spatial organization of the endoplasmic reticulum-mitochondria relationship. EMBO J 2010, 29:2715-2723.

27. Zhang K, Wong HN, Song B, Miller CN, Scheuner D, Kaufman RJ: ! The unfolded protein response sensor IRE1alpha is required at 2 distinct steps in B cell lymphopoiesis. J Clin Invest 2005, 115:268-281. This paper reveals that Ire1, but not XBP1, is required in early B cell development for the induction of the machinery that mediates Ig gene rearrangement. www.sciencedirect.com

42. Schubert U, Anton LC, Gibbs J, Norbury CC, Yewdell JW, !! Bennink JR: Rapid degradation of a large fraction of newly synthesized proteins by proteasomes. Nature 2000, 404:770-774. This is the first report of rapidly degraded proteins (RDP). 43. Dantuma NP, Lindsten K, Glas R, Jellne M, Masucci MG: Short! lived green fluorescent proteins for quantifying ubiquitin/ Current Opinion in Cell Biology 2011, 23:216–222

Author's personal copy

222 Cell regulation

proteasome-dependent proteolysis in living cells. Nat Biotechnol 2000, 18:538-543. This work devises elegant reporters of UPS activity. 44. Bence NF, Sampat RM, Kopito RR: Impairment of the ubiquitinproteasome system by protein aggregation. Science 2001, 292:1552-1555. 45. Bennett EJ, Bence NF, Jayakumar R, Kopito RR: Global impairment of the ubiquitin-proteasome system by nuclear or cytoplasmic protein aggregates precedes inclusion body formation. Mol Cell 2005, 17:351-365. 46. Hanna J, Finley D: A proteasome for all occasions. FEBS Lett 2007, 581:2854-2861. 47. Bianchi G, Oliva L, Cascio P, Pengo N, Fontana F, Cerruti F, Orsi A, ! Pasqualetto E, Mezghrani A, Calbi V et al.: The proteasome load versus capacity balance determines apoptotic sensitivity of multiple myeloma cells to proteasome inhibition. Blood 2009, 113:3040-3049. This study implicates the size of the proteasome compartment and the functional proteasome workload in the mechanisms that sensitize multiple myelomas to proteasome inhibition. 48. Cenci S, Mezghrani A, Cascio P, Bianchi G, Cerruti F, Fra A, !! Lelouard H, Masciarelli S, Mattioli L, Oliva L et al.: Progressively impaired proteasomal capacity during terminal plasma cell differentiation. EMBO J 2006, 25:1104-1113. This study describes an unexpected decrease of proteasomes in differentiating B cells, linking Ab production to cell lifespan control. 49. Macagno A, Gilliet M, Sallusto F, Lanzavecchia A, Nestle FO, Groettrup M: Dendritic cells up-regulate immunoproteasomes and the proteasome regulator PA28 during maturation. Eur J Immunol 1999, 29:4037-4042. 50. Kwak MK, Wakabayashi N, Greenlaw JL, Yamamoto M, Kensler TW: Antioxidants enhance mammalian proteasome expression through the Keap1-Nrf2 signaling pathway. Mol Cell Biol 2003, 23:8786-8794. 51. Radhakrishnan SK, Lee CS, Young P, Beskow A, Chan JY, ! Deshaies RJ: Transcription factor Nrf1 mediates the proteasome recovery pathway after proteasome inhibition in mammalian cells. Mol Cell 2010, 38:17-28. This study links proteasome biogenesis to Nrf1-dependent responses. 52. Aguilaniu H, Gustafsson L, Rigoulet M, Nystrom T: Asymmetric ! inheritance of oxidatively damaged proteins during cytokinesis. Science 2003, 299:1751-1753. A seminal paper demonstrating that carbonylated proteins, an irreversible oxidative damage product, accumulate as a function of replicative age and are asymmetrically segregated during mitosis, being retained by mother cells during cytokinesis. This active process requires Sir2, a lifespan determinant, raising the possibility that irreversibly damaged proteins may act as timers of age. 53. Lindner AB, Madden R, Demarez A, Stewart EJ, Taddei F: Asymmetric segregation of protein aggregates is associated with cellular aging and rejuvenation. Proc Natl Acad Sci USA 2008, 105:3076-3081. 54. Macara IG, Mili S: Polarity and differential inheritance— universal attributes of life? Cell 2008, 135:801-812. 55. Arking R: The Biology of Aging: Observations and Principles. 3rd ed.. New York: Oxford University Press; 2005. 56. Goldberg AL: Protein degradation and protection against misfolded or damaged proteins. Nature 2003, 426:895-899. 57. Zhang Q, Powers ET, Nieva J, Huff ME, Dendle MA, Bieschke J, Glabe CG, Eschenmoser A, Wentworth P, Lerner RA et al.: Metabolite-initiated protein misfolding may trigger Alzheimer’s disease. Proc Natl Acad Sci USA 2004, 101: 4752-4757. 58. Kiffin R, Kaushik S, Zeng M, Bandyopadhyay U, Zhang C, Massey AC, Martinez-Vicente M, Cuervo AM: Altered dynamics of the lysosomal receptor for chaperone-mediated autophagy with age. J Cell Sci 2007, 120:782-791. 59. Balch WE, Morimoto RI, Dillin A, Kelly JW: Adapting proteostasis for disease intervention. Science 2008, 319:916-919.

Current Opinion in Cell Biology 2011, 23:216–222

60. Martinez-Vicente M, Sovak G, Cuervo AM: Protein degradation and aging. Exp Gerontol 2005, 40:622-633. 61. Ben-Zvi A, Miller EA, Morimoto RI: Collapse of proteostasis ! represents an early molecular event in Caenorhabditis elegans aging. Proc Natl Acad Sci USA 2009, 106:14914-14919. Through the expression of metastable reporters of protein misfolding in C. elegans, this elegant study links defective proteostasis to aging. 62. Cohen E, Bieschke J, Perciavalle RM, Kelly JW, Dillin A: Opposing activities protect against age-onset proteotoxicity. Science 2006, 313:1604-1610. 63. Cohen E, Paulsson JF, Blinder P, Burstyn-Cohen T, Du D, Estepa G, Adame A, Pham HM, Holzenberger M, Kelly JW et al.: Reduced IGF-1 signaling delays age-associated proteotoxicity in mice. Cell 2009, 139:1157-1169. 64. Kenyon C, Chang J, Gensch E, Rudner A, Tabtiang R: A C. !! elegans mutant that lives twice as long as wild type. Nature 1993, 366:461-464. One of the first reports on mutants with altered longevity. 65. Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, Even PC, Cervera P, Le Bouc Y: IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature 2003, 421:182-187. 66. Kenyon C: The plasticity of aging: insights from long-lived mutants. Cell 2005, 120:449-460. 67. Hsu A-L, Murphy CT, Kenyon C: Regulation of aging and agerelated disease by DAF-16 and heat-shock factor. Science 2003, 300:1142-1145. 68. Morley JF, Morimoto RI: Regulation of longevity in Caenorhabditis elegans by heat shock factor and molecular chaperones. Mol Biol Cell 2004, 15:657-664. 69. San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, Spicka I, Petrucci MT, Palumbo A, Samoilova OS et al.: Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med 2008, 359:906-917. 70. Mateos M-V, Richardson PG, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, Spicka I, Petrucci MT, Palumbo A et al.: Bortezomib plus melphalan and prednisone compared with melphalan and prednisone in previously untreated multiple myeloma: updated follow-up and impact of subsequent therapy in the phase III VISTA trial. J Clin Oncol 2010, 28:2259-2266. 71. Rajkumar SV, Richardson PG, Hideshima T, Anderson KC: Proteasome inhibition as a novel therapeutic target in human cancer. J Clin Oncol 2005, 23:630-639. 72. Meister S, Schubert U, Neubert K, Herrmann K, Burger R, Gramatzki M, Hahn S, Schreiber S, Wilhelm S, Herrmann M et al.: Extensive immunoglobulin production sensitizes myeloma cells for proteasome inhibition. Cancer Res 2007, 67:1783-1792. 73. Kisselev AF: Joining the army of proteasome inhibitors. Chem Biol 2008, 15:419-421. 74. Levine B, Kroemer G: Autophagy in the pathogenesis of disease. Cell 2008, 132:27-42. 75. Ma Y, Hendershot LM: The role of the unfolded protein response in tumour development: friend or foe? Nat Rev Cancer 2004, 4:966-977. 76. Moenner M, Pluquet O, Bouchecareilh M, Chevet E: Integrated endoplasmic reticulum stress responses in cancer. Cancer Res 2007, 67:10631-10634. 77. Mathew R, Karantza-Wadsworth V, White E: Role of autophagy in cancer. Nat Rev Cancer 2007, 7:961-967. 78. Dang CV, Kim J-w, Gao P, Yustein J: The interplay between MYC and HIF in cancer. Nat Rev Cancer 2008, 8:51-56. 79. Anelli T, Sitia R: Physiology and pathology of proteostasis in the early secretory compartment. Semin Cell Dev Biol 2010, 21:520-525.

www.sciencedirect.com

Lihat lebih banyak...

Comentários

Copyright © 2017 DADOSPDF Inc.