Preprint
Concept Paper

Toward systemic lipofuscin removal

Altmetrics

Downloads

1625

Views

1400

Comments

0

A peer-reviewed article of this preprint also exists.

This version is not peer-reviewed

Submitted:

09 May 2024

Posted:

10 May 2024

You are already at the latest version

Alerts
Abstract
Lipofuscin is indigestible garbage that accumulates in the autophagic vesicles and cytosol of post-mitotic cells with age. Drs. Brunk and Terman postulated that lipofuscin accumulation is the main or at least a major driving factor in aging. They even posited that the evolution of memory is the reason why we get lipofuscin at all, as stable synaptic connections must be maintained over time, meaning that the somas of neurons must also remain in the same locale. In other words, they cannot dilute out their garbage over time through cell division. Mechanistically, their position certainly makes sense given that rendering a large percentage of a post-mitotic cell’s lysosomes useless must almost certainly negatively affect that cell and the surrounding microenvironment. It may be the case that lipofuscin accumulation is the main issue with regard to current age-related disease. Degradation in situ may be an insurmountable task currently. However, a method of systemic lipofuscin removal is discussed herein.
Keywords: 
Subject: Biology and Life Sciences  -   Aging

Graphical abstract:

A.
Preprints 106034 i001
B.
Preprints 106034 i002
A. Normal autophagy in a young, healthy cell. B. Autophagy impairment with age due to lipofuscin accumulation.

Introduction:

Biological aging is a complex molecular process that takes place over time in all organisms. However, organisms have evolved mechanisms to repair various forms of age-related damage. For example, DNA repair enzymes exist that can fix damage in nuclear DNA. Mitophagy enables the degradation of damaged mitochondria. The immune system, when one is young at least, eliminates at least some proportion of one’s senescent cells.
There are many theories about why we age, but one stands out as being the most plausible based on the evolutionary and mechanistic evidence. That is the “garbage catastrophe theory of aging.” Drs. Brunk and Terman postulated years ago that the problem of aging can essentially be summed up as a “garbage disposal issue [1].” The main idea is that basically old molecules are sometimes damaged in ways that prevent the lysosomes from breaking them down properly, and over time these damaged, old molecules accumulate inside the lysosomes. Eventually, the lysosomes become full of this indigestible garbage, i.e., “lipofuscin”, and cannot perform their normal function - then there is a garbage back-up and the cell starts to decline health-wise.
There are two arguments for lipofuscin removal being the most important goal of anti-aging science currently. One is evolutionary and one is mechanistic.

Evolutionary argument:

In nature, there are only a handful of organisms that can be said to be essentially biological immortal. Hydra vulgaris (i.e., magnipapillata) is one of such organisms, and the reason for this might be that its indigestible garbage is essentially released from its body over time. It has three cell lineages - ectodermal epithelial, interstitial, and endodermal epithelial. All the epithelial cells in its body column are stem cells that continuously divide - displacing cells toward its extremities. The cells at the extremities slough off eventually [2,3]. In terms of the interstitial lineage, the differentiated cells that its stem cells produce are closely associated with epithelial cells and so are continuously displaced as well. This is a convenient way to dispose of lipofuscin - i.e., through dilution and cell shedding. However, continuous replacement of neurons may not allow for the stable inter-neuronal interactions required for long-term memory [4].
Additionally, H. vulgaris displaces and sheds extracellular matrix (ECM), i.e., mesoglea, associated with its epithelial cells [5]. However, mesoglea in the head region remains stationary. Mesoglea in this region appears to be turned over instead, and rather slowly at that.
Lobsters continually grow throughout life; their fully differentiated cells express telomerase, allowing them to keep dividing as needed [6]. This includes the cells of their central nervous system, which allows for adult neurogenesis [7]. They also shed their shells periodically. Thus, the same logic appears to apply to them. However, their growth process does not appear to be fast enough to prevent lipofuscin from accumulating over time [8]. Notably, lipofuscin accumulation in eyestalk ganglia [9] is used as a gauge of biological age in lobsters (and myocardial lipofuscin accumulation can be used a marker of chronological age in humans [10]). Lobsters can retain memories, but only for a short time span [11].
Naked mole rats are also very long lived, and it has been shown that they have unusually active autophagic systems [12,13]. They also have very effective anti-cancer defenses [14]. However, even with enhanced autophagy, naked mole rats still accumulate lipofuscin in their post-mitotic tissues [15].
It appears as though all animals that age, e.g., flies [16], worms [17,18], lobsters, naked mole rats, mice [19], non-human primates [20], and humans [21] accumulate lipofuscin in their post-mitotic tissues. None of the aforementioned organisms seem to possess any evolutionarily “built-in” ways of exporting the lipofuscin that accumulates in their post-mitotic cells from their bodies, presumably because that would be an unnecessary expenditure of energy in light of procreation.
Export from the post-mitotic cells themselves is possible through exocytosis [22], extracellular vesicle secretion, or secretory autophagy. That part is not too energetically costly. However, from in vitro studies, it does not seem as though lipofuscin is exported from post-mitotic cells very often [23]. It also has only rarely been observed in vivo [24].
More importantly, when exported, there would ultimately be nowhere for the garbage to go except to be picked up by tissue-resident or circulating phagocytes, which themselves become bloated with lipofuscin. (Transfer of lipofuscin to tissue-resident phagocytes through tunneling nanotubes [TNTs] or partial cell fusion is also theoretically possible.)
Lipofuscin within aged tissue-resident macrophages is perhaps mostly derived from damaged molecules generated by their own, internal metabolic processes - rather than the phagocytosis of extracellular, lipid-saturated debris or efferocytosis in the context of aged, lipofuscin-laden cells [25].
Crucially, there is no evidence in the literature that tissue-resident or circulating phagocytes efficiently leave the body through migration to the gastrointestinal tract, urogenital tract, skin, or lungs, except possibly when there are infections in those areas.

Mechanistic argument:

Lipofuscin is broadly a complex amalgam of highly oxidized cross-linked macromolecules, including proteins, lipids, sugars, and metal cations. It varies in composition between species, individuals, cell types, and plausibly even cells of the same type [21,26,27,28].
While it was originally widely believed that lipofuscin is inert, it may in fact permeabilize or otherwise destabilize lysosomes and promote apoptosis or necrosis [21,29,30,31,32]. Even if the damaged molecules are mostly sequestered within lysosomes and are not actively harmful to the cell, the fact that many lysosomes become full of garbage and therefore are almost surely unable to perform their normal functions nearly as well just logically seems as though it must be a major problem for the cell. If a critical threshold is reached in enough cells in a tissue, e.g., the brain, it clearly would be problematic. The cells may try to produce more lysosomes - but will eventually reach capacity.
Along these lines, lipofuscin accumulation decreases the ability of cells to adapt to amino acid starvation [33] and increases their susceptibility to oxidative stress [34]. Increases in the dietary intake of metal cations such as Fe2+, which plays a key role in the formation of lipofuscin, augments lipofuscin accumulation [35,36,37] and speeds up aging [38]. Manganese acts similarly [39]. Furthermore, artificial lipofuscin loading into human cells results in a significant loss of cellular viability [40,41]. Another study showed that the dietary intake of artificial lipofuscin shortens the lifespan of Drosophila melanogaster [42].
With regard to humans, it has been demonstrated that multiple neuronal cell subtypes become densely packed with lipofuscin granules with age [43,44,45]. In large motor neurons of centenarians, lipofuscin constitutes up to 75% of total cytoplasmic volume [46]. Other post-mitotic cell types also accumulate substantial amounts of lipofuscin [10,47,48]. Lipofuscin-laden lysosomes are often much larger than typical lysosomes. The typical size of a lysosome in a fed, unaged cell is ~100 nm-500 nm in diameter [49]. In contrast, lipofuscin granules are generally 1-5 microns in diameter [46].
Finally, lipofuscin accumulation also explains the downward spiral of functionality that is seen in aging - i.e., the rapid acceleration in decline starting around 60-70 years of age [50]. That is because the decline in autophagy probably increases the rate at which lipofuscin is formed by allowing aggregates to stay around longer and develop further oxidative damage. Also, lipofuscin accumulation may lead to more free radical production [51], thus accelerating its accumulation as lipofuscin is heavily comprised of oxidatively damaged biomolecules. Furthermore, eventually, when autophagy levels have decreased substantially in many cells in a tissue, the rate of accumulation of other forms of age-related damage probably accelerates as well, which could further accelerate lipofuscin accumulation.

Clearance of Undigested Rubbish via Extraction (CURE):

I propose a strategy for systemic lipofuscin removal called “Clearance of Undigested Rubbish via Extraction” (CURE). As formulated currently, CURE involves three steps. The first step is to replace a patient’s endogenous tissue-resident macrophages (TRMs) with edited variants. The second step is to administer a small molecule that induces these edited TRMs to transiently become hyper-motile and export molecular instructions for the secretion of mixed-age lysosomes from target cells. The third step is for them each to phagocytose an experimentally determined number of those lysosomes and then asymmetrically divide, wherein one progeny cell inherits all the phagosomes containing exported lysosomes and then migrates to an extraction point in the body.
There appear to be three TRM subsets: one is completely self-renewing, another is partially self-renewing, and the last is completely reseeded by HSCs. However, this was shown to be the case in a parabiotic system with a lack of prior depletion [52]. Under continuous endogenous depletion conditions, those subsets may be reseeded entirely by HSC-derived TRMs. For example, when TRMs were cleared in mice using a CD45 antibody-drug conjugate (ADC), they were replaced by HSC-derived monocytes [53]. Notably, HSCs were recently shown to contribute to TRM population maintenance over time in non-human primates at all sites tested even in the absence of conditioning [54]. Thus, HSC-mediated TRM replacement may be a viable option. If so, patient HSCs could be harvested and edited ex vivo or edited in vivo using a dual adenovirus system to produce certain genetic alterations and install the necessary therapeutic genes under a macrophage-specific promoter [55,56].
Systemic depletion of TRMs could then potentially be effected via small molecule CSF1R inhibitors in combination with anti-CSF1R and anti-CD45 ADCs [53,57,58,59,60,61,62,63,64,65]. If the HSCs are edited ex vivo, the CD45 ADC would also suffice as non-genotoxic conditioning for subsequent transplant [66]. If the HSCs are edited in vivo, their cd45 gene could be altered to preclude recognition of the corresponding protein by the CD45 ADC, thus allowing for in vivo selection of the edited HSCs [67].
While the FDA-approved small molecule CSF1R inhibitor PLX3397 can deplete peripheral TRMs as well as microglia, it may not do so as efficiently for certain subtypes [62,63]. However, the csf1r gene in patient HSCs could be edited to confer resistance to PLX3397, allowing for the continuous depletion of peripheral TRMs over a longer period of time [61]. Transplanted HSCs could encode a diphtheria toxin-resistance marker [68]. That way, progeny TRMs could secrete a CD11b or CD64 immunotoxin within their host tissues after arriving at their niches, helping to eliminate any remaining endogenous TRMs.
After systemic TRM replacement, a small molecule could be administered to induce hyper-motility and the production of an RNA delivery vector. This vector would induce lipofuscin export. While lysosomal exocytosis is a possibility for cellular ejection of lipofuscin, this would release it in a bare form, which may break apart and be more difficult for TRMs to phagocytose than lysosomes. They could also not count the number of phagocytosis events without binding to a specified ligand. A better option may be to enact secretory autophagy of lipofuscin-laden lysosomes [69]. TFEB overexpression would generate new lysosomes in the cell to compensate for some of the remaining unladen or partially lysosomes being exported via secretory autophagy. (TFEB-mediated lysosomal exocytosis should be inhibited, however.)
TRM hyper-motility could be induced via a self-generated chemokine gradient and a synthetic gene circuit like a noisy repressilator [70,71]. Alternatively, for microglia, hyper-motility could potentially be effected via inducible LRRK2 inhibition [72]. LRRK2 inhibition may have the opposite effect on peripheral TRMs as it does on microglia, however [73]. Thus, for peripheral TRMs at least, Bmal1 inhibition may be more suitable [74]. Bmal1 inhibition may increase microglial motility as well.
COURIER is a cell-to-cell communication system that was recently developed. It involves the secretion of mRNA that is packaged in protein nanocages [75]. The packaging capacity of COURIER is at least 9.8 kb. Alternative protein nanocage architectures could theoretically allow for larger cargo capacities if necessary. A polyprotein could be expressed that includes the tobacco etch virus protease, which would cleave it apart after translation. 2A sequences could also be used to express multiple proteins.
The cargo mRNA could encode a nanobody targeting LAMP-1 with a fused domain that interacts with Sec22b - as well as an LC3-interaction region (LIR) [76]. FKBP51 overexpression may also be important [77]. If more packaging space is available, overexpression of Sec22b, Stx3/4, and SNAP23/29 may also help [69]. Furthermore, autophagosome-lysosome fusion could be inhibited via a dominant-negative STX17 variant [ [78]].
The next step would be for the hyper-motile TRMs to phagocytose the mixed-age lysosomes. Notably, mitochondria secreted via secretory autophagy seem to lack an encapsulating membrane after export [79]; thus, in the case of lysosomes, a lysosomal membrane protein, e.g., LAMP-1, could be targeted by TRMs [80]. The final step would be for the garbage-disposal cells to migrate to an extraction point in the body when full.
The edited TRMs could also employ a synthetic gene circuit to count the number of lysosomes they phagocytose [81,82]. In this context, the phagosomes containing said lysosomes should be unable to fuse with TRM lysosomes. Once an experimentally determined number of lysosomes have been phagocytosed, the given TRM would asymmetrically divide; one progeny cell would inherit all of the aforementioned phagosomes [83,84]. That progeny cell would also irreversibly express a receptor mediating its chemoattraction to the gastrointestinal lumen by responding to a blood-brain barrier (BBB)-permeable small molecule produced by the patient’s gut microbiome bacteria [85]. Alternatively, if necessary, a device could be placed in the peritoneal space that slowly releases a chemoattractant for the TRMs - allowing for non-invasive, albeit manually effected egress [86,87,88].
While in most tissues TRMs make up ~10–15% of the constituent cells, in some tissues they are less prevalent [89,90]. Induced replication of edited TRMs may be necessary in some tissues, which could be achieved via small molecule administration in combination with tuned TRM subset-specific promoters or miRNAs. It will be important to ensure that this does not cause excessive inflammation. M1 macrophages are typically considered inflammatory, while M2 macrophages are anti-inflammatory. Thus, it may be best to skew TRM polarity towards M2.
Random motility should only be induced when periodic treatment is required. Otherwise, it may impair the ability of the TRMs to fight infection.
Some of the edited TRMs may wander outside of their host tissues, but perhaps they would assume the identity of whatever tissue they end up in.
Eliminating the TRMs of elderly patients initially may release a large quantity of lipofuscin into tissues. In this case, asymmetric division of the edited TRMs could initially involve lysosomal inheritance rather than phagosomal inheritance.
Elderly patients will likely need an extensive first lipofuscin removal session. Once the garbage back-up is cleared, oxidatively damaged mitochondria would then be mitophagocytosed and cytoplasmic lipofuscin granules would be engulfed by autophagosomes and delivered to new lysosomes. Thus, new lysosomes might be quickly overwhelmed again - and they should be exported as well. Aged cells may also have autophagosomes that have been present for too long in the cytoplasm. Their outer membranes may have become “corrupted" by oxidatively warped lipids; thus, they may not even be able to fuse with new lysosomes. They might have to be exported as well. Also, if ECM turnover efficiency is restored to youthful levels, cross-linked proteins could be excised from the surrounding ECM and taken up; at least some cross-links may be indigestible by lysosomal enzymes [91].
Ideally, whole-body lipofuscin removal would perhaps be effected once every decade - starting at ~30 years of age.

Testing CURE in animal models:

Proof-of-concept for lipofuscin removal could potentially be undertaken in an animal model with a multitude of transgenes installed in a genomic safe harbor locus [55] once secretory autophagy of lysosomes can be reliably induced in a wide variety of target cell types.
The mouse TRMs could inducibly become hyper-motile and secrete a peptide that activates a synthetic receptor in target cells [92], triggering transient TFEB overexpression and mixed-age lysosome secretion.
In short-lived species, like mice and rats, lipofuscin may not have enough time to accumulate to pathological levels before they die of cancer. It is estimated that 50-90% of aged mice die of cancer [93]. Even still, it was shown that in the cerebral cortex neurons of lamina Vb in 630-700-day old rats, lipofuscin occupied 23% of the soma volume [94,95]. This could still certainly have a negative physiological effect. Unsurprisingly, we do see a cognitive decline in mice with age [96]. However, even the oldest mice do not develop age-related neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, or amyotrophic lateral sclerosis [97]. Ostensibly, they simply do not live long enough for sufficient build-up of lipofuscin in their neurons. Other conditions like age-related macular degeneration and sarcopenia do occur in mice, but whether their most severe cases are as bad as the most severe human cases is unclear to me. It is also possible that some mouse tissues accumulate lipofuscin more rapidly than others due to cell type differences in metabolic rates, etc. Some types of human neurons, for example, have not accumulated much lipofuscin by the time others are nearly full[98].
One way to test CURE would be to utilize a mouse model with a Cre-inducible, neuron-specific lysosomal storage disorder in combination with genetically-altered microglial variants [99].
Also, extraction from the peritoneal space would perhaps be easiest initially, as deschloroclozapine [87,88] is BBB-permeable and can be slowly released from biodegradable beads placed there laparoscopically [86,87].
For age-related proof-of-concept, non-human primates would be the best model organisms. However, perhaps pigs, cats, or dogs would be useful model organisms in this context, as well.

Discussion:

Another possible option for removing lipofuscin in the future involves TRM partial cell fusion with post-mitotic cells and acquisition of aged lysosomes.
Fixing nuclear DNA mutations and damage in stem cells and long-lived post-mitotic cells on a fundamental level may be the most difficult challenge we face with aging. Notably, it has become clear that nuclear DNA mutations damage accumulate with age in post-mitotic cells as well as stem cells [100].
There may only be only feasible strategy for addressing this on fundamental level in the relatively near future - “whole-body induced cell turnover” [101]. Edited TRMs could eliminate adult stem cells via immunotoxins. They could also asymmetrically divide, wherein one progeny cell dedifferentiates into an iPSC via Yamanaka factor expression. The empty niche could potentially then guide the iPSC into engrafting and differentiating into the appropriate adult stem cell type. Then, the new, edited adult stem cell could be induced to kill tissue-resident cells and divide to repopulate the tissue. This could be a viable strategy even in the brain, if done slowly over time - and it may be necessary in the short-term. The telomeres of the edited, adult stem cells could be elongated via hTERT overexpression to enable more rapid repopulation of tissues than is typical.
However, iPSCs can form teratomas in vivo, so this may not be appropriate. The progeny cell of the TRMs that is for reseeding adult stem cells may need to more directly convert to the given stem cell type instead.
Dr. Aubrey de Grey and Ben Zealley have suggested allotopic expression as a means of addressing mtDNA mutations with age [102]. An intercellular communication system involving DNA transfer may be possible in the future. Alternatively, bacteria can conjugate with mitochondria, although whether second strand synthesis would naturally occur after DNA transfer is unclear [103].

Funding

Funding not received for the study.

Authors' contributions

M.R. wrote the paper.

Ethics approval and consent to participate

N/A

Consent for publication

N/A

Availability of data and material

N/A

Acknowledgements

The graphical abstract was created with BioRender.com.

Competing interests

The author declares that he has no competing interests.

References

  1. Terman A and Brunk UT. Lipofuscin. The International Journal of Biochemistry & Cell Biology 2004;36(8):1400–1404; [CrossRef]
  2. Siebert S, Farrell JA, Cazet JF, et al. Stem Cell Differentiation Trajectories in Hydra Resolved at Single-Cell Resolution. Science 2019;365(6451):eaav9314; [CrossRef]
  3. Murad R, Macias-Muñoz A, Wong A, et al. Coordinated Gene Expression and Chromatin Regulation during Hydra Head Regeneration. Genome Biology and Evolution 2021;13(12):evab221; [CrossRef]
  4. Terman A, Brunk UT. Is aging the price for memory? Biogerontology (2005) 6:205–210. [CrossRef]
  5. Aufschnaiter R, Zamir EA, Little CD, Özbek S, Münder S, David CN, et al. In vivo imaging of basement membrane movement: ECM patterning shapes Hydra polyps. Journal of Cell Science 2011;124:4027–38. [CrossRef]
  6. Klapper W, Kühne K, Singh KK, Heidorn K, Parwaresch R, Krupp G. Longevity of lobsters is linked to ubiquitous telomerase expression. FEBS Letters (1998) 439:143–146. [CrossRef]
  7. Beltz BS, Sandeman DC. Regulation of life-long neurogenesis in the decapod crustacean brain. Arthropod Structure & Development (2003) 32:39–60. [CrossRef]
  8. Peregrim I. Why we age — a new evolutionary view. Biologia (2017) 72:475–485. [CrossRef]
  9. Sheehy M, Shelton P, Wickins J, Belchier M, Gaten E. Ageing the European lobster Homarus gammarus by the lipofuscin in its eyestalk ganglia. Mar Ecol Prog Ser (1996) 143:99–111. [CrossRef]
  10. Kakimoto Y, Okada C, Kawabe N, Sasaki A, Tsukamoto H, Nagao R, Osawa M. Myocardial lipofuscin accumulation in ageing and sudden cardiac death. Sci Rep (2019) 9:3304. [CrossRef]
  11. Karavanich C, Atema J. Individual recognition and memory in lobster dominance. Animal Behaviour (1998) 56:1553–1560. [CrossRef]
  12. Zhao S, Lin L, Kan G, Xu C, Tang Q, Yu C, Sun W, Cai L, Xu C, Cui S. High autophagy in the naked mole rat may play a significant role in maintaining good health. Cell Physiol Biochem (2014) 33:321–332. [CrossRef]
  13. Triplett JC, Tramutola A, Swomley A, Kirk J, Grimes K, Lewis K, Orr M, Rodriguez K, Cai J, Klein JB, et al. Age-related changes in the proteostasis network in the brain of the naked mole-rat: Implications promoting healthy longevity. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease (2015) 1852:2213–2224. [CrossRef]
  14. Hadi F, Kulaberoglu Y, Lazarus KA, Bach K, Ugur R, Beattie P, Smith ESJ, Khaled WT. Transformation of naked mole-rat cells. Nature (2020) 583:E1–E7. [CrossRef]
  15. Edrey YH, Hanes M, Pinto M, Mele J, Buffenstein R. Successful aging and sustained good health in the naked mole rat: a long-lived mammalian model for biogerontology and biomedical research. ILAR J (2011) 52:41–53. [CrossRef]
  16. Panno JP, Nair KK. Effects of increased lifespan on chromatin condensation in the adult male housefly. Mech Ageing Dev (1986) 35:31–38. [CrossRef]
  17. Clokey GV, Jacobson LA. The autofluorescent “lipofuscin granules” in the intestinal cells of Caenorhabditis elegans are secondary lysosomes. Mech Ageing Dev (1986) 35:79–94. [CrossRef]
  18. Houthoofd K, Braeckman BP, Lenaerts I, Brys K, De Vreese A, Van Eygen S, Vanfleteren JR. Ageing is reversed, and metabolism is reset to young levels in recovering dauer larvae of C. elegans. Exp Gerontol (2002) 37:1015–1021. [CrossRef]
  19. Goyal VK. Lipofuscin pigment accumulation in the central nervous system of the mouse during aging. Exp Gerontol (1982) 17:89–94. [CrossRef]
  20. Gilissen EP, Staneva-Dobrovski L. Distinct Types of Lipofuscin Pigment in the Hippocampus and Cerebellum of Aged Cheirogaleid Primates. The Anatomical Record (2013) 296:1895–1906. [CrossRef]
  21. Moreno-García A, Kun A, Calero O, Medina M, Calero M. An Overview of the Role of Lipofuscin in Age-Related Neurodegeneration. Frontiers in Neuroscience (2018) 12: https://www.frontiersin.org/article/10.3389/fnins.2018.00464 [Accessed March 31, 2022].
  22. Gray DA and Woulfe J. Lipofuscin and Aging: A Matter of Toxic Waste. Science of Aging Knowledge Environment 2005;2005(5):re1–re1; [CrossRef]
  23. Terman A, Brunk UT. Is Lipofuscin Eliminated from Cells? Investigative Ophthalmology & Visual Science (1999) 40:2463–2464.
  24. Wang L, Xiao C-Y, Li J-H, Tang G-C, Xiao S-S. Transport and Possible Outcome of Lipofuscin in Mouse Myocardium. Adv Gerontol 2022;12:247–63. [CrossRef]
  25. Burns JC, Cotleur B, Walther DM, Bajrami B, Rubino SJ, Wei R, Franchimont N, Cotman SL, Ransohoff RM, Mingueneau M. Differential accumulation of storage bodies with aging defines discrete subsets of microglia in the healthy brain. eLife (2020) 9:e57495. [CrossRef]
  26. Boellaard JW and Schlote W. Ultrastructural Heterogeneity of Neuronal Lipofuscin in the Normal Human Cerebral Cortex. Acta Neuropathol 1986;71(3–4):285–294; [CrossRef]
  27. Sohal RS, Wolfe LS. Chapter 11 Lipofuscin: characteristics and significance. In: Swaab DF, Fliers E, Mirmiran M, Van Gool WA, Van Haaren F, editors. Progress in Brain Research, vol. 70, Elsevier; 1986, p. 171–83. [CrossRef]
  28. Sheehy MRJ. Individual variation in, and the effect of rearing temperature and body size on, the concentration of fluorescent morphological lipofuscin in the brains of freshwater crayfish, Cherax cuspidatus (Crustacea: Parastacidae). Comparative Biochemistry and Physiology Part A: Physiology 1990;96:281–6. [CrossRef]
  29. Brunk UT, Terman A. Lipofuscin: mechanisms of age-related accumulation and influence on cell function. Free Radic Biol Med (2002) 33:611–619. [CrossRef]
  30. Kurz T, Terman A, Gustafsson B, et al. Lysosomes and oxidative stress in aging and apoptosis. Biochimica et Biophysica Acta (BBA) - General Subjects 2008;1780(11):1291–1303; [CrossRef]
  31. Gabandé-Rodríguez E, Keane L, Capasso M. Microglial phagocytosis in aging and Alzheimer’s disease. Journal of Neuroscience Research 2020;98(2):284–298; [CrossRef]
  32. Pan C, Banerjee K, Lehmann GL, et al. Lipofuscin causes atypical necroptosis through lysosomal membrane permeabilization. Proceedings of the National Academy of Sciences 2021;118(47):e2100122118; [CrossRef]
  33. Terman A, Dalen H, Brunk UT. Ceroid/lipofuscin-loaded human fibroblasts show decreased survival time and diminished autophagocytosis during amino acid starvation. Experimental Gerontology 1999;34(8):943–957; [CrossRef]
  34. Terman A, Abrahamsson N, Brunk UT. Ceroid/lipofuscin-loaded human fibroblasts show increased susceptibility to oxidative stress. Exp Gerontol 1999;34(6):755–770; [CrossRef]
  35. Lv Z, Jiang H, Xu H, et al. Increased iron levels correlate with the selective nigral dopaminergic neuron degeneration in Parkinson’s disease. J Neural Transm 2011;118(3):361–369; [CrossRef]
  36. Maccarinelli F, Pagani A, Cozzi A, et al. A novel neuroferritinopathy mouse model (FTL 498InsTC) shows progressive brain iron dysregulation, morphological signs of early neurodegeneration and motor coordination deficits. Neurobiology of Disease 2015;81:119–133; [CrossRef]
  37. Bhoiwala D, Song Y, Cwanger A, et al. High iron diet causes elevation of retinal iron levels and RPE autofluorescence. Investigative Ophthalmology & Visual Science 2015;56(7):4203.
  38. Mangan D. Iron: an underrated factor in aging. Aging 2021;13(19):23407–23415; [CrossRef]
  39. Ohgami N, Yajima I, Iida M, et al. Manganese-mediated acceleration of age-related hearing loss in mice. Sci Rep 2016;6(1):36306; [CrossRef]
  40. Höhn A, Grune T. Lipofuscin: formation, effects and role of macroautophagy. Redox Biol 2013;1(1):140–144; [CrossRef]
  41. von Zglinicki T, Nilsson E, Döcke WD, et al. Lipofuscin accumulation and ageing of fibroblasts. Gerontology 1995;41 Suppl 2:95–108; [CrossRef]
  42. Tsakiri EN, Iliaki KK, Höhn A, et al. Diet-derived advanced glycation end products or lipofuscin disrupts proteostasis and reduces life span in Drosophila melanogaster. Free Radic Biol Med 2013;65:1155–1163; [CrossRef]
  43. Mann DM, Yates PO, Stamp JE. The relationship between lipofuscin pigment and ageing in the human nervous system. J Neurol Sci (1978) 37:83–93. [CrossRef]
  44. Goyal VK. Lipofuscin pigment accumulation in human brain during aging. Experimental Gerontology 1982;17(6):481–487; [CrossRef]
  45. Benavides SH, Monserrat AJ, Fariña S, et al. Sequential histochemical studies of neuronal lipofuscin in human cerebral cortex from the first to the ninth decade of life. Archives of Gerontology and Geriatrics 2002;34(3):219–231; [CrossRef]
  46. Yin D. Biochemical basis of lipofuscin, ceroid, and age pigment-like fluorophores. Free Radical Biology and Medicine 1996;21(6):871–888; [CrossRef]
  47. Wing GL, Blanchard GC, Weiter JJ. The topography and age relationship of lipofuscin concentration in the retinal pigment epithelium. Investigative Ophthalmology & Visual Science (1978) 17:601–607.
  48. Dayan D, Abrahami I, Buchner A, Gorsky M, Chimovitz N. Lipid pigment (lipofuscin) in human perioral muscles with aging. Experimental Gerontology (1988) 23:97–102. [CrossRef]
  49. Xu H, Ren D. Lysosomal Physiology. Annu Rev Physiol (2015) 77:57–80. [CrossRef]
  50. Kang, Y.-K., Min, B., Eom, J. & Park, J. S. Different phases of aging in mouse old skeletal muscle. Aging (Albany NY) 14, 143–160 (2022).
  51. Vida C, de Toda IM, Cruces J, et al. Role of macrophages in age-related oxidative stress and lipofuscin accumulation in mice. Redox Biol 2017;12:423–437; [CrossRef]
  52. Dick SA, Wong A, Hamidzada H, Nejat S, Nechanitzky R, Vohra S, et al. Three tissue resident macrophage subsets coexist across organs with conserved origins and life cycles. Science Immunology 2022;7:eabf7777. [CrossRef]
  53. Gustafsson K, Rhee C, Frodermann V, Scadden EW, Li D, Iwamoto Y, et al. Clearing and replacing tissue-resident myeloid cells with an anti-CD45 antibody–drug conjugate. Blood Advances 2023;7:6964–73. [CrossRef]
  54. Rahmberg AR, Wu C, Shin T, Hong SG, Pei L, Markowitz TE, et al. Ongoing production of tissue-resident macrophages from hematopoietic stem cells in healthy adult macaques. Blood Adv 2023;8:523–37. [CrossRef]
  55. Fischer K, Kraner-Scheiber S, Petersen B, Rieblinger B, Buermann A, Flisikowska T, et al. Efficient production of multi-modified pigs for xenotransplantation by ‘combineering’, gene stacking and gene editing. Scientific Reports 2016;6:29081. [CrossRef]
  56. Wang H, Georgakopoulou A, Zhang W, Kim J, Gil S, Ehrhardt A, et al. HDAd6/35++ - A new helper-dependent adenovirus vector platform for in vivo transduction of hematopoietic stem cells. Molecular Therapy Methods & Clinical Development 2023;29:213–26. [CrossRef]
  57. Sung CYW, Hayase N, Yuen PST, Lee J, Fernandez K, Hu X, et al. Macrophage Depletion Protects Against Cisplatin-Induced Ototoxicity and Nephrotoxicity. bioRxiv 2023:2023.11.16.567274. [CrossRef]
  58. Lund H, Pieber M, Parsa R, Han J, Grommisch D, Ewing E, et al. Competitive repopulation of an empty microglial niche yields functionally distinct subsets of microglia-like cells. Nat Commun 2018;9:4845. [CrossRef]
  59. Hillmer AT, Holden D, Fowles K, Nabulsi N, West BL, Carson RE, et al. Microglial depletion and activation: A [11C]PBR28 PET study in nonhuman primates. EJNMMI Res 2017;7:59. [CrossRef]
  60. Green KN, Crapser JD, Hohsfield LA. To Kill Microglia: A Case for CSF1R Inhibitors. Trends Immunol 2020;41:771–84. [CrossRef]
  61. Chadarevian JP, Lombroso SI, Peet GC, Hasselmann J, Tu C, Marzan DE, et al. Engineering an inhibitor-resistant human CSF1R variant for microglia replacement. Journal of Experimental Medicine 2022;220:e20220857. [CrossRef]
  62. Claeys W, Verhaege D, Van Imschoot G, Van Wonterghem E, Van Acker L, Amelinck L, et al. Limitations of PLX3397 as a microglial investigational tool: peripheral and off-target effects dictate the response to inflammation. Front Immunol 2023;14:1283711. [CrossRef]
  63. Shaikh SN, Willis EF, Dierich M, Xu Y, Stuart SJS, Gobe GC, et al. CSF-1R inhibitor PLX3397 attenuates peripheral and brain chronic GVHD and improves functional outcomes in mice. J Neuroinflammation 2023;20:300. [CrossRef]
  64. Butowski N, Colman H, De Groot JF, Omuro AM, Nayak L, Wen PY, et al. Orally administered colony stimulating factor 1 receptor inhibitor PLX3397 in recurrent glioblastoma: an Ivy Foundation Early Phase Clinical Trials Consortium phase II study. Neuro Oncol 2016;18:557–64. [CrossRef]
  65. MacDonald KPA, Palmer JS, Cronau S, Seppanen E, Olver S, Raffelt NC, et al. An antibody against the colony-stimulating factor 1 receptor depletes the resident subset of monocytes and tissue- and tumor-associated macrophages but does not inhibit inflammation. Blood 2010;116:3955–63. [CrossRef]
  66. Hayal TB, Wu C, Abraham D, Demirci S, Palchaudhuri R, Lanieri L, et al. The Impact of CD45-Antibody-Drug Conjugate Conditioning on Clonal Dynamics and Immune Tolerance Post HSPC Transplantation in Rhesus Macaques. Blood 2023;142:3419. [CrossRef]
  67. Wellhausen N, O’Connell RP, Lesch S, Engel NW, Rennels AK, Gonzales D, et al. Epitope base editing CD45 in hematopoietic cells enables universal blood cancer immune therapy. Sci Transl Med 2023;15:eadi1145. [CrossRef]
  68. Picco G, Petti C, Trusolino L, Bertotti A, Medico E. A diphtheria toxin resistance marker for in vitro and in vivo selection of stably transduced human cells. Sci Rep 2015;5:14721. [CrossRef]
  69. Dedicated SNAREs and specialized TRIM cargo receptors mediate secretory autophagy. The EMBO Journal 2017;36:42–60. [CrossRef]
  70. Insall RH, Paschke P, Tweedy L. Steering yourself by the bootstraps: how cells create their own gradients for chemotaxis. Trends in Cell Biology 2022;32:585–96. [CrossRef]
  71. Tigges M, Marquez-Lago TT, Stelling J, Fussenegger M. A tunable synthetic mammalian oscillator. Nature 2009;457:309–12. [CrossRef]
  72. Choi I, Kim B, Byun J-W, Baik SH, Huh YH, Kim J-H, et al. LRRK2 G2019S mutation attenuates microglial motility by inhibiting focal adhesion kinase. Nat Commun 2015;6:8255. [CrossRef]
  73. Russo I, Bubacco L, Greggio E. LRRK2 as a target for modulating immune system responses. Neurobiology of Disease 2022;169:105724. [CrossRef]
  74. Kitchen GB, Cunningham PS, Poolman TM, Iqbal M, Maidstone R, Baxter M, et al. The clock gene Bmal1 inhibits macrophage motility, phagocytosis, and impairs defense against pneumonia. Proc Natl Acad Sci U S A 2020;117:1543–51. [CrossRef]
  75. Horns F, Martinez JA, Fan C, Haque M, Linton JM, Tobin V, et al. Engineering RNA export for measurement and manipulation of living cells. Cell 2023;186:3642-3658.e32. [CrossRef]
  76. Birgisdottir ÅB, Lamark T, Johansen T. The LIR motif - crucial for selective autophagy. J Cell Sci 2013;126(Pt 15):3237–3247; [CrossRef]
  77. Martinelli S, Anderzhanova EA, Bajaj T, Wiechmann S, Dethloff F, Weckmann K, et al. Stress-primed secretory autophagy promotes extracellular BDNF maturation by enhancing MMP9 secretion. Nat Commun 2021;12:4643. [CrossRef]
  78. Uematsu M, Nishimura T, Sakamaki Y, et al. Accumulation of undegraded autophagosomes by expression of dominant-negative STX17 (syntaxin 17) mutants. Autophagy 2017;13(8):1452–1464; [CrossRef]
  79. Tan HWS, Lu G, Dong H, Cho Y-L, Natalia A, Wang L, et al. A degradative to secretory autophagy switch mediates mitochondria clearance in the absence of the mATG8-conjugation machinery. Nat Commun 2022;13:3720. [CrossRef]
  80. Morrissey MA, Williamson AP, Steinbach AM, Roberts EW, Kern N, Headley MB, et al. Chimeric antigen receptors that trigger phagocytosis. eLife 2018;7:e36688. [CrossRef]
  81. Friedland AE, Lu TK, Wang X, Shi D, Church G, Collins JJ. Synthetic Gene Networks that Count. Science 2009;324:1199–202. [CrossRef]
  82. VanHook AM. Macrophages don’t take more than they can eat. Science Signaling 2017;10:eaao1183. [CrossRef]
  83. Watson JL, Krüger LK, Ben-Sasson AJ, Bittleston A, Shahbazi MN, Planelles-Herrero VJ, et al. Synthetic Par polarity induces cytoskeleton asymmetry in unpolarized mammalian cells. Cell 2023;186:4710-4727.e35. [CrossRef]
  84. Loeffler D, Wehling A, Schneiter F, Zhang Y, Müller-Bötticher N, Hoppe PS, et al. Asymmetric lysosome inheritance predicts activation of haematopoietic stem cells. Nature 2019;573:426–9. [CrossRef]
  85. Ronda C, Chen SP, Cabral V, Yaung SJ, Wang HH. Metagenomic engineering of the mammalian gut microbiome in situ. Nat Methods 2019;16:167–70. [CrossRef]
  86. Wang X, Maxwell KG, Wang K, Bowers DT, Flanders JA, Liu W, et al. A nanofibrous encapsulation device for safe delivery of insulin-producing cells to treat type 1 diabetes. Sci Transl Med 2021;13:eabb4601. [CrossRef]
  87. Park JS, Rhau B, Hermann A, McNally KA, Zhou C, Gong D, et al. Synthetic control of mammalian-cell motility by engineering chemotaxis to an orthogonal bioinert chemical signal. Proc Natl Acad Sci USA 2014;111:5896–901. [CrossRef]
  88. Nagai Y, Miyakawa N, Takuwa H, Hori Y, Oyama K, Ji B, et al. Deschloroclozapine, a potent and selective chemogenetic actuator enables rapid neuronal and behavioral modulations in mice and monkeys. Nat Neurosci 2020;23:1157–67. [CrossRef]
  89. Italiani P, Boraschi D. New Insights Into Tissue Macrophages: From Their Origin to the Development of Memory. Immune Netw 2015;15:167–76. [CrossRef]
  90. Wang SK, Cepko CL. Targeting Microglia to Treat Degenerative Eye Diseases. Front Immunol 2022;13. [CrossRef]
  91. Streeter MD, Rowan S, Ray J, et al. Generation and Characterization of Anti-Glucosepane Antibodies Enabling Direct Detection of Glucosepane in Retinal Tissue. ACS Chem Biol 2020;15(10):2655–2661; [CrossRef]
  92. Scheller L, Strittmatter T, Fuchs D, Bojar D, Fussenegger M. Generalized extracellular molecule sensor platform for programming cellular behavior. Nat Chem Biol 2018;14:723–9. [CrossRef]
  93. Seluanov A, Gladyshev VN, Vijg J, et al. Mechanisms of cancer resistance in long-lived mammals. Nat Rev Cancer 2018;18(7):433–441; [CrossRef]
  94. Samorajski T, Ordy JM, Rady-Reimer P. Lipofuscin pigment accumulation in the nervous system of aging mice. The Anatomical Record (1968) 160:555–573. [CrossRef]
  95. Brizzee KR, Johnson FA. Depth distribution of lipofuscin pigment in cerebral cortex of albino rat. Acta Neuropathol (1970) 16:205–219. [CrossRef]
  96. Yanai S, Endo S. Functional Aging in Male C57BL/6J Mice Across the Life-Span: A Systematic Behavioral Analysis of Motor, Emotional, and Memory Function to Define an Aging Phenotype. Frontiers in Aging Neuroscience 2021;13.
  97. Lutz CM, Osborne MA. Optimizing mouse models of neurodegenerative disorders: are therapeutics in sight? Future Neurology 2014;9(1):67–75; [CrossRef]
  98. Double KL, Dedov VN, Fedorow H, et al. The comparative biology of neuromelanin and lipofuscin in the human brain. Cell Mol Life Sci 2008;65(11):1669–1682; [CrossRef]
  99. Spampanato C, Feeney E, Li L, Cardone M, Lim J-A, Annunziata F, et al. Transcription factor EB (TFEB) is a new therapeutic target for Pompe disease. EMBO Mol Med 2013;5:691–706. [CrossRef]
  100. Abascal F, Harvey LMR, Mitchell E, Lawson ARJ, Lensing SV, Ellis P, et al. Somatic mutation landscapes at single-molecule resolution. Nature 2021;593:405–10. [CrossRef]
  101. Cortese FAB, Santostasi G. Whole-Body Induced Cell Turnover: A Proposed Intervention for Age-Related Damage and Associated Pathology. Rejuvenation Res 2016;19:322–36. [CrossRef]
  102. Zealley B, de Grey ADNJ. Strategies for Engineered Negligible Senescence. Gerontology 2013;59:183–9. [CrossRef]
  103. Yoon YG, Koob MD. Transformation of isolated mammalian mitochondria by bacterial conjugation. Nucleic Acids Res 2005;33:e139. [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

© 2024 MDPI (Basel, Switzerland) unless otherwise stated