Interplay between Mitophagy and Inflammasomes in Neurological

Mar 27, 2019 - Department of Neurology, University of Miami Miller School of Medicine , Miami , Florida 33136 , United States. ACS Chem. Neurosci. , A...
2 downloads 0 Views 1MB Size
Subscriber access provided by Bethel University

Review

Interplay between Mitophagy and inflammasome in Neurological Disorders Radhika Kesharwani, Deepaneeta Sarmah, Harpreet Kaur, Leela Mounika, Geetesh Verma, Veeresh Pabbala, Vignesh Kotian, Kiran Kalia, Anupom Borah, Kunjan R. Dave, Dileep R Yavagal, and Pallab Bhattacharya ACS Chem. Neurosci., Just Accepted Manuscript • DOI: 10.1021/acschemneuro.9b00117 • Publication Date (Web): 27 Mar 2019 Downloaded from http://pubs.acs.org on March 27, 2019

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

ACS Chemical Neuroscience

Interplay between Mitophagy and inflammasome in Neurological Disorders

2 3

Radhika Kesharwani1†, Deepaneeta Sarmah1†, Harpreet Kaur1, Leela Mounika1,

4

Geetesh Verma1, Veeresh Pabbala1, Vignesh Kotian1, Kiran Kalia1, Anupom Borah2,

5

Kunjan R Dave3, Dileep R Yavagal3 and Pallab Bhattacharya1*

6 7 8 9 10 11 12 13

1Department

of Pharmacology and Toxicology,National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India.2 Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India.3Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA.

†Authors

have equal contribution

14 15 16

*Address of correspondence:

17 18 19 20 21 22 23 24 25

Pallab Bhattacharya, Ph.D Assistant Professor, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad Gandhinagar-382355, Gujarat, India. Email: [email protected] [email protected] Phone: +91 79 66745555 Fax: +91 79 66745560

26 27 28 29 30 31 32 33 34 35 36 37

1 ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

List of Abbreviations

2

NLR - Nucleotide-binding oligomerization domain (NOD)-like receptors

3

ASC-Apoptosis speck-like protein containing a caspase recruitment domain (CARD)

4

NLRP3 – NLR family pyrin domain containing protein 3

5

NLRP1 - NLR family pyrin domain containing protein 1

6

NLRC4 - NLR family CARD domain containing protein 4

7

Atg– Autophagy related genes

8

ULK1 – Unc 51 like autophagy activating kinase 1

9

LC3 – Light chain kinase 3

10

GABARAP – GABA type A receptor associated protein

11

ROS – Reactive Oxygen Species

12

Bnip3 – BCL 2 interacting protein 3

13

FUNDC1 - FUN14 domain-containing protein 1

14

CK2 – Casein kinase 2

15

DRP1 – Dynamin related protein 1

16

OPA1 – Mitochondrial dynamin like GTPase

17

OMM – Outer mitochondrial membrane

18

IMM – Inner mitochondrial membrane

19

PINK1 - PTEN-induced putative kinase

20

CCCP – Carbonyl cyanide m-chlorophenyl hydrazone

21

MMP – Mitochondrial membrane potential

22

BAK - Bcl2 associated athano genes

23

PLEKHM1 - Pleckstrin homology domain containing protein family member 1

24

AMBRA – Autophagy and beclin 1 regulator 1

25

TAXBP1-Tax1-binding protein 1

26

SQSTM1-Sequestosome 1

27

NBR1-Neighbor of BRCA1 gene 1

28

PARL- Presenilin Associated Rhomboid Like

29

PTEN-L-Phosphatase and tensin homolog-L 2 ACS Paragon Plus Environment

Page 2 of 40

Page 3 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1

ASC-Apoptosis-Associated Speck-like Protein

2

CIITA-Class II Major Histocompatibility Complex Transactivator

3

IPAF-ICE-Protease Activating Factor

4

NAIP-NLR Family Apoptosis Inhibitory Protein

5

CARD-Caspase recruitment domains

6

ASIC- Acid-sensing ion channel 1

7

AMPK- AMP-activated protein kinase

8

MAPK- Mitogen-activated protein kinase

9

NFKB- Nuclear factor kappa-light-chain-enhancer of activated B cells)

10

Nrf2- Nuclear factor (erythroid-derived 2)-like 2

11

MFF- MITOCHONDRIAL FISSION FACTOR;

12

Trx1-Thioredoxin-1

13

APC-Antigen presenting cell

14

NR4A1- Nuclear Receptor Subfamily 4 Group A Member 1

15

JNK - C-Jun N-terminal kinase

16

TLR4-Toll-like receptor-4

17

ANSCs- Adult neural stem cells

18

SVZ-Sub-ventricular zone

19

PPAR- Peroxisome proliferator-activated receptors

20

PGC1-α- Peroxisome proliferator-activated receptor gamma coactivator 1-alpha

21

DFCP1- Double FYVE-containing protein 1

22

PTM-Post-translational modifications

23

TBK1- Serine/Threonine-Protein Kinase-1

24

VCP-Valosin-containing protein

25

HLA-Human leukocyte antigen

26

MttHtt- Mutant huntigtin gene

27

CCL4- C-C motif chemokine 4

28

IGF-1- Insulin-like growth factor 1

29

HMGB-1- High mobility group box 1 protein 3 ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

PGF-2α - Prostaglandin F2α

2

I-FABP- Fatty Acid Binding Protein 1

3

DAMP- Damage-associated molecular patterns

4

PAMP-Pathogen-associated molecular patterns

5

Wdfy3- WD Repeat and FYVE Domain Containing 3

6

NOS- Nitric oxide synthase

7

4 ACS Paragon Plus Environment

Page 4 of 40

Page 5 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1

Abstract

2

Mitophagy and inflammasome have a pivotal role in the development of

3

neuropathology. Molecular mechanisms behind mitophagy and inflammasome are

4

well understood, but lacunae prevail in understanding the crosstalk between them in

5

various neurological disorders. As mitochondrial dysfunction is the prime event in

6

neurodegeneration, the clearance of impaired mitochondria is one of the main task for

7

maintaining cell integrity in majority of neuropathologies. Along with it, inflammasome

8

activation also plays a major role which is usually followed by mitochondrial

9

dysfunction. The present review highlight basics of autophagy, mitophagy and

10

inflammasome, the molecular mechanisms involved and more importantly it tries to

11

elaborate the interplay between mitophagy and inflammasome in various neurological

12

disorders. This will help in upgrading the reader’s understanding in exploring the link

13

between mitophagy and inflammasome that has been dealt with limitations in past

14

studies.

15 16

Keywords: Cerebral ischemia, mitophagy, inflammasome, neurodegenerative

17

disorder, neuroinflammation

18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 5 ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 40

1

1. Introduction

2

The genesis of neurological disorders and their symptoms are different but at the

3

molecular level they have common attributes such as oxidative damage, aggregation

4

of misfolded proteins, mitochondrial dysfunction, impairment of autophagy, and

5

neuroinflammation1, 2. Impaired autophagy and neuroinflammation are the foremost

6

events involved in several neurological disorders3, 4. Misfolded proteins, such as α-

7

synuclein in Parkinson’s disease and amyloid β in Alzheimer’s disease, along with

8

oxidative stress play central role in the progression of neurodegeneration1. This

9

promotes activation of innate immune system to upregulate signaling of inflammasome

10

complexes (NLRP3, NLRP1 and NLRC4) in the microglia for neuroinflammation.

11

These mechanisms in some instances are also activated within the astrocytes and

12

neurons that aggravates the disease5. In healthy cells, factors like nutrient deprivation,

13

enhanced calcium influx and oxidative stress can lead to mitochondrial dysfunction6,

14

7.

15

known as mitophagy. This process of mitophagy gets dysregulated in neurological

16

diseases8.

17

neuroinflammation and mitophagy, hence keeping this in mind, the present review

18

provides a brief overview of mitophagy and inflammasome, their mechanisms, and

19

their associated cross talk in various neurological disorders.

Impaired mitochondria can be eliminated via specialized process of autophagy Recent

studies

report

that

there

is

a

connection

between

20 21

2. Autophagy and Mitophagy

22

Autophagy or self-eating is the process of degradation of impaired intracellular

23

components which is mediated by autophagy related proteins (Atg) and lysosomes 9.

24

Autophagy can be categorized into three types macroautophagy, microautophagy and

25

chaperon-mediated autophagy

26

comprises of five steps a) initiation of isolation membrane, b) elongation of isolation

27

membrane, c) formation of autophagosome, d) fusion of autophagosome and

28

lysosome and e) degradation of auto-phagolysosome 8. Autophagosome biogenesis

29

is regulated by unc51 like autophagy acting kinase 1 (ULK1), autophagy related

30

protein complex Atg16-Atg5-Atg12 and Atg 8 family proteins, while biogenesis and

31

maturation of autophagosome is regulated by microtubule-associated protein 1 light

32

chain 3 (LC3) and gamma-aminobutyric acid type 1 receptor (GABARAP). Soluble N-

33

ethylmaleimide sensitive factor (NSF) attachment protein receptor (SNARE) proteins

34

help in the fusion of autophagosome to lysosome and then the cargo is degraded by

10.

Mechanistically, the process of autophagy

6 ACS Paragon Plus Environment

Page 7 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

11.

1

the lysosomal hydrolases

2

well as cargo selective. Non-selective autophagy plays major role during nutrient

3

deprivation in order to recycle essential nutrients from the cell, while cargo-selective

4

autophagy accounts for the removal of damaged organelles and aggregation prone

5

proteins that can be toxic for the cell 12. Cargo selective autophagy can be of various

6

types depending on the target such as aggrephagy (aggregated proteins), mitophagy

7

(mitochondria), ribophagy (ribosomes), peroxyphagy (peroxisomes), reticulophagy

8

(endoplasmic reticulum) and xenophagy (pathogens).

9

Selective autophagy of mitochondria is called mitophagy. It is a cellular quality control

10

process to maintain mitochondria’s integrity and its functional state8. The mechanism

11

of mitophagy was first identified in yeast and Uth1p gene was found to be an important

12

regulator of mitophagy

13

mitochondrial depolarisation favours sequestration of damaged mitochondria inside

14

the autophagosome which is followed by its fusion with lysosome that ultimately leads

15

to its degradation

16

promotes the clearance of damaged mitochondria which is responsible for the

17

generation of reactive oxygen species (ROS) as well as proapoptotic signals and

18

depletion of ATP

19

event associated with the development of pathology of several neurological ailments

20

like Parkinson’s disease, cerebral ischemia, Alzheimer’s disease and Amyotrophic

21

Lateral Sclerosis. Therefore, removal of damaged mitochondria is a prime requirement

22

for halting progression of neuropathology. Figure 1 illustrates the molecular

23

mechanisms behind mitophagy.

14.

15.

13.

Macroautophagy or autophagy can be non-selective as

Studies carried out in mammalian cells suggest that

Mitophagy plays central role in preventing cell death, as it

It is highly accepted that mitochondrial dysfunction is a major

7 ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1 2

Figure 1: Illustration representing overview of molecular mechanisms behind

3

mitophagy

4

1) Mitochondrial depolarization followed by opening of mPTP caused by increased

5

levels of ROS 2) Recruitment of PINK1 from the cytosol to OMM due to mitochondrial

6

depolarization 3) Recruitment of Parkin to OMM induced by PINK1 4) Ubiquitination

7

of adaptor proteins (OPTN, p62, SQSTM1 and TAXBP1) present on OMM induced by

8

parkin 5) Tethering of ubiquitinated adaptor proteins with AMBRA1, a component of

9

autophagosomal membrane via LIR-binding domain 6) Fusion of autophagosome and

10

lysosome via interaction between HOPS and PLEKHM1 component of AP and Rab7

11

which is a component of lysosome 7) Degradation of damaged or superfluous

12

mitochondria via mitophagy.

13 14

3 Molecular mechanisms of mitophagy

15

3.1 Receptor mediated mitophagy

16

A) Bnip3/Nix: Bnip3 (BCL2/adenovirus E1B 19 kDa interacting protein) and Nix

17

(known as BNIP3L, a homolog of Bnip3) are pro-apoptotic mitochondrial proteins

18

where the former has Bcl-2 homology-3 domain (BH3) 16. The BH3 domain facilitates 8 ACS Paragon Plus Environment

Page 8 of 40

Page 9 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1

entry of Bnip3 to the outer mitochondrial membrane (OMM), which can be inhibited by

2

phosphorylation at C-terminus due to diminished interaction between bnip3 and OPA1

3

(a mitochondrial fusion regulator protein)17. Bnip3 regulates mitophagy during hypoxia

4

while

5

Bnip3/Nix is reported to be expressed in liver, kidney, brain, smooth muscles of

6

healthy cells but it is not present ubiquitously

7

many neurological disorders that modulates function of Bnip3/Nix

8

conditions promote attachment of hypoxia-inducible factor 1 (HIF-1) to the promoter

9

site of Bnip3/Nix, increasing expression of the same leading to mitophagy and thus

Nix regulates mitophagy during the development of erythroid lineage 19.

18.

Hypoxia is one of the key factors in

22,23.

20.

Hypoxic

10

reduces levels of ROS and mitigates apoptosis21,

11

Bnip3 also leads to enhanced mitophagy and thus cell death 24. Reports suggest that

12

Bnip3 is upregulated significantly in a delayed manner following cerebral ischemia in

13

neonatal stroke model, which is said to be responsible for cell death. It was observed

14

that Bnip3 gene silencing was able to prevent neuronal cell death following cerebral

15

ischemia 24.

16

B) FUNDC1 (FUN14 domain-containing protein 1): FUNDC1 is a protein present on

17

the OMM and is another important player of mitophagy

18

components within its structure, a 3-transmembrane domain and a N-terminal

19

cytosolic LIR-motif which binds with LC3 and GABARAP 25. It regulates autophagy via

20

recruitment of LC3 to mitochondria and by regulating mitochondrial fusion and fission

21

proteins, OPA1 and dynamin related protein 1 (DRP1) respectively 25, 26. Under normal

22

conditions, FUNDC1 is inhibited by negative phosphorylation by Src kinase at tyr18

23

and by casein kinase (CK2) at ser13 25,27. Hypoxic conditions or mitochondrial damage

24

activates PGAM5, which in turn activates CK2 and thus promotes phosphorylation.

25

LC3 then binds to damaged mitochondria, which then undergoes mitophagy

26

Recently it was reported that activation of Bcl-xl inhibits PGAM5 which leads to

27

inhibition of mitophagy28.

28

C) Prohibitin 2 (PHB2): Although the role of OMM proteins has been characterized

29

well during mitophagy, still extensive research is required to understand components

30

of inner mitochondrial membrane (IMM) that are involved in mitophagy. Recently,

31

PHB2 was characterized as a novel IMM protein and acts as a mitophagy receptor for

32

Parkin-mediated mitophagy in mammalian cells

33

rupture of OMM, that is achieved by PARK2 mediated ubiquitination and degradation

34

of OMM proteins followed by expression of PHB2 on OMM which further facilitates

29.

But excessive activation of

25.

It has two important

27.

Mitochondrial damage leads to

9 ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

30.

Page 10 of 40

1

binding of LC3 leading to mitophagy

2

achieved by USP14 inhibition. Results from co-immunoprecipitation assay showed

3

that interaction of PHB2 with LC3 depends on the activity of PARK2

4

it has been found that PHB2 does not interact with the GABA-type A receptor-

5

associated protein (GABARAP) and GABARAP-L2. As ubiquitin proteasome system

6

(UPS) is impaired in several neurodegenerative disorders, UPS may be targeted for

7

therapeutic purposes. Many de-ubiquitinating (DUB) enzymes are being explored.

8

USP14 is one of the DUB enzymes which regulates UPS negatively

9

supports that inhibition of USP14 leads to enhanced mitochondrial clearance. It also

10

Mitochondrial membrane rupture can be 31.

Interestingly,

32.

Studies

induces OMM rupture followed by interaction of PHB2-LC3 leading to mitophagy 30.

11 12

3.2 Parkin-PINK dependent mitophagy

13

PINK1 is PTEN-induced putative kinase which belongs to a family of Ser/Thr kinase

14

and thereby having kinase activity along with mitochondrial target sequence and it also

15

acts as a sensor for detection of polarized state of mitochondria 33, 34. While Parkin is

16

a E3-Ubiquitin ligase, PINK1 acts as an upstream regulator of PARKIN. This is

17

supported by a study in which it was found that PINK1-KO phenotype was rescued

18

with the overexpression of PARKIN but overexpression of PINK1 was not able to

19

rescue the PARKIN-KO phenotype in drosophila

20

mitochondria having proteins containing mitochondrial target sequence (MTS) and

21

single membrane spanning domain (SMSD) get entry into IMM through the

22

translocase of outer mitochondrial membrane (TOM) and translocase of inner

23

mitochondrial membrane (TIM) complex. Studies have reported two forms of PINK1,

24

a 64-kDa Full length PINK1 and a 52-kDa PINK137. Under normal conditions, the

25

former is present predominantly that enters the IMM by cleavage of MTS and is

26

converted into 60-kDa form. This is further cleaved by PARL (Presenilin associated

27

Rhomboid like protein) leading to formation of a 52-kDa form which is rapidly

28

processed and degraded by proteases and thus maintains the levels of PINK1 low and

29

inhibits mitophagy in healthy mitochondria 37, 38.

30

Under pathological conditions, mitochondria undergoes depolarization. To mimic the

31

same physiological conditions, studies have been carried out by using Carbonyl

32

cyanide m-chlorophenyl hydrazone (CCCP), which is a mitochondrial uncoupler39.

33

CCCP drastically reduces mitochondrial membrane potential (MMP), which blocks

34

transport of PINK1 to IMM and thus PINK1 will not be processed by PARL leading to

35, 36.

10 ACS Paragon Plus Environment

In healthy cells, polarized

Page 11 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1

accumulation of the 60-kDa form of PINK1 followed by recruitment of PARKIN37, 40.

2

This recruitment of PARKIN facilitates ubiquitination of OMM adaptor proteins like p62,

3

optineurin, SQSTM1, NBR1, TAXBP1 and this ubiquitination solely depends on the

4

TBK1 a Ser/Thrkinase that promotes phosphorylation of RAB7A, an activator of

5

mitophagy33,

6

autophagosome through their LIR motif (LC3-interacting region) to induce mitophagy

7

and promotes biogenesis of autophagosome via binding with AMBRA1 (an autophagy

8

promoting protein), followed by fusion of autophagosome with the lysosome via

9

autophagosomal proteins PLEKHM1 and HOPS and lysosomal protein-Rab7 and

39, 41.

These adaptor proteins followed by ubiquitination

42, 43.

targets

10

promotes mitophagy

11

was found to promote mitophagy independent of PINK1/PARKIN, via activation of

12

IKKα which in turn phosphorylates AMBRA, an activator of mitophagy

13

studies have suggested the role of PARKIN in apoptosis, as Parkin inhibits BAK, a

14

pro-apoptotic factor and thereby inhibits apoptosis and promotes clearance of

15

damaged mitochondria. Along with this it also promotes clearance of apoptotic

16

mitochondria and restricts their potential pro-inflammatory effect 44.

17

Mitochondrial outer membrane Rho GTPases Miro1/2 (Miro) are the adaptor proteins

18

that anchors mitochondria to motor proteins, which results in mitochondrial arrest

19

facilitating mitophagy. In healthy cells, Miro interacts with Parkin independent of

20

PINK1. However damaged mitochondria disrupt pool of Miro-Parkin due to

21

overexpression of PINK1 leading to ubiquitination and degradation of Miro and thus

22

impairs mitophagy

23

homolog-long (PTEN-L) prevents PINK1/PARKIN mediated mitophagy by preventing

24

translocation of Parkin as well as promotes phosphorylation of ubiquitin 45.

44.

Recently a novel ligase, identified as HUWE1 E3 ligase, 43.

Recent

Recently it was been identified that phosphatase and tensin

25 26

3. Inflammasomes

27

Inflammasomes are cytosolic complexes composed of three chief components:

28

sensor-NLRs, adaptor-ASC, effectors-pro-caspase1

29

types: canonical and non-canonical which activates caspase-1 and caspase-4,5,11

30

respectively 48-50. Innate immune response is the first line of immune defence against

31

invading pathogens and endogenous danger signals by pattern recognition receptors

32

(PRRs) through sensing the pathogen- and danger- associated molecular patterns

33

(PAMPs and DAMPs) 51, 52. Dendritic cells, microglia, astrocytes, neutrophils, epithelial

34

cells, macrophages and monocytes express huge amount of PRRs. PRRs are of

46, 47.

11 ACS Paragon Plus Environment

Inflammasomes are of two

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 40

1

various types including members of Toll-like receptors (TLRs), C-type lectins (CTLs),

2

nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) or absent in

3

melanoma 2 (AIM2)-like receptors (ALRs). TLRs are present on the extracellular

4

membranes, while NLRs are present in the cytosol. NLRs are mainly responsible for

5

formation of inflammasomes

6

NODs (NOD1, 2, NOD3/NLRC3, NOD4/NLRC5, NOD5/NLRX1, CIITA), NLRPs

7

(NLRP1–14,also called the NALPs) and ICE protein-activating factor (IPAF; NLRC4

8

and NAIP) 5. NLRs consists of a tripartite structure including C-terminal leucine-rich

9

repeats (LRRs) domain, NACHT or NOD (nucleotide-binding-oligomerization domain)

10

an intermediate domain and N-terminal effector domain can be pyrin domain (PYD) or

11

caspase recruitment domain (CARD)54. After sensing a stimuli, NLR undergoes

12

oligomerization by the interaction between their NACHT domains, then the

13

oligomerized NLR interacts with ASC through its PYD domain, followed by recruitment

14

of procaspase-1 by ASC via interaction of CARD/CARD. This is followed by cleavage

15

and activation of pro-caspase-1 to caspase-1, which further stimulates the maturation

16

of pro-inflammatory cytokines pro-interleukin 1β (IL-1β) and pro-IL-18 to active IL-1β

17

and IL-18

18

Gasdermin-D leading to pyroptosis and release of cytokines 56, 57. Indeed activation of

19

inflammasome is very essential in host-defence mechanism, although excessive

20

activation can damage the host cells. Recent study suggests that this excessive

21

inhibition is kept in check by Cullin1 (CUL1) component of the Skp1-Cullin1-F-box E3

22

ligase which binds with NRPL3 and promotes ubiquitination, not to degrade the protein

23

but to prevent activation of inflammasome

24

mitochondrial

25

inflammatory cytokines can also trigger mitochondrial damage60. So, it is quite

26

ambiguous that what comes first.

55.

53.

There are three subfamilies within the NLR family-

This is followed by maturation of cytokines and promotes cleavage of

damage

promote

activation

58.

of

Several studies suggest that inflammasomes59.

However,

27 28 29

4. Interplay between mitophagy and inflammasomes in neurological diseases A) Cerebral Ischemia

30

Activation of inflammasome is one of the events involved in cerebral ischemia5, 61. First

31

evidence of involvement of NLRP1 inflammasome in cerebral ischemia was reported

32

by Abulafia et al

33

receptor pyrin domain-containing 3 (NLRP3) inflammasome is reported to be

34

associated in cerebral ischemia. Apart from this, NLRP1, NLRC4, AIM2 are also

62.

The nucleotide binding domain leucine-rich repeat-containing

12 ACS Paragon Plus Environment

Page 13 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1

involved in the inflammatory cascade in cerebral ischemia5. In cerebral ischemia levels

2

of ROS are elevated leading to NLRP3 inflammasome activation, which results in

3

release of IL-1β and IL-18 causing neuroinflammation3, 5, 63. NLRP1 is reported to be

4

activated in response to acid sensing ion channel (ASIC) activation, which further

5

release IL-1β and IL-18 and aggravate neuroinflammation 5. ATP depletion also

6

stimulates the activation of NLRP1 inflammasome via AMP-activated protein kinase

7

(AMPK). Nuclear factor kappa B (NFκB) and mitogen-activated factor (MAPK) lowers

8

the expression and activation of NLRP1 and NLRP3 inflammasomes under ischemic

9

condition 64.

10

NLRP3, NLRP1, NLRC4 and AIM2 inflammasomes are expressed and activated in

11

microglia and exacerbate the ischemic condition

12

inflammasome complex regulates the inflammatory response, apoptotic and pyroptotic

13

death of microglial cells under ischemic condition

14

suppresses the activation of inflammasomes post ischemia

15

nicotine worsens cerebral ischemic condition by stimulating the inflammasome

16

activation via inhibition of ER-β signaling in post ischemic brain

17

that nuclear factor erythroid 2-related factor 2 (Nrf2) inhibits the activation of NLRP3

18

inflammasome by regulating the thioredoxin 1 (Trx1)/ thioredoxin interacting protein

19

(TXNIP) complex in the model of cerebral ischemia. It is suggested that under

20

oxidative stress conditions, Trx1/TXNIP complex dissociates and release TXNIP

21

which further stimulates the activation of NLRP3 inflammasome and Nrf2 acts as an

22

antioxidant agent which plays a protective role in the animal model of cerebral

23

ischemia 67.

24

Cerebral ischemia involves depletion in ATP and increase in AMP due to mitochondrial

25

dysfunction leading to

26

mitochondrial fission factor (MFF) which activates DRP1 and thus stimulates

27

mitochondrial fission and mitophagy

28

during reticulocyte maturation and in cerebral ischemia reperfusion-induced

29

mitophagy 69, 70. Acidic post-conditioning (APC) is a condition in which CO2 is inhaled

30

during reperfusion after cerebral ischemia leading to mild acidosis. It is reported that

31

APC maintains the mitochondrial membrane potential

32

sensor present in the mitochondria of neuronal cells which reduces the mitochondrial

33

membrane potential following oxidative stress 72. Findings suggest that APC alleviates

34

cerebral I/R injury via activation of mitophagy by recruiting PARK2 in mitochondria in

65.

65.

In addition to this, NLRC4 Estrogen receptor β (ER-β) 66.

66.

It is reported that It is also reported

activation of AMPK, promoting phosphorylation of 7, 68.

BNIP3L is mainly involved in mitophagy

13 ACS Paragon Plus Environment

71.

ASIC1 is a main proton

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

73.

Page 14 of 40

Peroxynitrite (ONOO-) is the main marker of

1

a mouse model of cerebral ischemia

2

reactive nitrogen species which promotes nitration of proteins. This further activates

3

mitochondrial fission and thus activates mitophagy

4

group A member 1 (NR4A1) is involved in regulating mitochondrial function. It is

5

reported that NR4A1 exacerbates the cerebral I/R injury by reducing the expression

6

of Mfn2 through MAPK-ERK-CREB signaling pathway

7

reperfusion injury the PINK1/Parkin/p62 pathway is activated to induce mitophagy

8

(Figure 2).

74.

Nuclear receptor subfamily 4

75.

In cerebral ischemia/ 76

9 10

Figure

2:

Illustration

representing

interplay

11

inflammasome in cerebral ischemia: Process of cerebral ischemia involves multiple

12

signalling pathways that activate or inhibit mitophagy (1-6) and inflammasome (7). 1)

13

Hypoxia occurring during cerebral ischemia increase levels of Bnip3/Nix via activation

14

of HIF-1, followed by activation of Beclin-1 which ultimately induces mitophagy 2)

15

Reperfusion injury following cerebral ischemia can produce ROS which also activates

16

mitophagy via PINK1/Parkin activation. 3) Decreased levels of ATP and increased

17

levels of AMP following mitochondrial dysfunction activates AMPK followed by fission 14 ACS Paragon Plus Environment

between

mitophagy

and

Page 15 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1

of mitochondria due to activation of MFF and Drp1, a mitochondrial fission protein and

2

thus promotes mitophagy 4) Mitochondrial fission is also promoted by nitration of

3

proteins due to generation of RNS which activates mitophagy 5) Mitophagy is also

4

promoted by increased levels of LC3II, p62 and Beclin1 6) Acidic post-conditioning

5

(APC) activates PARK2 during ischemic condition and thus facilitates mitophagy 7)

6

Increased levels of ROS and activation of AMPK activates NLRP3 and NLRP1

7

respectively, which in turn activates Il-1β, IL-18 via activation of caspase-1 and

8

promotes neuroinflammation.

9

B) Parkinson’s disease (PD)

10 11

Oxidative stress and activation of both microglial cells and inflammasomes play a key

12

role in the pathology of PD as they promote neuroinflammation and neurodegeneration

13

77

14

which in turn activates c-Abl kinase which facilitates activation of NLRP3

15

inflammasomes

16

microglial cells which can also activate inflammasomes 79. Neurotoxins like rotenone,

17

MPTP, paraquat, dieldrin, manganese, salolinon, etc. increase levels of H2O2 which

18

activates JNK MAPK and subsequently activate caspase-1 and 3 leading to cell death

19

and promotes induction of PD

20

of α-synuclein via activation of inflammasome 81.

21

Crosstalk between caspase-1 and NLRP3 have been suggested by studies which

22

support that α-synuclein promotes activation of TLR2 which facilitates formation of

23

NLRP3 assembly and ultimately promotes synthesis of IL-1ß

24

levels of IL-1ß promotes generation of ROS and also promotes release of cathepsin-

25

B which finally upregulates NLRP3

26

synuclein also promotes activation of TLR4/NF-kB and NLRP3/caspase-1 signaling

27

mechanism in adult neural stem cells (ANSCs), which was confirmed by knockdown

28

of either caspase-1 or NLRP3 which inhibited proliferation of ANSCs

29

reported that miRNA-7 activates neurogenesis in subventricular zone (SVZ) via

30

inhibition of NLRP3/caspase-1 signalling mechanism in ANSCs 84.

31

Recently, researchers found that there was increased levels of NLRP3 within

32

mesencephalic neurons of PD patients

33

inflammation and death of dopaminergic neurons in which they have shown that

34

inhibition of hepatic inflammasomes by siNLRP3 (silencing RNA) leads to decreased

(Figure 3). More specifically, generation of ROS leads to increased oxidative stress 78.

ROS

also increases the activity of cathepsin-B in activated

80.

Activation of caspase-1 also promotes aggregation

79, 83.

82,83.

The increased

Furthermore, it has been reported that α-

85.

84.

It is also

Studies have described the role of liver

15 ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 40

1

spreading of inflammatory cytokines to brain and thus halts the progression of

2

neuroinflammation and also the degeneration of dopaminergic neurons

3

Mitochondrial serine protease HtrA2 inhibits activation of NLRP3 and AIM2

4

inflammasomes which was confirmed by disruption of HtrA2 activity leading to

5

upregulation in NLRP3 and AIM2 inflammasomes in macrophages

6

demonstrated that MPTP promotes impairment in autophagy via induction of

7

PI3K/Akt/mTOR pathway which was confirmed by assessing levels of Beclin-1 and

8

LC3. Levels of which was found to be decreased 88. Many PPAR ß/delta agonists are

9

reported to have anti-inflammatory properties but mechanisms are yet to be explored

87.

86.

It has been

10

89.

11

agonist and it was found that it protected dopaminergic neurons in the midbrain,

12

increased levels of dopamine in the striatum and also inhibited activation of NLRP3

13

inflammasomes in astrocytes in a MPTP-mouse model of PD 89.

14

ß-arrestin-2 mediated dopamine D2-receptor (Drd2) signaling mechanism is

15

considered as potential anti-inflammatory target. In one study they used Drd2 agonist

16

and they found that it suppressed activation of NLRP3 inflammasome and also

17

decreased levels of caspase-1 and IL-1ß in MPTP-induced mouse model

18

and PARKIN are the widely-studied protein related to PD. PINK1-PARKIN pathway is

19

considered as most important pathway related to mitophagy

20

of mitochondria, mitochondrial rhomboid protease (PRAL) instead of PGAM5,

21

(phosphoglycerate mutase which promotes mitochondrial division by phosphorylation

22

of Drp1) cleaves PINK1 which leads to accumulation of PINK1 in the OMM

92.

23

Deficiency of PGAM5 leads to development of PD-like movement disorder

93.

24

Functions of PGAM5 are regulated by Syntaxin 17 which is a novel mitochondrial

25

protein mediating normal division of mitochondria under healthy conditions but upon

26

stimulation of mitophagy, syntaxin 17 detaches from Drp1 and binds with Atg14L

27

promoting formation of autophagosomes and also promotes elongation of

28

mitochondria. Upon mitophagy, cleaved PGAM5 promotes dephosphorylation of

29

FUNDC1 which promotes excessive breakdown of mitochondria

30

fusion is mediated by mitofusins and Opa1, while fission is mediated by Drp1

31

Mitochondrial fusion and fission are important for maintaining mitochondrial

32

homeostasis which is mediated by PINK1 and PARKIN, which was confirmed by one

33

study in which it was found that mitofusin (Mfn) is the mitochondrial fusion protein

34

which is ubiquitinated by PINK1 and PARKIN. Knockdown of Mfn1 and Mfn2 results

In a study researchers used GW501516, a selective and high-affinity PPAR ß/delta

16 ACS Paragon Plus Environment

91.

90.

PINK1

Upon depolarization

94.

Mitochondrial 95, 96.

Page 17 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1

in altered MMP and inhibits recruitment of Parkin on OMM 97. Recently, it was identified

2

that retro-translocation of Mfn2 promotes detachment of mitochondria from ER

3

thereby, accelerates mitophagy

4

extract (GSE) preserves mitochondrial function, improved function of flight muscle,

5

prolonged life span via activation of mitophagy in PINK1 mutant drosophila model of

6

PD 99. Rotenone , a neurotoxin , used to develop model of PD induces autophagy and

7

mitophagy in PC12 cells100. Rotenone inhibits mitochondrial biogenesis and reduces

8

the mitochondrial membrane potential. PINK1 inhibits mitochondrial biogenesis by

9

inhibiting PGC1-α, which in turn inhibits PINK1/PARKIN protein and ultimately inhibits

10

mitophagy. Which indicates that mutual antagonism exists between PINK1/PARKIN

11

and PGC1-α.This maintains mitochondrial homeostasis in rotenone-induced

12

neurotoxicity 101.

98.

More recently, it was discovered that grape skin

13 14

Figure

3:

Illustration

15

inflammasome in Parkinson’s disease: Mitochondrial dysfunction in PD arises as a

16

result of exposure to neurotoxins. A) Increased ROS due to mitochondrial dysfunction

17

activates c-Abl and cathepsin B which further activates NLRP3 inflammasome. This

18

then

activates

Il-1β

representing

via

activation

interplay

of

between

caspase-1/3

17 ACS Paragon Plus Environment

and

mitophagy

finally

and

promotes

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

neuroinflammation B) Increased levels of ROS as well as activation of caspase-1/3

2

induces the misfolding of wild-type α-synuclein to toxic α-synuclein which promotes

3

extensive degradation of mitochondria, promotes cleavage of PGAM5 instead of

4

PINK1 via activation of PARL. Further the cleaved PGAM5 dephosphorylates

5

FUNDC1 resulting into extensive mitochondrial breakdown C) Exposure to

6

neurotoxins also generates H2O2 which activates Jnk-MAPK pathway promoting

7

neuroinflammation via activation of caspase-1/3.

8

C) Alzheimer ’s disease

9

Inflammation plays a major role in the pathogenesis of AD, which is supported by

10

studies indicating enhanced levels of inflammatory cytokines in the brain sample of

11

AD patients

12

pathogenesis of AD, as a study found that NLRP3 enhances accumulation of Aβ

13

protein

14

overload of neuritic plaque in transgenic mice model of AD 104. Priming of microglia by

15

β-amyloid pathology resulted in activation of IL-1β leading to significant cognitive

16

decline as compared with wild type LPS-treated mice. But it was reported that priming

17

of microglia does not activate IL-1β or NFκB p65 nuclear translocation. But upon

18

secondary stimulation, it activates the IL-1β and also p62 nuclear translocation, from

19

this it was confirmed that secondary inflammatory insults induces cognitive

20

impairments specifically in in-vivo models 105.

21

Glial maturation factor (GMF) is a pro-inflammatory protein which activates glial cells

22

to stimulate neuroinflammation and neurodegeneration in AD. GMF is mainly present

23

and expressed in proximity to Aβ and tau in the temporal cortex of human AD brain.

24

Upon prolonged accumulation and aggregation of Aβ peptide, glial cells are activated

25

and secrete pro inflammatory cytokines. GMF enhances the activation of NLRP3

26

inflammasome and promotes neuroinflammation in AD 106. Although the link between

27

inflammasome and mitophagy in AD is yet not clearly understood, a link may be

28

supported by the study which suggests that mitophagy is impaired in AD resulting in

29

increased oxidative stress and deficiency of cellular energy triggering the

30

accumulation of Aβ peptide and tau protein, which further impairs mitophagy and

31

activates NLRP3 inflammasome

32

potential (MMP) due to which recruitment of PINK1/Parkin in the outer membrane of

33

dysfunctional mitochondria is halted 108. Accumulation of tau protein impairs mitophagy

34

by inhibiting the recruitment of Parkin in dysfunctional mitochondria by sequestering it

103.

102.

NLRP3 inflammasome was also found to be important in the

It is also reported that absence of NLRP3 inflammasome reduces the

107.

Tau protein increases mitochondrial membrane

18 ACS Paragon Plus Environment

Page 18 of 40

Page 19 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

109.

1

in cytosol

Post-translational modifications (PTMs) in the tau protein leads to

2

impairment of mitophagy and this idea came after a study which suggests that NH2-

3

truncated tau protein promotes aberrant recruitment of Parkin and UCHL-1 modulator

4

of mitophagy which promotes deregulation of mitophagy 110. Aging is a natural process

5

which is associated with inflammation. This process is known as inflammaging. This

6

is mediated by inflammasome activation and is associated with the development of

7

deleterious conditions in the aging brain such as Alzheimer’s disease 111.

8 9

Fig 4: Illustration representing interplay between mitophagy and inflammasome

10

in Alzheimer’s disease: Mitochondrial dysfunction is the main pathological event

11

during AD which leads to accumulation of damaged mitochondria, which in turn

12

triggers production of ROS promoting accumulation of Aβ and Tau-proteins A) This in

13

turn activates microglia via release of glial maturation factor (GMF), followed by

14

activation of NLRP3-inflammasome which further promotes neuroinflammation. B)

15

Accumulation of Aβ and Tau-protein also impairs mitophagy by inhibiting recruitment

16

of PINK1/Parkin via two ways: either by increasing MMP or by directly promoting

17

sequestration of PINK1/Parkin in the cytosol.

18 19 ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 40

D) Amyotrophic Lateral Sclerosis

1 2

Amyotrophic lateral sclerosis (ALS) is a motor neurodegenerative disease involving

3

degeneration of both upper and lower motor neurons leading to motor and extra-motor

4

symptoms 112,113. Mitochondrial dysfunction and autophagy plays an important role in

5

ALS 114. Optineurin is an autophagy receptor that is recruited to the ubiquitinated OMM

6

proteins via its ubiquitin binding domain. On the other side it consists of light chain 3

7

(LC3) binding domain (LIR) which binds to LC3 for autophagosome formation favoured

8

by assistance of parkin 41. Optineurin momentarily binds to dysfunctional mitochondria

9

and allows the recruitment of double FYVE-containing protein 1 (DFCP1) for the

10

initiation of autophagosome formation and microtubule-associated protein LC3

11

recruitment 41.

12

In ALS there is mutation of optineurin E478G, as a result of which mitophagy is

13

subdued which exacerbates the pathology 41. Along with optineurin, ALS also involves

14

mutation of TANK-binding kinase 1 (TBK1) which phosphorylates optineurin in its

15

serine 177 residue and stimulates its binding to Atg8/LC3 and autophagic clearance

16

115.

17

required for binding of ubiquitin to LC3, which binds to LC3 via its LIR region. Studies

18

suggest that ALS-associated mutation in LIR region i.e. D337E, L341V reduces the

19

ability of binding of SQSTM1/p62 to LC3

20

containing protein (VCP) in the damaged outer mitochondrial membrane depends on

21

Parkin-mediated ubiquitination of damaged mitochondria 117. VCP undergoes mutation

22

during ALS, which leads to impairment of mitophagy 118.

23

Fibroblasts of ALS patients express C9orf72, which assists in protein synthesis,

24

undergoes mutation. This interacts with ULK1 and initiates autophagy

25

between inflammation and ALS was first established by Troost et. al., where they

26

found that brain of sporadic ALS patients expressed higher levels of MHC (major

27

histocompatibility complex) and HLA (human leucocyte antigens), which are important

28

components of inflammation

29

inflammation which were found to be activated in ALS-linked mutant SOD1 mice

30

Apart from these, reactive oxygen and nitrogen species were also found to be

31

upregulated. These reactive nitrogen are then converted into peroxynitrite leading to

32

protein nitration, which triggers the activation of caspase-1/inflammasome

33

involves accumulation of mutant superoxide dismutase 1 (SOD1)

34

sensed by ASC component of inflammasome as a result of which there is activation

Sequestosome 1 (SQSTM1) encodes for SQSTM1/p62 which is an adaptor protein

120.

116

(Figure 5). Localization of valosin-

119.

The link

NFкB and TLRs are important component of

20 ACS Paragon Plus Environment

123,

122.

121.

ALS

which can be

Page 21 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

124.

1

of caspase-1 and IL-1β in microglia which leads to neuroinflammation

Levels of

2

NLRP3 and other inflammasome components i.e. ASC, caspase-1, IL-1β and IL-18

3

were found to be overexpressed in the spinal cord of ALS-linked mutant SOD1 (G93A)

4

mice astrocytes and also in the tissue of ALS patient these levels are increased as

5

compared to healthy volunteers 125.

6 7

Figure 5: Illustration representing the role of genetic alteration and inflammatory

8

cascade in the development of ALS: A) Genetic alteration: 1) Mutations in TBK1

9

promotes phosphorylation of ubiquitinated adaptor proteins 2) E4784 mutation impairs

10

tethering of ubiquitinated adaptor proteins and AMBRA1 of AP and ultimately impairs

11

mitophagy 3) D337E and L341V mutation impairs autophagy via inhibiting interaction

12

of SQSTM1 and AMBRA1 of AP B) Inflammatory cascade: development of ALS

13

activates inflammasome directly via activation of 1) TLRs and NFkB 2) SOD1 or

14

indirectly via activation of 3) ROS and RNS which promotes nitration of protein.

15

Activation of inflammasome further activates caspase-1 followed by activation of Il-1β,

16

IL-18 and further triggers neuroinflammation.

17 18

E) Other neurological disorders 21 ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 40

1 2

Other neurological conditions have also shown the involvement of mitophagy and

3

inflammasomes. These include epilepsy, Huntington’s disease, multiple sclerosis and

4

autism. Details of each have been summarised in table 1.

5

1. Huntington’s disease (HD)

6

Huntington’s disease is associated with the mutation of huntingtin gene (mtHtt) which

7

activates a cascade of detrimental events resulting in neurodegeneration.

8

Mitochondrial dysfunction is one of the major events involved in HD. VCP translocates

9

to mitochondria and interacts with UBXD1 leading to degradation of the OMM protein

10

MCL1, as a result of which mitophagy increases126,

11

mediated mitophagy128. Huntingtin facilitates the association of p62 with integral

12

autophagosome component, however due to mutation in huntingtin, mitophagy is

13

impaired129. mtHtt also triggers the activation of inflammasomes via NFkβ. This further

14

activates neuroinflammation by upregulating IL-6, IL-8 and TNF-α130. mtHtt inhibits the

15

phosphorylation induced by IGF-1/Akt, leading to neurotoxicity131, 132.

16

127

. mtHtt disturbs PINK/Parkin

2. Epilepsy

17

In case of refractory temporal lobe epilepsy with hippocampal sclerosis, LC3B was

18

found to be upregulated133. Evidence of role of inflammasome in epilepsy has been

19

reported in many studies suggesting that deficiency of COX and its product PGF-2α

20

may be responsible for increased susceptibility towards epilepsy134. Overexpression of

21

genes related to pro-inflammatory cytokines and NOS have been found in the

22

olfactory-bulb of frontal lobe epilepsy patients135. Epilepsy enhances release of HMGB-

23

1, an inflammatory cytokine which promotes neuroinflammation via activation of TLR-

24

4136.

25

3. Multiple sclerosis (MS)

26

Involvement of mitophagy and inflammasome have been documented in MS patients.

27

Increased levels of ATG5 and parkin have been reported in body fluids of MS

28

patients137. Active involvement of caspase 1 has also been reported in MS which

29

directly leads to inflammation138 139.

30

4. Autism

31

Wdfy3 is an autism risk gene ideally required for maintaining mitochondrial

32

homeostasis. Mutation of this gene in autism leads to inhibition of mitophagy 140. AIM2

22 ACS Paragon Plus Environment

Page 23 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1

and NLRP3 have been found to be upregulated in the patients of autism, suggesting

2

the involvement of inflammasomes in autism141.

3 4 5

Table1: List of neurological diseases involving molecular mechanisms behind

6

mitophagy and inflammasome

7 Sr.No.

Neurological disease

Event

Target

Molecular mechanism

GAPDH

Abnormal interaction of polyglutamine repeats with the GAPDH prevents GAPDHmediated mitophagy

VCP

Mitophagy PINK1/Parkin

1

Huntington’s disease

Inflammasome

Presence of mtHtt facilitates interaction of VCP and UBXD1 results into degradation of OMM-protein MCL1 leads to uncontrolled mitophagy mtHtt impairs PINK1/Parkin-mediated mitophagy

C-terminal of Htt

Impaired p62-mediated mitophagy due to mutation in C-terminal of Htt

NFkB

mtHtt activates NFkB cascade and upregulates IL-6,IL-8,TNF-α and promotes neuroinflammation

CCL4 and MMP9

Upregulated in the brains of HD patients

IGF-1/Akt

mtHtt mediates neurotoxicity via inhibitory effect on phosphorylation induced by IGF-1/Akt

23 ACS Paragon Plus Environment

Reference

142

126, 127

128

129

130

143

131, 132

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

LC3B Mitophagy

PGF-2α

Epilepsy Inflammasome

2

Mitophagy

3

Page 24 of 40

LC3B upregulated in case of rTLE with hippocampal sclerosis Lack of PGF-2α increased susceptibility to the seizures in immature brain

Proinflammatory cytokines and NOS genes

Upregulated in olfactory bulbs of FLE patients

HMGB-1

Epilepsy promotes release of HMGB-1, an inflammatory cytokine, activates TLR-4 leads to neuroinflammation

ATG5 and Parkin

ATG5 and Parkin activates mitophagy

Caspase-1

Caspase-1 activation followed by activation of glial inflammasomes and pyroptosis in MS

Multiple sclerosis

133

134

135

136

137

138

Inflammasome

Mitophagy

4

Autism Inflammasome

ASC and Caspase-1

MS promotes activation of inflammasome via activation of ASC and caspase-1

Wdfy3

Mutation in Wdfy3 leads to inhibition of mitophagy

AIM2 and NLRP3

Upregulated in case of ASD patients, responsible for inflammasome activation

I-FABP

Compromised GI permeability promotes release of I-FABP followed by release of ATP, an activator of DAMP and activates inflammasome

139

140

141

141

1 2

5. Concluding Remarks

3

Mitophagy and inflammasome activation are two main cellular events involved in

4

several neurological diseases. The former is important for elimination of damaged 24 ACS Paragon Plus Environment

Page 25 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1

mitochondria while the later provides a molecular platform for activation of caspase-1

2

which secretes IL-1β in response to various insults leading to neuroinflammation in

3

various neurological diseases. Each and every neurological disease comes with its

4

unique pathogenesis. Although, mitophagy and inflammasome have been implicated

5

in various neurological diseases like AD, PD, HD, MS, etc. The interplay between the

6

two in various neurological disorders is yet not clear. Thus, here we have tried to find

7

the link between inflammasome and mitophagy in different neurological diseases that

8

was not put forward effectively. By developing some novel therapeutics against the

9

target which regulates mitophagy and inflammasome activation, one may pave a way

10

to halt the progression of neurological disorders.

11

6.

12

Authors acknowledge Department of Science and Technology (DST),Govt.of India for

13

their financial support through a grant (SB/YS/LS-196/2014), International Society for

14

Neurochemistry (ISN) Return Home grant, Department of Pharmaceuticals, Ministry

15

of Chemical and Fertilizers, Govt of India and National Institute of Pharmaceutical

16

Education and Research (NIPER) Ahmedabad, Gandhinagar, India.

17

Acknowledgements

7. Conflict of Interest

18

The authors declare that they have no conflicts of interest.

19

Authors Contributions:

20

RK, DS, PB, KD and DY conceived and designed the study. RK,DS,PB,

21

KD,AB,HK,LM,GV,VP,VK, KK and DY outlined the performed rigorous literature

22

search. RK, DS, PB, KD, KK, DY conceived and designed the figures and images. RK,

23

DS, PB, KD, KK, and DY wrote the manuscript.

24

References

25 26 27 28 29 30 31 32 33 34 35

1. Rane, P., Sarmah, D., Bhute, S., Kaur, H., Goswami, A., Kalia, K., Borah, A., Dave, K. R., Sharma, N., and Bhattacharya, P. (2018) Novel Targets for Parkinson’s Disease: Addressing Different Therapeutic Paradigms and Conundrums, ACS chemical neuroscience 10, 44-57. 2. Pravalika, K., Sarmah, D., Kaur, H., Vats, K., Saraf, J., Wanve, M., Kalia, K., Borah, A., Yavagal, D. R., and Dave, K. R. (2019) Trigonelline therapy confers neuroprotection by reduced glutathione mediated myeloperoxidase expression in animal model of ischemic stroke, Life sciences 216, 49-58. 3. Pravalika, K., Sarmah, D., Kaur, H., Wanve, M., Saraf, J., Kalia, K., Borah, A., Yavagal, D. R., Dave, K. R., and Bhattacharya, P. (2018) Myeloperoxidase and Neurological disorder: A crosstalk, ACS chemical neuroscience 9, 421-430. 25 ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49

4. Wanve, M., Kaur, H., Sarmah, D., Saraf, J., Pravalika, K., Vats, K., Kalia, K., Borah, A., Yavagal, D. R., and Dave, K. R. (2019) Therapeutic spectrum of interferon‐β in ischemic stroke, Journal of neuroscience research 97, 116-127. 5. Vats, K., Sarmah, D., Kaur, H., Wanve, M., Kalia, K., Borah, A., Dave, K. R., Yavagal, D. R., and Bhattacharya, P. (2018) Inflammasomes in stroke: a triggering role for acid‐sensing ion channels, Annals of the New York Academy of Sciences. 6. Sarmah, D., Kaur, H., Saraf, J., Vats, K., Pravalika, K., Wanve, M., Kalia, K., Borah, A., Kumar, A., and Wang, X. (2018) Mitochondrial dysfunction in stroke: implications of stem cell therapy, Translational stroke research, 1-16. 7. Saraf, J., Bhattacharya, P., Kalia, K., Borah, A., Sarmah, D., Kaur, H., Dave, K. R., and Yavagal, D. R. (2018) A Friend or Foe: Calcineurin across the Gamut of Neurological Disorders, ACS Central Science 4, 805-819. 8. Ashrafi, G., and Schwarz, T. (2013) The pathways of mitophagy for quality control and clearance of mitochondria, Cell death and differentiation 20, 31. 9. Glick, D., Barth, S., and Macleod, K. F. (2010) Autophagy: cellular and molecular mechanisms, The Journal of pathology 221, 3-12. 10. Lee, J., Giordano, S., and Zhang, J. (2012) Autophagy, mitochondria and oxidative stress: cross-talk and redox signalling, Biochemical Journal 441, 523-540. 11. Pickles, S., Vigié, P., and Youle, R. J. (2018) Mitophagy and quality control mechanisms in mitochondrial maintenance, Current Biology 28, R170-R185. 12. Martinez-Vicente, M. (2017) Neuronal mitophagy in neurodegenerative diseases, Frontiers in molecular neuroscience 10, 64. 13. Kissová, I., Deffieu, M., Manon, S., and Camougrand, N. (2004) Uth1p is involved in the autophagic degradation of mitochondria, Journal of Biological Chemistry 279, 39068-39074. 14. Rodriguez-Enriquez, S., Kim, I., Currin, R. T., and Lemasters, J. J. (2006) Tracker dyes to probe mitochondrial autophagy (mitophagy) in rat hepatocytes, Autophagy 2, 39-46. 15. Lemasters, J. J. (2014) Variants of mitochondrial autophagy: Types 1 and 2 mitophagy and micromitophagy (Type 3), Redox biology 2, 749-754. 16. Chinnadurai, G., Vijayalingam, S., and Gibson, S. B. (2009) BNIP3 subfamily BH3only proteins: mitochondrial stress sensors in normal and pathological functions, Oncogene 27, S114. 17. Liu, K. E., and Frazier, W. A. (2015) Phosphorylation of the BNIP3 C-terminus inhibits mitochondrial damage and cell death without blocking autophagy, PloS one 10, e0129667. 18. Sandoval, H., Thiagarajan, P., Dasgupta, S. K., Schumacher, A., Prchal, J. T., Chen, M., and Wang, J. (2008) Essential role for Nix in autophagic maturation of erythroid cells, Nature 454, 232. 19. Gálvez, A. S., Brunskill, E. W., Marreez, Y., Benner, B. J., Regula, K. M., Kirschenbaum, L. A., and Dorn, G. W. (2006) Distinct pathways regulate proapoptotic Nix and BNip3 in cardiac stress, Journal of Biological Chemistry 281, 1442-1448. 20. Bellot, G., Garcia-Medina, R., Gounon, P., Chiche, J., Roux, D., Pouysségur, J., and Mazure, N. M. (2009) Hypoxia-induced autophagy is mediated through hypoxiainducible factor induction of BNIP3 and BNIP3L via their BH3 domains, Molecular and cellular biology 29, 2570-2581. 21. Bruick, R. K. (2000) Expression of the gene encoding the proapoptotic Nip3 protein is induced by hypoxia, Proceedings of the National Academy of Sciences 97, 90829087.

26 ACS Paragon Plus Environment

Page 26 of 40

Page 27 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48

ACS Chemical Neuroscience

22. Guo, K., Searfoss, G., Krolikowski, D., Pagnoni, M., Franks, C., Clark, K., Yu, K., Jaye, M., and Ivashchenko, Y. (2001) Hypoxia induces the expression of the proapoptotic gene BNIP3, Cell death and differentiation 8, 367. 23. Zhang, H., Bosch-Marce, M., Shimoda, L. A., Tan, Y. S., Baek, J. H., Wesley, J. B., Gonzalez, F. J., and Semenza, G. L. (2008) Mitochondrial autophagy is an HIF-1dependent adaptive metabolic response to hypoxia, Journal of Biological Chemistry 283, 10892-10903. 24. Shi, R. Y., Zhu, S. H., Li, V., Gibson, S. B., Xu, X. S., and Kong, J. M. (2014) BNIP3 interacting with LC3 triggers excessive mitophagy in delayed neuronal death in stroke, CNS neuroscience & therapeutics 20, 1045-1055. 25. Liu, L., Feng, D., Chen, G., Chen, M., Zheng, Q., Song, P., Ma, Q., Zhu, C., Wang, R., and Qi, W. (2012) Mitochondrial outer-membrane protein FUNDC1 mediates hypoxia-induced mitophagy in mammalian cells, Nature cell biology 14, 177. 26. Chen, M., Chen, Z., Wang, Y., Tan, Z., Zhu, C., Li, Y., Han, Z., Chen, L., Gao, R., and Liu, L. (2016) Mitophagy receptor FUNDC1 regulates mitochondrial dynamics and mitophagy, Autophagy 12, 689-702. 27. Chen, G., Han, Z., Feng, D., Chen, Y., Chen, L., Wu, H., Huang, L., Zhou, C., Cai, X., and Fu, C. (2014) A regulatory signaling loop comprising the PGAM5 phosphatase and CK2 controls receptor-mediated mitophagy, Molecular cell 54, 362-377. 28. Wu, H., Xue, D., Chen, G., Han, Z., Huang, L., Zhu, C., Wang, X., Jin, H., Wang, J., and Zhu, Y. (2014) The BCL2L1 and PGAM5 axis defines hypoxia-induced receptor-mediated mitophagy, Autophagy 10, 1712-1725. 29. Lahiri, V., and Klionsky, D. J. (2017) PHB2/prohibitin 2: An inner membrane mitophagy receptor, Cell research 27, 311. 30. Wei, Y., Chiang, W.-C., Sumpter, R., Mishra, P., and Levine, B. (2017) Prohibitin 2 is an inner mitochondrial membrane mitophagy receptor, Cell 168, 224-238. e210. 31. Yoshii, S. R., Kishi, C., Ishihara, N., and Mizushima, N. (2011) Parkin mediates proteasome-dependent protein degradation and rupture of the outer mitochondrial membrane, Journal of Biological Chemistry, jbc. M110. 209338. 32. Kim, H. T., and Goldberg, A. L. (2017) The deubiquitinating enzyme Usp14 allosterically inhibits multiple proteasomal activities and ubiquitin-independent proteolysis, Journal of Biological Chemistry, jbc. M116. 763128. 33. Geisler, S., Holmström, K. M., Skujat, D., Fiesel, F. C., Rothfuss, O. C., Kahle, P. J., and Springer, W. (2010) PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1, Nature cell biology 12, 119. 34. Matsuda, N., Sato, S., Shiba, K., Okatsu, K., Saisho, K., Gautier, C. A., Sou, Y.s., Saiki, S., Kawajiri, S., and Sato, F. (2010) PINK1 stabilized by mitochondrial depolarization recruits Parkin to damaged mitochondria and activates latent Parkin for mitophagy, The Journal of cell biology 189, 211-221. 35. Clark, I. E., Dodson, M. W., Jiang, C., Cao, J. H., Huh, J. R., Seol, J. H., Yoo, S. J., Hay, B. A., and Guo, M. (2006) Drosophila pink1 is required for mitochondrial function and interacts genetically with parkin, Nature 441, 1162. 36. Park, J., Lee, S. B., Lee, S., Kim, Y., Song, S., Kim, S., Bae, E., Kim, J., Shong, M., and Kim, J.-M. (2006) Mitochondrial dysfunction in Drosophila PINK1 mutants is complemented by parkin, Nature 441, 1157. 37. Jin, S. M., Lazarou, M., Wang, C., Kane, L. A., Narendra, D. P., and Youle, R. J. (2010) Mitochondrial membrane potential regulates PINK1 import and proteolytic destabilization by PARL, The Journal of cell biology 191, 933-942.

27 ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49

38. Meissner, C., Lorenz, H., Weihofen, A., Selkoe, D. J., and Lemberg, M. K. (2011) The mitochondrial intramembrane protease PARL cleaves human Pink1 to regulate Pink1 trafficking, Journal of neurochemistry 117, 856-867. 39. Hollville, E., Carroll, R. G., Cullen, S. P., and Martin, S. J. (2014) Bcl-2 family proteins participate in mitochondrial quality control by regulating Parkin/PINK1dependent mitophagy, Molecular cell 55, 451-466. 40. Narendra, D. P., Jin, S. M., Tanaka, A., Suen, D.-F., Gautier, C. A., Shen, J., Cookson, M. R., and Youle, R. J. (2010) PINK1 is selectively stabilized on impaired mitochondria to activate Parkin, PLoS biology 8, e1000298. 41. Wong, Y. C., and Holzbaur, E. L. (2014) Optineurin is an autophagy receptor for damaged mitochondria in parkin-mediated mitophagy that is disrupted by an ALSlinked mutation, Proceedings of the National Academy of Sciences 111, E4439E4448. 42. Strappazzon, F., Nazio, F., Corrado, M., Cianfanelli, V., Romagnoli, A., Fimia, G., Campello, S., Nardacci, R., Piacentini, M., and Campanella, M. (2015) AMBRA1 is able to induce mitophagy via LC3 binding, regardless of PARKIN and p62/SQSTM1, Cell death and differentiation 22, 419. 43. Di Rita, A., Peschiaroli, A., Pasquale, D., Strobbe, D., Hu, Z., Gruber, J., Nygaard, M., Lambrughi, M., Melino, G., and Papaleo, E. (2018) HUWE1 E3 ligase promotes PINK1/PARKIN-independent mitophagy by regulating AMBRA1 activation via IKKα, Nature communications 9, 3755. 44. Bernardini, J. P., Brouwer, J. M., Tan, I. K., Sandow, J. J., Huang, S., Stafford, C. A., Bankovacki, A., Riffkin, C. D., Wardak, A. Z., and Czabotar, P. E. (2019) Parkin inhibits BAK and BAX apoptotic function by distinct mechanisms during mitophagy, The EMBO journal 38, e99916. 45. Wang, L., Cho, Y.-L., Tang, Y., Wang, J., Park, J.-E., Wu, Y., Wang, C., Tong, Y., Chawla, R., and Zhang, J. (2018) PTEN-L is a novel protein phosphatase for ubiquitin dephosphorylation to inhibit PINK1–Parkin-mediated mitophagy, Cell Research, 1. 46. Lu, A., and Wu, H. (2015) Structural mechanisms of inflammasome assembly, The FEBS journal 282, 435-444. 47. Broz, P., and Dixit, V. M. (2016) Inflammasomes: mechanism of assembly, regulation and signalling, Nature Reviews Immunology 16, 407. 48. Kayagaki, N., Wong, M. T., Stowe, I. B., Ramani, S. R., Gonzalez, L. C., AkashiTakamura, S., Miyake, K., Zhang, J., Lee, W. P., and Muszyński, A. (2013) Noncanonical inflammasome activation by intracellular LPS independent of TLR4, Science, 1240248. 49. Zanoni, I., Tan, Y., Di Gioia, M., Broggi, A., Ruan, J., Shi, J., Donado, C. A., Shao, F., Wu, H., and Springstead, J. R. (2016) An endogenous caspase-11 ligand elicits interleukin-1 release from living dendritic cells, Science 352, 1232-1236. 50. Shi, J., Zhao, Y., Wang, Y., Gao, W., Ding, J., Li, P., Hu, L., and Shao, F. (2014) Inflammatory caspases are innate immune receptors for intracellular LPS, Nature 514, 187. 51. Medzhitov, R. (2007) Recognition of microorganisms and activation of the immune response, Nature 449, 819. 52. Medzhitov, R. (2010) Innate immunity: quo vadis?, Nature immunology 11, 551. 53. Palm, N., Zhu, S., and Flavell, R. (2014) Inflammasomes, Cold Spring Harbor perspectives in biology 6, a016287-a016287. 54. Ye, Z., and Ting, J. P.-Y. (2008) NLR, the nucleotide-binding domain leucine-rich repeat containing gene family, Current opinion in immunology 20, 3-9.

28 ACS Paragon Plus Environment

Page 28 of 40

Page 29 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50

ACS Chemical Neuroscience

55. Walsh, J. G., Muruve, D. A., and Power, C. (2014) Inflammasomes in the CNS, Nature Reviews Neuroscience 15, 84. 56. Ding, J., Wang, K., Liu, W., She, Y., Sun, Q., Shi, J., Sun, H., Wang, D.-C., and Shao, F. (2016) Pore-forming activity and structural autoinhibition of the gasdermin family, Nature 535, 111. 57. Liu, X., Zhang, Z., Ruan, J., Pan, Y., Magupalli, V. G., Wu, H., and Lieberman, J. (2016) Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores, Nature 535, 153. 58. Wan, P., Zhang, Q., Liu, W., Jia, Y., Ai, S., Wang, T., Wang, W., Pan, P., Yang, G., and Xiang, Q. (2019) Cullin1 binds and promotes NLRP3 ubiquitination to repress systematic inflammasome activation, The FASEB Journal, fj. 201801681R. 59. Sarkar, S., Malovic, E., Harishchandra, D. S., Ghaisas, S., Panicker, N., Charli, A., Palanisamy, B. N., Rokad, D., Jin, H., and Anantharam, V. (2017) Mitochondrial impairment in microglia amplifies NLRP3 inflammasome proinflammatory signaling in cell culture and animal models of Parkinson’s disease, npj Parkinson's Disease 3, 30. 60. Ferri, A., Nencini, M., Cozzolino, M., Carrara, P., Moreno, S., and Carrì, M. T. (2008) Inflammatory cytokines increase mitochondrial damage in motoneuronal cells expressing mutant SOD1, Neurobiology of disease 32, 454-460. 61. Kaur, H., Sarmah, D., Saraf, J., Vats, K., Kalia, K., Borah, A., Yavagal, D. R., Dave, K. R., Ghosh, Z., and Bhattacharya, P. (2018) Noncoding RNAs in ischemic stroke: time to translate, Annals of the New York Academy of Sciences 1421, 19-36. 62. Abulafia, D. P., de Rivero Vaccari, J. P., Lozano, J. D., Lotocki, G., Keane, R. W., and Dietrich, W. D. (2009) Inhibition of the inflammasome complex reduces the inflammatory response after thromboembolic stroke in mice, Journal of Cerebral Blood Flow & Metabolism 29, 534-544. 63. Sarmah, D., Kaur, H., Saraf, J., Pravalika, K., Goswami, A., Kalia, K., Borah, A., Wang, X., Dave, K. R., and Yavagal, D. R. (2018) Getting closer to an effective intervention of ischemic stroke: the big promise of stem cell, Translational stroke research 9, 356-374. 64. Fann, D. Y.-W., Lim, Y.-A., Cheng, Y.-L., Lok, K.-Z., Chunduri, P., Baik, S.-H., Drummond, G. R., Dheen, S. T., Sobey, C. G., and Jo, D.-G. (2018) Evidence that NFκB and MAPK signaling promotes NLRP inflammasome activation in neurons following ischemic stroke, Molecular neurobiology 55, 1082-1096. 65. Poh, L., Kang, S.-W., Baik, S.-H., Ng, G. Y. Q., She, D. T., Balaganapathy, P., Dheen, S. T., Magnus, T., Gelderblom, M., and Sobey, C. G. (2018) Evidence that NLRC4 Inflammasome Mediates Apoptotic and Pyroptotic Microglial Death Following Ischemic Stroke, Brain, Behavior, and Immunity. 66. d’Adesky, N. D., de Rivero Vaccari, J. P., Bhattacharya, P., Schatz, M., PerezPinzon, M. A., Bramlett, H. M., and Raval, A. P. (2018) Nicotine alters estrogen receptor-Beta-regulated Inflammasome activity and exacerbates ischemic brain damage in female rats, International journal of molecular sciences 19. 67. Hou, Y., Wang, Y., He, Q., Li, L., Xie, H., Zhao, Y., and Zhao, J. (2018) Nrf2 inhibits NLRP3 inflammasome activation through regulating Trx1/TXNIP complex in cerebral ischemia reperfusion injury, Behavioural brain research 336, 32-39. 68. Toyama, E. Q., Herzig, S., Courchet, J., Lewis, T. L., Losón, O. C., Hellberg, K., Young, N. P., Chen, H., Polleux, F., and Chan, D. C. (2016) AMP-activated protein kinase mediates mitochondrial fission in response to energy stress, Science 351, 275281. 69. Dzierzak, E., and Philipsen, S. (2013) Erythropoiesis: development and differentiation, Cold Spring Harbor perspectives in medicine 3, a011601. 29 ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50

70. Yuan, Y., Zheng, Y., Zhang, X., Chen, Y., Wu, X., Wu, J., Shen, Z., Jiang, L., Wang, L., and Yang, W. (2017) BNIP3L/NIX-mediated mitophagy protects against ischemic brain injury independent of PARK2, Autophagy 13, 1754-1766. 71. Zhang, C.-h., Fan, Y.-y., Wang, X.-f., Xiong, J.-y., Tang, Y.-y., Gao, J.-q., Shen, Z., Song, X.-h., Zhang, J.-y., and Shen, Y. (2012) Acidic preconditioning protects against ischemia-induced brain injury, Neuroscience letters 523, 3-8. 72. Wang, Y., Zeng, W., Xiao, X., Huang, Y., Song, X., Yu, Z., Tang, D., Dong, X., Zhu, M., and Xu, T. (2013) Intracellular ASIC1a regulates mitochondrial permeability transition-dependent neuronal death, Cell death and differentiation 20, 1359. 73. Shen, Z., Zheng, Y., Wu, J., Chen, Y., Wu, X., Zhou, Y., Yuan, Y., Lu, S., Jiang, L., and Qin, Z. (2017) PARK2-dependent mitophagy induced by acidic postconditioning protects against focal cerebral ischemia and extends the reperfusion window, Autophagy 13, 473-485. 74. Feng, J., Chen, X., Guan, B., Li, C., Qiu, J., and Shen, J. (2018) Inhibition of Peroxynitrite-induced mitophagy activation attenuates cerebral ischemia-reperfusion injury, Molecular neurobiology, 1-18. 75. Zhang, Z., and Yu, J. (2018) NR4A1 Promotes Cerebral Ischemia Reperfusion Injury by Repressing Mfn2-Mediated Mitophagy and Inactivating the MAPK–ERK– CREB Signaling Pathway, Neurochemical research, 1-15. 76. Lan, R., Wu, J.-T., Wu, T., Ma, Y.-Z., Wang, B.-Q., Zheng, H.-Z., Li, Y.-N., Wang, Y., Gu, C.-Q., and Zhang, Y. (2018) Mitophagy is activated in brain damage induced by cerebral ischemia and reperfusion via the PINK1/Parkin/p62 signalling pathway, Brain research bulletin 142, 63-77. 77. Joers, V., Tansey, M. G., Mulas, G., and Carta, A. R. (2017) Microglial phenotypes in Parkinson’s disease and animal models of the disease, Progress in neurobiology 155, 57-75. 78. Lawana, V., Singh, N., Sarkar, S., Charli, A., Jin, H., Anantharam, V., Kanthasamy, A. G., and Kanthasamy, A. (2017) Involvement of c-Abl kinase in microglial activation of NLRP3 inflammasome and impairment in autolysosomal system, Journal of Neuroimmune Pharmacology 12, 624-660. 79. Bai, H., Yang, B., Yu, W., Xiao, Y., Yu, D., and Zhang, Q. (2018) Cathepsin B links oxidative stress to the activation of NLRP3 inflammasome, Experimental cell research 362, 180-187. 80. . Chun, H. S., Gibson, G. E., DeGiorgio, L. A., Zhang, H., Kidd, V. J., and Son, J. H. (2001) Dopaminergic cell death induced by MPP+, oxidant and specific neurotoxicants shares the common molecular mechanism, Journal of neurochemistry 76, 1010-1021. 81. Wang, W., Nguyen, L. T., Burlak, C., Chegini, F., Guo, F., Chataway, T., Ju, S., Fisher, O. S., Miller, D. W., and Datta, D. (2016) Caspase-1 causes truncation and aggregation of the Parkinson’s disease-associated protein α-synuclein, Proceedings of the National Academy of Sciences 113, 9587-9592. 82. Codolo, G., Plotegher, N., Pozzobon, T., Brucale, M., Tessari, I., Bubacco, L., and de Bernard, M. (2013) Triggering of inflammasome by aggregated α–synuclein, an inflammatory response in synucleinopathies, PloS one 8, e55375. 83. Gustot, A., Gallea, J. I., Sarroukh, R., Celej, M. S., Ruysschaert, J.-M., and Raussens, V. (2015) Amyloid fibrils are the molecular trigger of inflammation in Parkinson's disease, Biochemical Journal 471, 323-333. 84. Fan, Z., Lu, M., Qiao, C., Zhou, Y., Ding, J.-H., and Hu, G. (2016) MicroRNA-7 enhances subventricular zone neurogenesis by inhibiting NLRP3/caspase-1 axis in adult neural stem cells, Molecular neurobiology 53, 7057-7069. 30 ACS Paragon Plus Environment

Page 30 of 40

Page 31 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49

ACS Chemical Neuroscience

85. von Herrmann, K. M., Salas, L. A., Martinez, E. M., Young, A. L., Howard, J. M., Feldman, M. S., Christensen, B. C., Wilkins, O. M., Lee, S. L., and Hickey, W. F. (2018) NLRP3 expression in mesencephalic neurons and characterization of a rare NLRP3 polymorphism associated with decreased risk of Parkinson’s disease, NPJ Parkinson's disease 4, 24. 86. Qiao, C., Zhang, Q., Jiang, Q., Zhang, T., Chen, M., Fan, Y., Ding, J., Lu, M., and Hu, G. (2018) Inhibition of the hepatic Nlrp3 protects dopaminergic neurons via attenuating systemic inflammation in a MPTP/p mouse model of Parkinson’s disease, Journal of neuroinflammation 15, 193. 87. Rodrigue-Gervais, I. G., Doiron, K., Champagne, C., Mayes, L., Leiva-Torres, G. A., Vanié, P., Douglas, T., Vidal, S. M., Alnemri, E. S., and Saleh, M. (2018) The mitochondrial protease HtrA2 restricts the NLRP3 and AIM2 inflammasomes, Scientific reports 8, 8446. 88. Giacoppo, S., Bramanti, P., and Mazzon, E. (2017) Triggering of inflammasome by impaired autophagy in response to acute experimental Parkinson’s disease: Involvement of the PI3K/Akt/mTOR pathway, Neuroreport 28, 996. 89. Chen, L., Xue, L., Zheng, J., Tian, X., Zhang, Y., and Tong, Q. (2019) PPARß/δ agonist alleviates NLRP3 inflammasome-mediated neuroinflammation in the MPTP mouse model of Parkinson’s disease, Behavioural brain research 356, 483-489. 90. Zhu, J., Hu, Z., Han, X., Wang, D., Jiang, Q., Ding, J., Xiao, M., Wang, C., Lu, M., and Hu, G. (2018) Dopamine D2 receptor restricts astrocytic NLRP3 inflammasome activation via enhancing the interaction of β-arrestin2 and NLRP3, Cell Death & Differentiation, 1. 91. Grenier, K., McLelland, G.-L., and Fon, E. A. (2013) Parkin-and PINK1-dependent mitophagy in neurons: will the real pathway please stand up?, Frontiers in neurology 4, 100. 92. Sekine, S., Kanamaru, Y., Koike, M., Nishihara, A., Okada, M., Kinoshita, H., Kamiyama, M., Maruyama, J., Uchiyama, Y., and Ishihara, N. (2012) Rhomboid protease PARL mediates the mitochondrial membrane potential loss-induced cleavage of PGAM5, Journal of Biological Chemistry, jbc. M112. 357509. 93. Lu, W., Karuppagounder, S. S., Springer, D. A., Allen, M. D., Zheng, L., Chao, B., Zhang, Y., Dawson, V. L., Dawson, T. M., and Lenardo, M. (2014) Genetic deficiency of the mitochondrial protein PGAM5 causes a Parkinson’s-like movement disorder, Nature communications 5, 4930. 94. Sugo, M., Kimura, H., Arasaki, K., Amemiya, T., Hirota, N., Dohmae, N., Imai, Y., Inoshita, T., Shiba‐Fukushima, K., and Hattori, N. (2018) Syntaxin 17 regulates the localization and function of PGAM5 in mitochondrial division and mitophagy, The EMBO journal, e98899. 95. Mishra, P., and Chan, D. C. (2016) Metabolic regulation of mitochondrial dynamics, J Cell Biol 212, 379-387. 96. Wai, T., and Langer, T. (2016) Mitochondrial dynamics and metabolic regulation, Trends in Endocrinology & Metabolism 27, 105-117. 97. Pickrell, A. M., and Youle, R. J. (2015) The roles of PINK1, parkin, and mitochondrial fidelity in Parkinson’s disease, Neuron 85, 257-273. 98. McLelland, G.-L., and Fon, E. A. (2018) MFN2 retrotranslocation boosts mitophagy by uncoupling mitochondria from the ER, Autophagy 14, 1658-1660. 99. Wu, Z., Wu, A., Dong, J., Sigears, A., and Lu, B. (2018) Grape skin extract improves muscle function and extends lifespan of a Drosophila model of Parkinson's disease through activation of mitophagy, Experimental gerontology 113, 10-17.

31 ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49

100. Pan-Montojo, F., Anichtchik, O., Dening, Y., Knels, L., Pursche, S., Jung, R., Jackson, S., Gille, G., Spillantini, M. G., and Reichmann, H. (2010) Progression of Parkinson's disease pathology is reproduced by intragastric administration of rotenone in mice, PloS one 5, e8762. 101. Peng, K., Xiao, J., Yang, L., Ye, F., Cao, J., and Sai, Y. (2018) Mutual Antagonism of PINK1/Parkin and PGC-1α Contributes to Maintenance of Mitochondrial Homeostasis in Rotenone-Induced Neurotoxicity, Neurotoxicity research, 1-13. 102. Morimoto, K., Horio, J., Satoh, H., Sue, L., Beach, T., Arita, S., Tooyama, I., and Konishi, Y. (2011) Expression profiles of cytokines in the brains of Alzheimer's disease (AD) patients compared to the brains of non-demented patients with and without increasing AD pathology, Journal of Alzheimer's Disease 25, 59-76. 103. Heneka, M. T., Kummer, M. P., Stutz, A., Delekate, A., Schwartz, S., VieiraSaecker, A., Griep, A., Axt, D., Remus, A., and Tzeng, T.-C. (2013) NLRP3 is activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice, Nature 493, 674. 104. Shi, J. Q., Zhang, C. C., Sun, X. L., Cheng, X. X., Wang, J. B., Zhang, Y. D., Xu, J., and Zou, H. Q. (2013) Antimalarial Drug Artemisinin Extenuates Amyloidogenesis and Neuroinflammation in APP swe/PS 1dE9 Transgenic Mice via Inhibition of Nuclear Factor‐κ B and NLRP 3 Inflammasome Activation, CNS neuroscience & therapeutics 19, 262-268. 105. Lopez-Rodriguez, A. B., Hennessy, E., Murray, C., Lewis, A., de Barra, N., Fagan, S., Rooney, M., Nazmi, A., and Cunningham, C. (2018) Microglial and Astrocyte priming in the APP/PS1 model of Alzheimer's Disease: increased vulnerability to acute inflammation and cognitive deficits, bioRxiv, 344218. 106. Ahmed, M. E., Iyer, S., Thangavel, R., Kempuraj, D., Selvakumar, G. P., Raikwar, S. P., Zaheer, S., and Zaheer, A. (2017) Co-Localization of glia maturation factor with NLRP3 inflammasome and autophagosome markers in human Alzheimer’s disease brain, Journal of Alzheimer's Disease 60, 1143-1160. 107. Kerr, J. S., Adriaanse, B. A., Greig, N. H., Mattson, M. P., Cader, M. Z., Bohr, V. A., and Fang, E. F. (2017) Mitophagy and Alzheimer’s disease: cellular and molecular mechanisms, Trends in neurosciences 40, 151-166. 108. Hu, Y., Li, X.-C., Wang, Z.-h., Luo, Y., Zhang, X., Liu, X.-P., Feng, Q., Wang, Q., Yue, Z., and Chen, Z. (2016) Tau accumulation impairs mitophagy via increasing mitochondrial membrane potential and reducing mitochondrial Parkin, Oncotarget 7, 17356. 109. Cummins, N., Tweedie, A., Zuryn, S., Bertran‐Gonzalez, J., and Götz, J. (2018) Disease‐associated tau impairs mitophagy by inhibiting Parkin translocation to mitochondria, The EMBO journal, e99360. 110. Corsetti, V., Florenzano, F., Atlante, A., Bobba, A., Ciotti, M., Natale, F., Della Valle, F., Borreca, A., Manca, A., and Meli, G. (2015) NH2-truncated human tau induces deregulated mitophagy in neurons by aberrant recruitment of Parkin and UCHL-1: implications in Alzheimer's disease, Human molecular genetics 24, 30583081. 111. Mejias, N. H., Martinez, C. C., Stephens, M. E., and de Rivero Vaccari, J. P. (2018) Contribution of the inflammasome to inflammaging, Journal of Inflammation 15, 23. 112. Van Der Graaff, M., de Jong, J., Baas, F., and De Visser, M. (2009) Upper motor neuron and extra-motor neuron involvement in amyotrophic lateral sclerosis: a clinical and brain imaging review, Neuromuscular Disorders 19, 53-58.

32 ACS Paragon Plus Environment

Page 32 of 40

Page 33 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49

ACS Chemical Neuroscience

113. Rosen, D. R., Siddique, T., Patterson, D., Figlewicz, D. A., Sapp, P., Hentati, A., Donaldson, D., Goto, J., O'Regan, J. P., and Deng, H.-X. (1993) Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis, Nature 362, 59. 114. Rogers, R. S., Tungtur, S., Tanaka, T., Nadeau, L. L., Badawi, Y., Wang, H., Ni, H.-M., Ding, W.-X., and Nishimune, H. (2017) Impaired mitophagy plays a role in denervation of neuromuscular junctions in ALS mice, Frontiers in neuroscience 11, 473. 115. Moore, A. S., and Holzbaur, E. L. (2016) Dynamic recruitment and activation of ALS-associated TBK1 with its target optineurin are required for efficient mitophagy, Proceedings of the National Academy of Sciences 113, E3349-E3358. 116. Goode, A., Butler, K., Long, J., Cavey, J., Scott, D., Shaw, B., Sollenberger, J., Gell, C., Johansen, T., and Oldham, N. J. (2016) Defective recognition of LC3B by mutant SQSTM1/p62 implicates impairment of autophagy as a pathogenic mechanism in ALS-FTLD, Autophagy 12, 1094-1104. 117. Kim, N. C., Tresse, E., Kolaitis, R.-M., Molliex, A., Thomas, R. E., Alami, N. H., Wang, B., Joshi, A., Smith, R. B., and Ritson, G. P. (2013) VCP is essential for mitochondrial quality control by PINK1/Parkin and this function is impaired by VCP mutations, Neuron 78, 65-80. 118. Johnson, J. O., Mandrioli, J., Benatar, M., Abramzon, Y., Van Deerlin, V. M., Trojanowski, J. Q., Gibbs, J. R., Brunetti, M., Gronka, S., and Wuu, J. (2010) Exome sequencing reveals VCP mutations as a cause of familial ALS, Neuron 68, 857-864. 119. Sullivan, P. M., Zhou, X., Robins, A. M., Paushter, D. H., Kim, D., Smolka, M. B., and Hu, F. (2016) The ALS/FTLD associated protein C9orf72 associates with SMCR8 and WDR41 to regulate the autophagy-lysosome pathway, Acta neuropathologica communications 4, 51. 120. Troost, D., Van den Oord, J., and JONG, J. V. D. (1990) Immunohistochemical characterization of the inflammatory infiltrate in amyotrophic lateral sclerosis, Neuropathology and applied neurobiology 16, 401-410. 121. Gugliandolo, A., Giacoppo, S., Bramanti, P., and Mazzon, E. (2018) NLRP3 Inflammasome Activation in a Transgenic Amyotrophic Lateral Sclerosis Model, Inflammation 41, 93-103. 122. Bellezza, I., Grottelli, S., Costanzi, E., Scarpelli, P., Pigna, E., Morozzi, G., Mezzasoma, L., Peirce, M. J., Moresi, V., and Adamo, S. (2018) Peroxynitrite activates the NLRP3 inflammasome cascade in SOD1 (G93A) mouse model of amyotrophic lateral sclerosis, Molecular neurobiology 55, 2350-2361. 123. Chattopadhyay, M., and Valentine, J. S. (2009) Aggregation of copper–zinc superoxide dismutase in familial and sporadic ALS, Antioxidants & redox signaling 11, 1603-1614. 124. Meissner, F., Molawi, K., and Zychlinsky, A. (2010) Mutant superoxide dismutase 1-induced IL-1β accelerates ALS pathogenesis, Proceedings of the National Academy of Sciences 107, 13046-13050. 125. Johann, S., Heitzer, M., Kanagaratnam, M., Goswami, A., Rizo, T., Weis, J., Troost, D., and Beyer, C. (2015) NLRP3 inflammasome is expressed by astrocytes in the SOD1 mouse model of ALS and in human sporadic ALS patients, Glia 63, 22602273. 126. Guo, X., and Qi, X. (2017) VCP cooperates with UBXD1 to degrade mitochondrial outer membrane protein MCL1 in model of Huntington's disease, Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1863, 552-559.

33 ACS Paragon Plus Environment

ACS Chemical Neuroscience 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50

127. Guo, X., Sun, X., Hu, D., Wang, Y.-J., Fujioka, H., Vyas, R., Chakrapani, S., Joshi, A. U., Luo, Y., and Mochly-Rosen, D. (2016) VCP recruitment to mitochondria causes mitophagy impairment and neurodegeneration in models of Huntington’s disease, Nature communications 7, 12646. 128. Khalil, B., El Fissi, N., Aouane, A., Cabirol-Pol, M.-J., Rival, T., and Liévens, J.C. (2015) PINK1-induced mitophagy promotes neuroprotection in Huntington’s disease, Cell death & disease 6, e1617. 129. Rui, Y.-N., Xu, Z., Patel, B., Chen, Z., Chen, D., Tito, A., David, G., Sun, Y., Stimming, E. F., and Bellen, H. J. (2015) Huntingtin functions as a scaffold for selective macroautophagy, Nature cell biology 17, 262. 130. Björkqvist, M., Wild, E. J., Thiele, J., Silvestroni, A., Andre, R., Lahiri, N., Raibon, E., Lee, R. V., Benn, C. L., and Soulet, D. (2008) A novel pathogenic pathway of immune activation detectable before clinical onset in Huntington's disease, Journal of Experimental Medicine 205, 1869-1877. 131. Humbert, S., Bryson, E. A., Cordelières, F. P., Connors, N. C., Datta, S. R., Finkbeiner, S., Greenberg, M. E., and Saudou, F. (2002) The IGF-1/Akt pathway is neuroprotective in Huntington's disease and involves Huntingtin phosphorylation by Akt, Developmental cell 2, 831-837. 132. Colin, E., Régulier, E., Perrin, V., Dürr, A., Brice, A., Aebischer, P., Déglon, N., Humbert, S., and Saudou, F. (2005) Akt is altered in an animal model of Huntington's disease and in patients, European Journal of Neuroscience 21, 1478-1488. 133. Wu, M., Liu, X., Chi, X., Zhang, L., Xiong, W., Chiang, S. M. V., Zhou, D., and Li, J. (2018) Mitophagy in Refractory Temporal Lobe Epilepsy Patients with Hippocampal Sclerosis, Cellular and molecular neurobiology 38, 479-486. 134. Chung, J.-I., Kim, A. Y., Lee, S. H., and Baik, E. J. (2013) Seizure susceptibility in immature brain due to lack of COX-2-induced PGF2α, Experimental neurology 249, 95-103. 135. Mercado-Gómez, O. F., Córdova-Dávalos, L., García-Betanzo, D., Rocha, L., Alonso-Vanegas, M. A., Cienfuegos, J., and Guevara-Guzmán, R. (2018) Overexpression of inflammatory-related and nitric oxide synthase genes in olfactory bulbs from frontal lobe epilepsy patients, Epilepsy research 148, 37-43. 136. Park, J. S., Gamboni-Robertson, F., He, Q., Svetkauskaite, D., Kim, J.-Y., Strassheim, D., Sohn, J.-W., Yamada, S., Maruyama, I., and Banerjee, A. (2005) High Mobility Group Box 1 protein (HMGB1) interacts with multiple Toll like receptors, American Journal of Physiology-Cell Physiology. 137. Patergnani, S., Castellazzi, M., Bonora, M., Marchi, S., Casetta, I., Pugliatti, M., Giorgi, C., Granieri, E., and Pinton, P. (2018) Autophagy and mitophagy elements are increased in body fluids of multiple sclerosis-affected individuals, J Neurol Neurosurg Psychiatry 89, 439-441. 138. McKenzie, B. A., Mamik, M. K., Saito, L. B., Boghozian, R., Monaco, M. C., Major, E. O., Lu, J.-Q., Branton, W. G., and Power, C. (2018) Caspase-1 inhibition prevents glial inflammasome activation and pyroptosis in models of multiple sclerosis, Proceedings of the National Academy of Sciences, 201722041. 139. Keane, R. W., Dietrich, W. D., and de Rivero Vaccari, J. P. (2018) inflammasome Proteins as Biomarkers of Multiple sclerosis, Frontiers in neurology 9, 135. 140. Napoli, E., Song, G., Panoutsopoulos, A., Riyadh, M. A., Kaushik, G., Halmai, J., Levenson, R., Zarbalis, K. S., and Giulivi, C. (2018) Beyond autophagy: a novel role for autism-linked Wdfy3 in brain mitophagy, Scientific reports 8, 11348. 141. Saresella, M., Piancone, F., Marventano, I., Zoppis, M., Hernis, A., Zanette, M., Trabattoni, D., Chiappedi, M., Ghezzo, A., and Canevini, M. P. (2016) Multiple 34 ACS Paragon Plus Environment

Page 34 of 40

Page 35 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Chemical Neuroscience

1 2 3 4 5 6 7 8

inflammasome complexes are activated in autistic spectrum disorders, Brain, behavior, and immunity 57, 125-133. 142. Hwang, S., Disatnik, M. H., and Mochly‐Rosen, D. (2015) Impaired GAPDH‐induced mitophagy contributes to the pathology of Huntington's disease, EMBO molecular medicine 7, 1307-1326. 143. Silvestroni, A., Faull, R. L., Strand, A. D., and Möller, T. (2009) Distinct neuroinflammatory profile in post-mortem human Huntington's disease, Neuroreport 20, 1098-1103.

9

Table of Contents Graphic

10

Inflammasome

Mitophagy

11

Cerebral Ischemia Multiple Sclerosis Parkinson’s Disease Alzheimer’s Disease Amyotrophic Lateral Sclerosis Autism Epilepsy Huntington’s Disease

35 ACS Paragon Plus Environment

ACS Chemical Neuroscience

Figure 1 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41

ACS Paragon Plus Environment

Page 36 of 40

Page 37 of 40

ACS Chemical Neuroscience

Figure 2 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41

ACS Paragon Plus Environment

ACS Chemical Neuroscience

Figure 3 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41

ACS Paragon Plus Environment

Page 38 of 40

Page 39 of 40 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41

ACS Chemical Neuroscience

ACS Paragon Plus Environment

ACS Chemical Neuroscience

Figure 5 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41

ACS Paragon Plus Environment

Page 40 of 40