Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
KINASE MUTATION-ASSOCIATED NEURODEGENERATIVE DISORDERS
Document Type and Number:
WIPO Patent Application WO/2019/040877
Kind Code:
A1
Abstract:
The present technology relates to methods for treating, preventing, and/or ameliorating kinase mutation- associated neurodegenerative diseases, including BRAFV600E-associated neurodegenerative diseases, in a subject in need thereof. In particular aspects, the present technology relates to the use of BRAF, MEK, and/or CSF-IR inhibitors to treat, prevent, and/or ameliorate kinase mutation-associated neurodegenerative diseases, including BRAFV600E- associated neurodegenerative diseases.

Inventors:
GEISSMANN FREDERIC (US)
MASS ELVIRA (US)
Application Number:
PCT/US2018/047964
Publication Date:
February 28, 2019
Filing Date:
August 24, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MEMORIAL SLOAN KETTERING CANCER CENTER (US)
International Classes:
A61K31/506; A61K31/513; A61K31/519; A61K31/5377; A61K39/395; A61K45/00
Foreign References:
US20100183600A12010-07-22
US20160009663A12016-01-14
US20150030588A12015-01-29
US20150354012A12015-12-10
US20170321281A12017-11-09
Other References:
PRABOWO ET AL.: "BRAF V600E Mutation Is Associated with mTOR Signaling Activation in Glioneuronal Tumors", BRAIN PATHOLOGY, vol. 24, no. 1, 13 August 2013 (2013-08-13), pages 52 - 66, XP055578147
MASS ET AL.: "A Somatic Mutation in Erythro-Myeloid progenitors Causes Neurodegenerative Disease", NATURE, vol. 549, no. 7672, 21 September 2017 (2017-09-21), pages 389 - 393, XP055578150
SATOH ET AL.: "B-RAF Mutant Alleles Associated with Langerhans Cell Histiocytosis, a Granulomatous Pediatric Disease", PLOS ONE, vol. 7, 10 April 2010 (2010-04-10), pages 1 - 9, XP055362288
See also references of EP 3672596A4
Attorney, Agent or Firm:
EWING, James F. et al. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A method for treating or preventing BRAFV600E-associated neurodegenerative disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a BRAF, MEK, and/or CSF-1R inhibitor, or a pharmaceutically acceptable salt thereof.

2. The method of claim 1, wherein at least a portion of the resident macrophages in the central nervous system of the subject are BRAFV600E+.

3. A method for treating or preventing BRAFV600E-associated neurodegenerative disease comprising:

(a) isolating resident macrophages from a neuronal environment of the subject;

(b) determining whether the resident macrophages express BRAFV600E+; and

(c) administering to the subject a therapeutically effective amount of a BRAF, MEK, and/or CSF-1R inhibitor, or a pharmaceutically acceptable salt thereof, when the isolated resident macrophages express BRAFV600E+.

4. The method of any one of claims 1-3, wherein the neurodegenerative disease is characterized by one or more of impaired cognitive functions, dementia, ataxia, dysarthria, reduced motor coordination and synchrony as compared to a normal control subject, paralysis, microglia accumulation, astrogliosis, microglia phagocytosis, demyelination, neuronal loss in the central nervous system, synaptic loss in the central nervous system, and amyloid precursor protein (APP) deposits in the brain.

5. The method of any one of claims 1-4, wherein the BRAF inhibitor is selected from the group consisting of vemurafenib, dabrafenib, encorafenib, PLX7904, PLX8394, GDC-0879, LGX818, and PLX4720, the MEK inhibitor is selected from the group consisting of AZD8330, refametinib, E6201, MEK162 (binimetinib), PD0325901, pimasertib, R04987655, selumetinib, TAK-733, GDC-0623, WX-544, cobimetinib, and trametinib, and the CSF-1R inhibitor is selected from the group consisting of GW2580, BLZ945, pexidartinib (PLX3397), ARRY-382, PLX7486, and JNJ-40346527.

6. The method of any one of claims 1-5, wherein the route of administration of the BRAF, MEK, or CSF-1R inhibitor is parenteral, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, oral, sublingual, intranasal, intracerebral, intrathecal, intravaginal, transdermal, rectal, by inhalation, or topical.

7. The method of any one of claims 1-6, wherein treatment of the neurodegenerative disease comprises one or more of improving cognitive functions, reducing dementia, reducing ataxia, reducing dysarthria, increasing motor coordination and synchrony, relieving paralysis, reducing microglia accumulation, reducing astrogliosis, reducing microglia phagocytosis, reducing demyelination, reducing neuronal loss, reducing synaptic loss, or reducing amyloid precursor protein (APP) expression in the brain as compared to an untreated control.

8. The method of claim 5, wherein the BRAF inhibitor is vemurafenib.

9. The method of claim 5, wherein the BRAF inhibitor is PLX4720.

10. A method for treating or preventing neurodegenerative disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a PI 3- kinase inhibitor, or a pharmaceutically acceptable salt thereof, wherein at least a portion of the resident macrophages in the central nervous system of the subject comprise one or more PI 3- kinase mutations.

11. The method of claim 10, wherein at least a portion of the resident macrophages in the central nervous system of the subject are PIK3CAH1047R+.

12. The method of claim 10 or 11, wherein the neurodegenerative disease is characterized by one or more of impaired cognitive functions, dementia, ataxia, dysarthria, reduced motor coordination and synchrony as compared to a normal control subject, paralysis, microglia accumulation, astrogliosis, microglia phagocytosis, demyelination, neuronal loss in the central nervous system, synaptic loss in the central nervous system, and amyloid precursor protein (APP) deposits in the brain.

13. The method of any one of claims 10-12, wherein the PI 3-kinase inhibitor is selected from the group consisting of idelalisib, BKM120, GDC-0980, PF-04691502, XL147, IPI-145, BYL719, SF1126, BAY80-6946, GSK2126458, NVP-BEZ235, GDC-0941, PX-866, XL765, and ZSTK474.

14. The method of any one of claims 10-13, wherein the route of administration of the PI 3- kinase inhibitor is parenteral, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, oral, sublingual, intranasal, intracerebral, intrathecal, intravaginal, transdermal, rectal, by inhalation, or topical.

Description:
KINASE MUTATION-ASSOCIATED

NEURODEGENERATIVE DISORDERS

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims the benefit of and priority to U.S. Provisional Patent

Application No. 62/550,536, filed August 25, 2017, the entire contents of which are incorporated herein by reference.

STATEMENT OF RIGHTS UNDER FEDERALLY-SPONSORED RESEARCH

[0002] This invention was made with government support under grant number CA008748 awarded by the National Institutes of Health. The government has certain rights in the invention.

TECHNICAL FIELD

[0003] The present disclosure relates to a method for treatment of neurodegenerative disorders.

BACKGROUND

[0004] The pathophysiology of neurodegenerative diseases is poorly understood and

therapeutic options are few. Neurodegenerative diseases are hallmarked by progressive neuronal dysfunction and loss, and chronic glial activation. Whether microglial activation, which is viewed in general as a secondary process, is harmful or protective in neurodegeneration remains unclear.

[0005] BRAF is a serine/threonine-protein kinase that is part of the RAS/MAPK/ERK signaling pathway, which affects cell senescence or proliferation, differentiation, and secretion, depending on the cell type. The BRAF V600E point mutation results in constitutive ERK activation, and is present in numerous tumors including melanomas, thyroid, colon and liver carcinoma, and hairy cell leukemia (HCL), as well as in clonal macrophage disorders known as histiocytoses. Histiocytoses display considerable heterogeneity in terms of prognostic and clinical presentation, and are characterized by the occurrence of neurodegenerative syndromes. Microglia belong to the lineage of tissue macrophages that develop during organogenesis from yolk-sac erythro- myeloid progenitors (EMPs) distinct from haematopoietic stem cells. However, the cellular consequences of BRAF V600E expression in microglia and the role of BRAF inhibitors for treating neurodegenerative disease have not been investigated. SUMMARY

[0006] The technology of the present disclosure is based on the observation that the conditional expression of a BRAF V600E allele in a small number of erythro-myeloid progenitors (EMPs) does not grossly affect embryonic development, but results in the accumulation of BRAF V600E macrophage clones in various tissues and causes neurodegeneration.

[0007] By developing novel genetically and phenotypically accurate murine models of disease, it is possible to comprehensively explore the effects of BRAF V600E expression in tissue macrophages and other myeloid cells. The animal model described herein overcomes a limitation of previous murine models where constitutive expression of Cre resulted in a very high frequency of cells expressing BRAF V600E within hematopoietic cells of different lineages, which may not accurately model the behavior of a limited number of BRAF V600E progenitors of a particular hematopoietic lineage in competition with wild type progenitors in patients.

[0008] In one aspect, the present disclosure provides a method for treating or preventing BRAF V600E -associated neurodegenerative disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a BRAF, MEK, and/or CSF-IR inhibitor or a pharmaceutically acceptable salt thereof. In some embodiments, at least a portion of the resident macrophages in the central nervous system of the subject are BRAF V600E+ .

[0009] In one aspect, the present disclosure provides a method for treating or preventing BRAF V600E -associated neurodegenerative disease comprising: (a) isolating resident macrophages from a neuronal environment of the subject; (b) determining whether the resident macrophages express BRAF V600E+ ; and (c) administering to the subject a therapeutically effective amount of a BRAF, MEK, and/or CSF-IR inhibitor, or a pharmaceutically acceptable salt thereof, when the isolated resident macrophages express BRAF V600E+ .

[0010] In some embodiments of the methods disclosed herein, the neurodegenerative disease is characterized by one or more of impaired cognitive functions, dementia, ataxia, dysarthria, reduced motor coordination and synchrony as compared to a normal control subject, paralysis, microglia accumulation, astrogliosis, microglia phagocytosis, demyelination, neuronal loss in the central nervous system, synaptic loss in the central nervous system, and amyloid precursor protein (APP) deposits in the brain.

[0011] In some embodiments of the methods disclosed herein, the BRAF inhibitor is selected from the group consisting of vemurafenib, dabrafenib, encorafenib, PLX7904, PLX8394, GDC- 0879, LGX818, and PLX4720, the MEK inhibitor is selected from the group consisting of AZD8330, refametinib, E6201, MEK162 (binimetinib), PD0325901, pimasertib, R04987655, selumetinib, TAK-733, GDC-0623, WX-544, cobimetinib, and trametinib, and the CSF-1R inhibitor is selected from the group consisting of GW2580, BLZ945, pexidartinib (PLX3397), ARRY-382, PLX7486, and JNJ-40346527. In some embodiments, the BRAF inhibitor is vemurafenib. In some embodiments, the BRAF inhibitor is PLX4720.

[0012] In some embodiments of the methods disclosed herein, the route of administration of the BRAF, MEK, or CSF-1R inhibitor is parenteral, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, oral, sublingual, intranasal, intracerebral, intrathecal, intravaginal, transdermal, rectal, by inhalation, or topical.

[0013] In some embodiments of the methods disclosed herein, treatment of the

neurodegenerative disease comprises one or more of improving cognitive functions, reducing dementia, reducing ataxia, reducing dysarthria, increasing motor coordination and synchrony, relieving paralysis, reducing microglia accumulation, reducing astrogliosis, reducing microglia phagocytosis, reducing demyelination, reducing neuronal loss, reducing synaptic loss, or reducing amyloid precursor protein (APP) expression in the brain as compared to an untreated control.

[0014] In one aspect, the present disclosure provides a method for treating or preventing neurodegenerative disease in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a PI 3 -kinase inhibitor or a pharmaceutically acceptable salt thereof, wherein at least a portion of the resident macrophages in the central nervous system of the subject comprise one or more PI 3-kinase mutations. In some embodiments, at least a portion of the resident macrophages in the central nervous system of the subject are

PIK3CA H1047R+ . In some embodiments, the PI 3-kinase inhibitor is selected from the group consisting of idelalisib, BKM120, GDC-0980, PF-04691502, XL147, IPI-145, BYL719, SF1126, BAY80-6946, GSK2126458, NVP-BEZ235, GDC-0941, PX-866, XL765, and ZSTK474.

[0015] In some embodiments of the methods disclosed herein, the route of administration of the PI 3-kinase inhibitor is parenteral, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, oral, sublingual, intranasal, intracerebral, intrathecal, intravaginal, transdermal, rectal, by inhalation, or topical.

[0016] In another aspect, the present disclosure relates to non-human animals, for example rodents, that conditionally express a mutant BRAF allele that confers a pathological phenotype on the non-human animal expressing the allele, embodiment, the pathological phenotype is histiocytosis.

[0017] In one embodiment, the non-human animal of the disclosure is characterized by expression of BRAF V600E in erythromyeloid progenitors.

[0018] In another aspect, the non-human animals comprise a mutant BRAF allele flanked upstream and downstream with site-specific recombinase recognition sites (SRRSs), and the non-human animal comprises a recombinase that recognizes the SRRSs, wherein the

recombinase is inducible.

[0019] In a another aspect, the present disclosure relates to a genetically modified mouse that comprises a nucleic acid construct comprising a mutant exon encoding a BRAF V600E mutation, wherein the mutant is flanked upstream and downstream by SRRSs and the mouse comprises an inducible recombinase gene encoding a recombinase. In one embodiment, the SRRSs are recognized by the inducible Cre recombinase.

[0020] In one aspect, the present disclosure relates to a genetically modified mouse comprising the genotype Csflr' Cre - BRAF V600E ; Rosa26 LSL - YFP .

[0021] In another aspect, the present disclosure relates to a method for recapitulating

development of neurodegeneration in clonal histiocytic disorders comprising: (a) providing a transgenic mouse whose genome comprises a BRAF V600E transgene and a Rosa26 LSL'YFP trmsgene, the transgenes operably linked to a tamoxifen-inducible regulatory sequence for expression of BRAF V600E and YFP in erythro-myeloid progenitors (EMPs) of said mouse, and Csflr Mer,CreMer ; (b) contacting said mouse in utero with 4-hydroxy-tamoxifen (OH-TAM) wherein expression of BRAF V600E and Rosa26 LSl - YFP is induced in EMPs of said mouse. The mouse (embryo) is exposed to OH-TAM at E8.5.

BRIEF DESCRIPTION OF THE DRAWINGS

[0022] FIG. 1 is a schematic showing that the erythro-myeloid progenitors (EMP) from the yolk sac colonize the fetal liver and give rise to macrophage (ΜΦ) precursors that colonize the embryo from E9.5 in a Cx3crl -dependent manner, to give rise to adult F4/80+ resident macrophages. ΜΦ specification, starting from El 0.25, is initiated by the expression of tissue- specific transcriptional regulators. [0023] FIG. 2 is a schematic showing the developmental diversity of the myeloid system. EMPs emerge in the yolk sac at E8.5, migrate to the fetal liver, and give rise to fetal

macrophages in vivo. Resident macrophages develop from EMPs in the absence of Myb and persist as residents in post-natal tissues. Within the embryo proper, the hemogenic endothelium of large arteries gives rise to Hematopoietic Stem Cells (HSCs) at -E10.5. HSCs migrate to the fetal liver and to the bone marrow, where they persist, self-renew throughout life, and give rise to adult-type red blood cells, lymphoid cells and short-lived myeloid cells.

[0024] FIGs. 3A-3D show constitutive expression of BRAF V600E in Csflr+ cells. (FIG. 3A) is a graphic showing the breeding scheme. (FIG. 3B) Embryonic lethality of Csflr lCre +;

BRAF Y600E ; Ro S a26 LSL - YFP mice, black bars represent the % of mice born from the cross depicted in (FIG. 3A) according to their genotype (n=39). (FIG. 3C) Brightlight (upper panel) and epifluorescence microscopy (lower panel) of Q 7r iCre +; BRAF WT ; Rosa26 LSL'YFP and Q 7r iCre +; BRAF Y600E Rosa26 LSL'YFP embryos showing hemorrhagic foci in the liver (arrow) and accumulation of YFP+ cells in fetal liver. Cross (†) indicates dead embryo. (FIG. 3D)

Q 7r iCre +; BRAF Y600E Rosa26 ESL'Y¥P mice are associated with 100% lethality beyond E14.5.

[0025] FIG. 4 shows Tamoxifen-inducible mouse models used to target EMP versus HSC. Csfl Mer,CreMer +; Rosa26 LSL - YFP do not target HSCs in adult mice, while pulse-labeling of

Cxcr4 CreERT2+ ; Rosa26 LS ^ YFP results in labeling of 10% HSCs. LSK: Lin-Scal+Kit+.

[0026] FIGs. 5A-5E show the results of experiments with a Csflr MeriCreMer ; BRAF V600E ;

Rosa26 LSL"YFP inducible model. (FIG. 5A) Breeding scheme. (FIG. 5B) % of mice born from the cross depicted in (FIG. 5A) according to their genotype (n=201). (FIGs. 5C and 5D) Flow cytometric analysis and IF analysis of livers of 4-week old mice. YFP+ cells are F4/80hi CDl lblo (gated on CD45+ cells). (FIG. 5E) Histologic analysis of the brain. Clusters of YFP+ cells in the cerebellum and brainstem. YFP+ cells also stain for CD68 (and Ibal, not shown).

[0027] FIGs. 6A-6D show neurological disease in the Csflr MeriCreMer ; BRAF V600E ; Rosa26 LSL" YFP model. (FIG. 6A) Test of limb-clasping reflexes of 6-8-month old mice. When lifted by the tail BRAF WT mice behave normally, extending their hind limbs. In contrast, BRAF V600E mice bend their legs towards their trunk. (FIG. 6B) Cumulative incidence rate of behavioral abnormalities. (FIG. 6C) Footprint assay assessing locomotion defects. Front paws were painted with red ink, hind paws with blue ink. BRAF V600E mice lose coordination; front/hind legs do not overlap, and later mice are paralyzed (far right panel). (FIG. 6D) Measurement of footprint assay. Numbers below graphs correspond to the measurements performed as displayed in (FIG. 6C)

[0028] FIGs. 7A and 7B show histological analysis of the spinal cord in Cre+ BRAF V600E mice and littermate controls. (FIG. 7A) Ibal+ microglia accumulate in the white matter. (FIG. 7B) Flow cytometry analysis of F4/80+ macrophages from the spinal cord. Littermates are color- coded.

[0029] FIGs. 8A and 8B show demyelination of the spinal cord of Cre+ BRAF V600E mice compared to wild type mice. (FIG. 8A) Luxol fast blue stain shows demyelination and leukocyte infiltration in the white matter of the spinal cord. (FIG. 8B) Flow cytometry analysis shows that CD3+ cells accumulate in the spinal cord in Cre+ BRAF V600E mice.

[0030] FIGs. 9A and 9B show histological analysis of the spinal cord from 6-month old BRAF V600E and control mice. Immunostaining was performed with CD68, GFP, and pERK antibodies.

[0031] FIG. 10 shows transcription factors that are differentially expressed between macrophages from the brain, liver, skin, kidney, and lung during development and in postnatal mice.

[0032] FIGs. 11A-11C show the analysis of the liver in 6-8 month old Csflr MeriCreMer ;

BRAF V600E ; Rosa26 LSL"YFP and littermates. (FIG. 11 A) Flow cytometry analysis of total and YFP+ Kupffer cells. Littermates are color-coded. (FIG. 11B) Histological analysis shows accumulation of YFP+ Kupffer cells (stained with a cross-reactive GFP antibody). (FIG. 11C) Serum analysis of liver enzymes.

[0033] FIG. 12 shows cumulative incidence rate of behavioral abnormalities in Csflr MenCreMer ; BRAF V600E ; Rosa26 LSL"YFP mice placed on long-term PLX4720 diet (PLX diet) at 3 months of age, and mice on normal control diet (Ctrl diet).

[0034] FIGs. 13A-13I. Targeting BRAF V600E in tissue-resident macrophages (FIGs. 13A and 13B) Breeding scheme for experimental mice and genotype distribution (n=342). (FIGs. 13C and 13D) YFP expression on BM LSK, blood leukocytes and microglia from 1 -month-old mice, representative of n=5 per group. (FIG. 13E) Proportion of YFP + F4/80 + cells in tissues from 1 -month-old mice. Circles represent individual mice. Unpaired two-tailed t-test. (FIG. 13F) A>T transversion encoding BRAF V600E in YFP + Kupffer cells at the Braf locus. Red and blue bars indicate forward and reverse strands. (FIG. 13G) Ki67 and cleaved Caspase 3 (Casp3) expression in YFP + microglia from 1 -month-old brains. n=5 per group. Unpaired two-tailed t- test. (FIG. 13H) GSEA of differentially expressed genes in YFP + microglia from BRAF VE (n=3) and littermates (n=2) mice, q-value < 0.01. (FIG. 131) Heatmap representation of selected genes from (FIG. 13H), values are displayed as z-score. See also FIG. 17.

[0035] FIGs. 14A-14H. Neurodegenerative disease in BRAF VE mice (FIG. 14A) Footprint assays. Front and hind paws are painted with red and blue ink, respectively, n = 20 mice per group. (FIG. 14B) Limb-clasping reflexes in 6-8-month old mice. n=10 per group. (FIG. 14C) Cumulative incidence of behavioral abnormalities in BRAF VE mice and controls. Log-rank (Mantel-Cox) test. (FIG. 14D) Overlap distance, and stride length of mice on control or PLX4270 diet from 1 month or 3 months of age. Average values ±s.d. for each group, 2way ANOVA, *p<0.05, **p<0.01, ***p<0.001. (FIG. 14E) Disease progression in mice from (d), average score excluding mice euthanized for paralysis († ). (FIG. 14F) Cumulative incidence of behavioral abnormalities in mice from (d). Log-rank (Mantel-Cox) test (FIG. 14G) Scheme depicting microglia (Ibal) and neuronal (NeuN) densities in brain regions from BRAF VE and BRAF m (n=4 per group). (FIG. 14H) Ibal staining and quantitative analysis of microglial accumulation (Ibal+ ), phagocytosis (Ibal+ /Lamp2+ ), astrogliosis (GFAP+ 308 ), relative synapse density (determined by Synaptophysin and Homerl), neuronal loss (NeuN) and amyloid precursor protein (APP) in brainstem from 5-9 month-old BRAF VE mice on control diet (n=4), BRAF VE mice on PLX diet (n=4-6), and BRAF m (n=4). One-way ANOVA. See also FIGs. 20 and 22

[0036] FIGs. 15A-15G. ERK activation in BRAF V600E microglia. (FIG. 15A) CD68, YFP and pERK staining in spinal cord from 7-month old mice. Scale bars=500μm, ΙΟμιη for insets. n=4 per group. (FIG. 15B) pERK + microglia in brainstem. Circles represent individual mice. One-way ANOVA. (FIG. 15C) ERK phosphorylation in spinal cords and brains from 6-9 month-old mice. Top: representative western blot, bottom: pERK/ERK ratio, n=5 per group. One-way ANOVA. (FIG. 15D) pERK expression in YFP + microglia from BRAF VE mice. n=5 per group. Scale bars=5μm. (FIG. 15E) Numbers of microglia from 5-9 month-old mice Circles represent individual mice. One-way ANOVA. (FIG. 15F) Heatmap representation of cell frequency among CD45 + in the brain littermates. n=3 per group. (FIG. 15G) Ki67 + and cleaved Caspase 3 + (Casp3) expression in YFP + microglia from 5-9 month-old BRAF VE mice. n=6 per group. Unpaired two-tailed t-test. See also FIG. 23.

[0037] FIGs. 16A-16I. Molecular features of ERK-activated microglia, and their presence in histiocytoses patients. (FIG. 16A) GSEA of differentially expressed genes in YFP + microglia from 6-7 month-old littermates. q-value <0.05. EMT: epithelial-mesenchymal transition. (FIG. 16B) Heatmap representation of selected DEG, values are displayed as z-score. (FIG. 16C) Validation of gene expression in YFP + microglia. n=5 per group. FMO:

fluorescence minus one. (FIG. 16D) Illb expression in spinal cords from 6-7 month-old mice. n=3 per group. Scale bars=10 μιη. (FIG. 16E) Bioplex analysis of Illb and 1117a in spinal cords from 6-9 month-old mice. Circles represent individual mice. Unpaired two-tailed t-test. (FIG. 16F) Collagen IV and collagen VI expression in spinal cord from (FIG. 16D). n=3 per group. Scale bars=l(^m. (FIG. 16G) CD163, pERK and BRAF V600E expression in Erdheim-Chester disease (ECD) brain tissue. Scale bar upper panels: 40μιη, lower panels: 5μιη. (FIG. 16H) Quantification of pERK microglia in control (n=6) and ECD (n=3) brains. Mann-Whitney test. (FIG. 161) Heatmap representation of selected genes from RNA-seq analysis of brain tissue from 5 control brains and 2 histiocytoses patients (Juvenile Xanthogranuloma (JXG) and

Langerhans Cell Histiocytosis (LCH)), values are displayed as z-score. q-value <0.01. See also FIG. 25

[0038] FIGs. 17A-17H. Analysis of one-month old Csflr Me CreMer ; BRA p LSL~ V600E .

LSL YFP

Rosa26 ' mice. (FIG. 17A) % of mice born from the cross depicted in Figure 13a according to their genotype (n=42), but no injection of hydroxy -tamoxifen (4-OHT) to test for adverse effects of 4- OHT administration. (FIG. 17B) Flow cytometry analysis of YFP expression on blood leukocytes. Representative for n=8 per genotype. (FIG. 17C) Flow cytometry analysis of YFP + cells in the liver. YFP + cells, present only in Q s f-i l - enCveMev+ (Cre + ) mice (upper panels), fall into the F4/80 + CDl lb + Kupffer cell gate (lower panels). Representative for n=8 per genotype. (FIG. 17D) YFP expression by immunofluorescence in the liver of BRAF VE mice and BRAF WT . YFP + cells are F4/80 Kupffer cells. Representative of n=6 mice per genotype. Scale bars=200 μπι (5 μπι for insets). (FIG. 17E) Total tissue-resident macrophages cell numbers per gram (FIG. 17G) of tissue were analyzed by flow cytometry in BRAF VE mice (n=4) and BRAF m (n=6). Circles represent individual mice. Unpaired two-tailed t-test. (FIG. 17F) In situ analysis of phospho-Hi stone H3 (pHis3) staining in YFP + cells from brains oiBRAF VE and BRAF m Circles represent individual mice (n=3). Unpaired two-tailed t- test. (FIG. 17G) RNA-seq analysis, heatmap representation of MAPK target genes in YFP + microglia from BRAF VE (n=3) and BRAF WT (n=2) mice, values are displayed as z-score. (FIG. 17H) Histological analysis of liver, lung, kidney and spleen in BRAF VE mice and BRAF m Representative of n=4 mice per genotype. Scale bars=10C^m (ΙΟμπι for insets).

[0039] FIGs. 18A-18F. Effect of constitutive BRAF V600E expression in Q/2r-expressing cells. (FIG. 18A) Breeding scheme. (FIG. 18B) Embryonic lethality of Csflr' Cre+ ; BRAF LSL~ V600E . R osa 26L SL - YFP mice, black bars represent the % of mice born from the cross depicted in (FIG. 18 A) according to their genotype (n=39). (FIG. 18C) Brightlight (upper panel) and epifluorescence microscopy (lower panel) of Csflr ,Cre+ BRAF VE and Csflr ,Cre+ ; BRAF WT embryos showing haemorrhagic foci in the liver (arrow) and accumulation of YFP + cells in fetal liver. † indicates dead embryo. Pictures are representative of n=3 per genotype. (FIG. 18D)

Mouse embryos found alive during different developmental stages. Csfl^^ ; BRAF LSL'V600E ; Rosa26 LSL - YFP mice are associated with 100 % lethality beyond E14.5. (FIG. 18E) Liver weight in E12.5 embryos. Circles represent individual mice. n=8 for WT; Cre " , n=14 for VE; Cre " , n=16 for VE; Cre " , n=12 for VE; Cre + . One-way ANOVA. (FIG. 18F) Flow cytometry analysis of Lin " Kit + blast, erythroid cell (Terl 19) and hematopoietic stem cell numbers (LSK

CD150 + CD48 " and CD150 " CD48 " ) in the E12.5 fetal liver and of E12.5 tissue-resident macrophages in the limbs, head and liver. Circles represent individual mice. n=4 for BRAF m and n=6 ioxBRAF 1 ^. Unpaired two-tailed t-test.

[0040] FIGs. 19A-19G. Analysis of CDllc Cre ; BRAF V600E mouse model. (FIG. 19A)

Kaplan-Meier survival curve ofBRAF VE (n=16) and BRAF m (n=66) controls. Log-rank (Mantel- Cox) test. (FIG. 19B) Representative photographs of lung and spleen from BRAF VE mice at time of death with representative BRAF WT control organs. (FIGs. 19C and 19D) Hematoxylin and eosin (HE) stain of lung tissue from BRAF VE and littermate controls. (FIG. 19E) CD68 immunohistochemistry (IHC) of BRAF VE lung tissue. (FIG. 19F) HE stain of liver tissue from BRAF VE and littermate controls with magnified view of granuloma in the BRAF VE liver. (FIG. 19G) HE stain of bone marrow (BM) from BRAF VE and littermate controls with CD68 IHC of BRAF VE mouse. All pictures for (FIGs. 19B-19GA) are representative of n=5 per genotype .

[0041] FIGs. 20A-20G. Longitudinal study and PLX treatment of the

Cs f l r MeriCreMer . BRA pV600E . Rosa26 LSL - YFP mice . ( FIGs . 20A and 20B) Latency to fall in the rotarod assay and footprint assay quantification for BRAF VE mice (n=7) md BRAF^ in^). (FIG. 20A) Rotarod assay at 1 to 4 months of age. Values are mean ± s.d. (FIG. 20B) Rotarod and footprint assay at 4 months of age displaying single values. Mice that are score 1 are labeled in red. (FIG. 20C) Footprint assay quantification of BRAF VE mice at score 1 and littermate controls. Circles represent individual mice. n= 10 for BRAF m and n=l 1 for BRAF VE . (FIG. 20D) Representative weight curve oiBRAF^ and BRAF VE mice on control or PLX4720 diet. (FIG. 20E) PLX4720 concentration in serum (ng/ml), liver and brain (ng/g) of 7-9 month old BRAF m (n=9) and BRAF VE mice placed on the diet at 1 (n=8) or at 3 months (n=3) of age.

Circles represent individual mice. (FIG. 20F) Footprint assay quantification from BRAF VE mice on PLX4720 diet at 1 month (n=8) or at 3 months (n=6) and control (Ctrl) diet (n=13) and BRAF WT (n=32, black). Mice reaching paralysis were excluded from further analysis. See also (FIG. 20G) where † indicates when BRAF VE animals were euthanized. Values are mean ± s.d. 2way ANOVA comparing treated and not treated BRAF VE mice. * p<0.05, **p<0.01,

*** p<0.001. (FIG. 20G) Disease progression for BRAF mice on control or PLX4720 diet. † indicates animal death due to paralysis.

[0042] FIGs. 21A and 21B. Microglia activation in the brain starts at early, preclinical stages. (FIG. 21A) Histological analysis by hematoxylin and eosin (HE) and Luxol-fast-blue-PAS (LFB- PAS) and immunohistochemistry analysis of T-cells (CD3), B- cells (B220) and astrocyte activation (GFAP) in one-month old BRAF VE mice and BRAF^. Representative of n=5 per for BRAF WT &ηά n=4 for BRAF 1 ^. (FIG. 21B) Immunohistochemistry analysis and quantification of Ibal + cell density, cortical neurons (NeuN) and expression of amyloid precursor protein (APP), a positive signal for neurodegeneration in one-month old BRAF VE mice and BRAF WT . Representative of n=5 per for BRAF WT and n=4 for BRAF VE . Circles represent individual mice. Scale bars=10C^m (ΙΟμπι for insets). Unpaired two-tailed t- test.

[0043] FIGs. 22A-22D. Neurodegenerative process in BRAF VE mice (FIG. 22A) Ibal and GFAP immunohistochemistry of brain and spinal cord from 6-month-old BRAF VE and BRAF WT . Anatomical regions of insets are indicated. Representative for n=5 BRAF m and n=4 BRAF VE . S cal e b ars= 50C^m for spinal cords and 1 mm for b rains (50 μπι for insets) . (FIG. 22B) Immunohistochemistry and immunofluorescence as used for quantification in FIG. 14H of brain stem for NeuN (neurons), APP (amyloid precursor protein) and GFAP (astrocytes), Ibal + /Lamp2 + cells (phagocytosis), Synaptophysin (Syn) and Homerl (synapse density) and staining with Luxol-fast-blue (LFB)-PAS. Scale bars=10C^m (ΙΟμπι for insets). Ibal/Lamp2 scale bar=25μm, Syn/Homerl scale bar=lC^m. Representative for 6-9 month old BRAF m (n=5), BRAF VE (n=4), and for BRAF VE on PLX diet (n=4-6). (FIG. 22C) LFB staining of spinal cord samples from (FIG. 22A). Scale bar=10C^m. (FIG. 22D)

Immunohistochemistry of brain stem for B220 (B-cells) from BRAF VE on control and PLX diet. Representative for n=4 per genotype. Scale bars=10 μπι(10μιη for insets).

[0044] FIGs. 23A-23G. Microglia and T-cell phenotype in BRAF VE mice.

(FIG. 23A) Representative pERK staining in Ibal + microglia as used for the quantification in

FIG. 15B in brain stem of 5-9 month old BRAF^ and BRAF^ mice on control or PLX diet. Scale bar=50 μπι. (FIG. 23B) Representative tSNE analysis of flow cytometry staining of CD45 + cells from the brain of paralyzed BRAF 1 ^ mice and littermate controls. Arrow indicates expansion of F4/80 + YFP + cells. Representative for n=3 per genotype. (FIG. 23C) FSC profile of YFP + and YFP " microglia from (FIG. 23B) from BRAF VE and BRAF m mice indicates an increase of YFP + microglia cell size. Representative for n=3 per genotype. (FIG. 23D) Proportion of YFP + F4/80 + cells in indicated organs analyzed by flow cytometry. The proportion of YFP + among F4/80 + cells from Cre + BRAF WT on control diet was normalized and set to 1. Analysis was performed on 5-8 month-old BRAF VE mice (n=5-6) and BRAF m mice (n=6) on control diet, and 7-9 month-old BRAF VE mice (n=6) and BRAF m mice (n=4) on PLX diet. Circles represent values for individual mice. One-way ANOVA. * p<0.05, **p<0.01, ***p<0.001. (FIG. 23E) CD3 immunohistochemistry of brain and spinal cord from 6-month-old BRAF VE and BRAF WT .

Anatomical regions of insets are indicated. Representative for n=5 BRAF WT and n=4 BRAF VE . Scale bars=50C^m for spinal cords and 1mm for brains (50μπι for insets). (FIGs. 23F and 23G) Analysis of CD8 + , CD4 + and Foxp3 + T-cell numbers (FIG. 23F) and proliferation (FIG. 23G) in brain/spinal cord by flow cytometry in 5-8 month-old BRAF VE (n=4) and BRAF m (n=6) on control diet, and 7-9 month-old BRAF VE (n=6) and BRAF WT (n=5) on PLX diet. Circles represent values for individual mice. One-way ANOVA.

[0045] FIGs. 24A-24F. BRAF VE mice analysis outside the central nervous system (FIG. 24A) Proportion of YFP + F4/80 + cells in indicated organs from analyzed by flow cytometry. The proportion of YFP + among F4/80 + cells from Cre + BRAF m (n=6) was normalized and set to 1. Circles represent values for individual BRAF VE mice (n=7). Unpaired two-tailed t-test. (FIG. 24B) Analysis of liver Kupffer cells as in (FIG. 24A) was performed on 5-8 month-old BRAF VE (n=5) and BRAF m (n=4) on control diet, and 7-9 month-old BRAF VE (n=6) and BRAF m mice (n=4) on PLX diet. Circles represent values for individual mice. One-way ANOVA. * p<0.05, **p<0.01 , ***p<0.001. (FIG. 24C) Immunofluorescent analysis of pERK in F4/80 + Kupffer cells from 5-8 month-old BRAF VE . Results are representative for n=3. (FIG. 24D)

Serum analysis of BRAF VE mice (score 1, n=6) and their littermates controls (n=6). ALB: albumin, ALP: alkaline phosphatase, ALT: alanine aminotransferase, AST: aspartate aminotransferase. (FIG. 24E) Gross liver, lung, kidney and spleen structure (HE,

Trichrome) of paralyzed BRAF VE and BRAF WT . Representative of n=7 per genotype. Scale bars=20C^m (Ι Ομπι for insets). (FIG. 24F) Liver and spleen gross organs from paralyzed BRAF VE and BRAF m Representative of n=5 per genotype.

[0046] FIGs. 25A-25C. Erdheim-Chester disease (ECD) patients (FIG. 25A) Table summarizing observed pathological and molecular findings in brain tissue of three ECD patients with neurologic presentations. BRAF status was determined by immunohistochemical analysis and by sequencing. Neuronal loss and demyelination was determined by immunohistochemistry of neurofilament and myelin basic protein (MBP). RF: Rosenthal fiber, n/a: not applicable/no tissue available for further analysis. (FIG. 25B)

Immunohistochemistry and immmunofluorescent analysis of ECD brain tissue for CD163, pERK and BRAF V600E (anti-BRAFVEl antibody). Upper panel scale bar=20C^m. Lower panel scale bar=5μm. (FIG. 25C) Immunohistochemistry analysis of ECD brain tissue for neurofilament and MBP shows areas of myelin deficits with preserved axons in the same region. Scale bar=20C^m .

[0047] FIGs. 26A-26C. Isolation of microglia, neuron, and glia nuclei by cell sorting in a patient with neurodegeneration. (FIG. 26A) Nuclei extracted from -350 mg of fresh frozen control brain tissue using a non-ionic detergent buffer containing DAPI and flow-sorted using anti-NeuN and anti-Pu. l . DN: double negative. (FIG. 26B) Top panel: Nuclei extraction and flow-sorting using anti-Pu.1 and anti-NeuN antibodies was performed in four different brain regions, two affected and two non-affected, obtained from a deceased patient with histiocytosis and neurodegenerative disease. Double negative (DN), NeuN+, and Pu.1+ populations were sorted for sequencing. Patient's blood was separated based on Lin+ (lymphocytes) and Lin- HLA-DR+ (myeloid cells, not shown). Bottom panel: FACS graph of nuclei isolated from 3 patients with Alzheimer's disease (AD), Parkinson's disease (PD), and Adult-Onset- Leukoencephalopathy with spheroids (ALSP). (FIG. 26C) Example of quality control of DNA obtained from AD patient. DNA is immediately isolated after nuclei sorting with QIAamp DNA Mini Kit (Qiagen). DNA quality is determined based on DNA Integrity Number (DIN) measured by Agilent 4200 TapeStation.

[0048] FIGs. 27A-27B. Analysis of BRAF V600E mutation in microglia, neuron, and glia nuclei from a 25-year old patient with neurodegeneration. (FIG. 27A) BRAF V600E allele frequency determined ddPCR in sorted nuclei from 8 brain areas from 1 patient with

neurodegeneration, whole brain tissue and blood were processed for gDNA extractions using QIAamp DNA Mini Kit (Qiagen) and quality and quantity was determined with Agilent 4200 TapeStation. BRAF V600E allele frequency is determined by ddPCR in the same DNA samples before library preparation. Multiple numbers indicate independent determinations. Counts <5 are not reproducible and below sensitivity level. (FIG. 27B) BRAF V600E allele frequency determined by targeted deep sequencing (HEME-PACT, mean sequencing depth 409x) in sorted nuclei from four of the above brain areas, whole brain tissue, and blood. Approximately 200 ng of DNA were used for library preparation with KAPA HyperPrep Kit (Roche). Libraries are then captured with custom-designed biotinylated probes (NimbleGen) and sequenced with Illumina HiSeq 2500. Counts < 4 (1%) are considered below sensitivity level. [0049] FIG. 28. Hippocampus of patient with neurodegenerative disease due to

BRAF V600E microglia. Immunohistochemistry analysis for CD163, Neurofilament- 1 (NF), and GFAP of post-mortem brain tissue from the hippocampal area.

[0050] FIG. 29. Preliminary analysis of somatic mosaicism from Heme-PACT targeted sequencing. Venn diagrams represent the number and repartition among cell types and tissues (brain regions and blood) of somatic variants (SNV and indel) detected by Heme-PACT sequencing and analysis. Variants with an allele frequency >35% are considered germline and eliminated from analysis. SNVs are validated bioinformatically and by ddPCR and Ampli-seq. This analysis suggests that mutations are either common to all tissues ($, with high 15%-35% allelic frequency) and considered early somatic events, or local to a cell type and brain region (with lower allelic frequencies) for neurons, microglia, and DN (double negative) cells suggesting their local development independent of blood cells. The 2 mutations common to hippocampus and cerebellum (*) are at higher allelic frequencies and correspond to BRAF V600E (11% and 21%) and a non-coding mutation in MSH6 (16% and 26%).

[0051] FIG. 30. Breeding strategy for experimental mice and genotype distribution (n=22). 4- OHT: 4-hydroxytamoxifen. E8.5, embryonic day 8.5.

[0052] FIGs. 31A-31B. Neurodegenerative disease in PIK3CA HR mice. (FIG. 31A)

Cumulative incidence of behavioral abnormalities in PIK3CA HR mice and control. (FIG. 31B) Footprint assay example, which measures the overlap distance between hind and front paws.

DETAILED DESCRIPTION

[0053] All patents, published applications and other publications and references are hereby incorporated by reference in their entirety into the present disclosure.

[0054] It is to be appreciated that certain aspects, modes, embodiments, variations and features of the present technology are described below in various levels of detail in order to provide a substantial understanding of the present technology.

Definitions

[0055] The definitions of certain terms as used in this specification are provided below. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this present technology belongs. [0056] As used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the content clearly dictates otherwise. For example, reference to "a cell" includes a combination of two or more cells, and the like.

[0057] As used herein, the term "about" encompasses the range of experimental error that may occur in a measurement and will be clear to the skilled artisan.

[0058] The embryonic stages used herein relate to embryonic day (for example Dl is El .0) of mouse development. This staging by "days" relate to in the female presence of a vaginal plug indicating that the mating occurred.

[0059] As used herein, "prevention," "prevent," or "preventing" of a disorder or condition refers to one or more compounds that, in a statistical sample, reduces the occurrence of the disorder or condition in the treated sample relative to an untreated control sample, or delays the onset of one or more symptoms of the disorder or condition relative to the untreated control sample.

[0060] As used herein, the terms "subject," "individual," or "patient" can be an individual organism, a vertebrate, a mammal, or a human.

[0061] As used herein, a "therapeutically effective amount" of a compound refers to compound or agent levels in which the physiological effects of a disease or disorder are, at a minimum, ameliorated. A therapeutically effective amount can be given in one or more administrations. The amount of a compound which constitutes a therapeutically effective amount will vary depending on the compound, the disorder and its severity, and the general health, age, sex, body weight and tolerance to drugs of the subject to be treated, but can be determined routinely by one of ordinary skill in the art.

[0062] "Treating," "treat," or "treatment" as used herein covers the treatment of a disease or disorder described herein, in a subject, such as a human, and includes: (i) inhibiting a disease or disorder, i.e., arresting its development; (ii) relieving a disease or disorder, i.e., causing regression of the disorder; (iii) slowing progression of the disorder; and/or (iv) inhibiting, relieving, or slowing progression of one or more symptoms of the disease or disorder.

[0063] It is also to be appreciated that the various modes of treatment or prevention of medical diseases and conditions as described are intended to mean "substantial," which includes total but also less than total treatment or prevention, and wherein some biologically or medically relevant result is achieved. The treatment may be a continuous prolonged treatment for a chronic disease or a single, or few time administrations for the treatment of an acute condition.

[0064] The term "administer," "administering," or "administration" as used in this disclosure refers to either directly administering a therapeutic agent, such as, but not limited to, a BRAF, MEK, or CSF-1R inhibitor, or a combination of thereof, to a subject.

[0065] In certain embodiments, routes of administration include, for example: intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, oral, sublingual, intranasal, intracerebral, intravaginal, transdermal, rectally, by inhalation, or topically, particularly to the ears, nose, eyes, or skin. In some embodiments, the administering is effected orally or by parenteral injection. The mode of administration can be left to the discretion of the practitioner, and will depend in-part upon the site of the medical condition. In most instances, administration results in the release of any agent described herein into the bloodstream. In specific embodiments, it may be desirable to administer locally to the area in need of treatment or blocking.

[0066] The term "neuronal environment" refers to "tissue" or cellular niche where the resident macrophages are found.

General

[0067] The technology of the present disclosure relates to the observation that conditional expression of a BRAF V600E allele in EMP clones in vivo does not grossly affect embryonic development, but results in the accumulation of BRAF V600E macrophage clones in various tissues, and is responsible for a neurodegenerative syndrome in adult mice that recapitulates for the first time one of the most poorly understood and adverse characteristics of human histiocytic neoplasms. Characterizing a yolk sac origin for Erdheim-Chester disease (ECD) and Langerhans Cell Histiocytosis (LCH) carries significant implications for the pathophysiology of these diseases, and in particular may illuminate the cause of neurodegenerative syndromes as well as liver/lung fibrosis in these disorders, which are only partially sensitive or even refractory to current therapy.

[0068] The involvement of somatic mutations that confer a proliferative, survival, or activation advantage (called "oncogenic" or "driver" mutations) in the clonal evolution of cancer is well known. Progress in sequencing technologies has revealed a high burden of somatic mutations, including driver mutations, in normal (non-tumoral) tissues and their association with neurodevelopmental disorders, neurodegeneration, and non-cancerous proliferative diseases. As an example, inherited mutations that activate the RAS/MAPK/ERK pathway are incompatible with life, or cause severe and early-onset tumoral, developmental, and neurodevelopmental disorders ("RASopathies") and are therefore rarely observed. However, somatic mosaicism for such mutations is frequently observed in "normal" tissues and results in distinct phenotypes that vary according to several factors related to the time at which mutations occur, including the cellular lineage(s) harboring the mutation, the size of the clone, defects in DNA repair, and the occurrence of secondary hits. For example, mosaicism for mutations activating the RAS pathway in the early embryo can cause RASopathies, while mosaicism restricted to melanocytes is responsible for limited melanocyte proliferation and activation and give rise to the common and giant nevi. Mosaicism for mutations activating the RAS pathway in macrophages causes histiocytic lesions, but causes leukemia in hematopoietic cells.

[0069] Tissue resident macrophages such as microglia in mice originate from erythro-myeloid precursors (EMP) after gastrulation. EMP-derived cells colonize the brain during embryogenesis to give rise to microglia that self-maintain in the adult brain due to their long life span and slow local proliferation. Therefore, resident macrophages, including microglia, develop and maintain independently from the hematopoietic stem cell (HSC) lineage that give rise to blood cells. A prediction from this model was that somatic mosaicism generated during development of microglia or later in life is stable over long periods of time as microglia self-maintain rather than turnover from blood derived progenitors. To test the hypothesis that somatic mutations that confer a proliferative/survival advantage and activate macrophages ("driver" mutations) may be pathogenic, BRAF V600E mutation - the most frequent mutation that drives constitutive activation of the RAS-MEK-ERK pathway - was expressed in a mosaic manner in EMPs in mouse embryos. It was observed that somatic mosaicism for the BRAF V600E in a limited number of embryonic EMP precursors does not have detectable effects on development but results in mosaicism of mutant macrophages well tolerated in all organs, except for the brain, where positive selection of microglia mutant clones is associated with the progressive development of the cardinal features of neurodegeneration: microgliosis, astrogliosis, synaptic loss, and neuronal death. Interestingly, as shown herein, treatment of adult mice with a BRAF V600E inhibitor (PLX) slows the course of the neurological disease and reduces its mortality.

The cell lineage of origin of histiocytoses.

[0070] The previous "mononuclear phagocyte system" model, supported by bone marrow transplantation in irradiated hosts and in vitro studies over the past four decades, held that tissue macrophages originate and renew from hematopoietic stem cells (HSCs) via circulating progenitors such as blood monocytes. In contrast, recent studies have led to a major paradigm shift identifying that tissue-resident macrophages, including brain microglia, liver Kupffer cells, kidney macrophages, and Langerhans cells of the epidermis, persist in adult tissues with little contribution from bone marrow progenitors and monocytes. Instead, these cells develop during embryogenesis from yolk sac erythro-myeloid progenitors (EMP), cells distinct from HSCs {See FIGs. 1 and 2). In the model that follows from these findings, two distinct types of myeloid cells are considered: (i) tissue-resident macrophages, which contribute to cellular niches involved in tissue growth, morphogenesis, homeostasis and repair; and (ii) HSC-derived

"passenger" leukocytes, including monocytes, which are recruited to inflamed or infected tissues to perform anti-microbial and immune-regulatory functions.

HSC origin of histiocytoses is demonstrated in a small number of cases.

[0071] In recent studies in human patients, the presence of a BRAF V600E mutation in CD34 + bone marrow cells or blood monocytes was found in a small proportion of LCH patients. In murine models, previous attempts to analyze the role of BRAF V600E expression in hematopoietic cells in murine models have not succeeded in recapitulating many characteristic features of LCH and ECD. Interestingly, hematopoietic phenotypes differed greatly depending on the cell in which BRAF V600E was expressed. Constitutive expression of Cre recombinase to target the expression of BRAF V600E to HSC-derived progenitors in Vav Cve BRAF V600E mice or in all myeloid cells in Csflr ie ; BRAF V600E mice did not result in histiocytoses, but in a lethal and transplantable hematopoietic disorder in mouse embryos. Targeted expression of BRAF V600E in CD1 lc + cells - that include dendritic cell precursors in the post-natal bone marrow - also resulted in an aggressive leukemic-like disease with reduced lifespan and accumulation of circulating CD1 lc + cells in bone marrow, blood and peripheral organs. Interestingly, however, targeted expression of BRAF V600E in Langerin + cells resulted in the moderate accumulation of Langerin + dendritic cells in the lung and liver, an observation consistent with clinical features of LCH, although it does not recapitulate neurodegenerative disease or fibrotic disorders of the liver and lung, which represent key clinical problems in these diseases. As outlined below,

Langerhans cells are part of the EMP-derived myeloid lineage, thus it is possible that both HSC- derived and EMP-derived resident macrophages could give rise to histiocytoses. Tissue-resident macrophages originate at least in part from EMPs from the yolk sac.

[0072] EMPs emerge from yolk sac hemogenic endothelium at embryonic day (E)8.25. They are distinguished from HSCs by the lack of lymphoid potential, both in vitro and in vivo, the lack of long term repopulating potential, and cell surface phenotype (Seal " CD45 low AA4.1 + , CD41 + , FCYRII/III + ). EMPS express Myb but, in contrast to HSCs, their commitment and differentiation into myeloid fate is unaltered in y£-deficient embryos, although their erythroid potential is blocked. In vivo, EMPs are generated from Tie2 + yolk sac ancestors from E7.5 to E10.5. They seed the fetal liver as early as E9.5. EMPs and EMP-derived Kit + cells can be detected until E14.5. EMPs are thus distinct from HSCs, which develop from the intra-embryonic hemogenic endothelium around embryonic day E10.5, require the transcription factor Myb to colonize the fetal liver and later the bone marrow, and give rise to specialized hematopoietic cells, including myeloid cells in postnatal mice. The present disclosure is based on several key observations (summarized below) that demonstrate that tissue-resident macrophages originate in large part from EMPs, and provide novel experimental tools and strategies for the study of histiocytoses. First, in two independent models of transplantation without irradiation the majority of F4/80 bnght macrophages are self-maintained and do not renew from bone marrow HSCs in most adult murine tissues, with the notable exception of the gut. These findings have been replicated by a number of investigators. Second, development of these resident F4/80 bnght macrophages is unaltered in y£-deficient mice, lacking HSCs, thus providing the first genetic indication that resident macrophages develop from a progenitor distinct from HSCs. Third, F4/80 bnght macrophages in most adult mouse tissues were traced to progenitors that express Kit, CD45 low , AA4.1, and the Csfl -receptor (Csflr) at E8.5, two days before HSCs are identified in the embryo, and developmental distinct from HSCs using Cre-mediated hydroxy-tamoxifen (OH-TAM) induced pulse-labeling in C sflr enCreMer and Tie2 er, re er embryos. Altogether, it has been shown that EMPs colonize the fetal liver and give rise to pre-macrophages that colonize the embryo and differentiate into tissue macrophages. Thus, the majority of resident tissue macrophages that are present in mouse tissues originate from Kit + , CD45 low , AA4.1 + , Csflr + yolk sac early hematopoietic progenitors (defined as EMP) and can self-maintain in adults.

Resident macrophages are a diverse family of professional phagocytes and "accessory cells" involved in tissue remodeling.

[0073] Resident macrophages are sessile but continuously explore their immediate

environment/niche using motile filopodia, recognize, and scavenge pathogens as well as unfit cells and cell debris, glycoproteins and lipids, and produce a large range of bioactive molecules and growth factors. In mammals, macrophages are found all over the body within each specialized tissue and their serous membranes. Long elongated dendritic filopodial processes frequently extend from their cell bodies and build up 3D network-like structures that provide for extensive contacts with specialized cells such as brain, liver, kidney, or epidermal cells, thereby allowing constant surveillance or scavenging of their tissue. Macrophages are involved in the clearance of apoptotic and senescent cells during organogenesis, in the brain, limbs and lung, in branching morphogenesis, and regulate blood and lymphatic vessel morphogenesis and maturation during fetal and postnatal development. They pursue this task in adults, contribute to the pruning of neuronal synapses, and scavenge and digest nuclei released daily by billions (2xlO u in humans) of maturing erythroblasts. Altogether, a considerable literature indicates that macrophages play a role as regulators of morphogenesis, homeostasis and tissue remodeling, inflammatory processes and tumor growth.

[0074] The distinction between monocytes and macrophages is relevant to disease

pathogenesis and may be relevant to the pathophysiology of histiocytoses. Rossi and colleagues demonstrated that the two cell types have distinct expansion mechanisms and distinct functions in the brain during Experimental Auto-immune Encephalitis (EAE); infiltrating monocytes are recruited via extravasation from blood vessels and produce inflammatory mediators important for disease progression but do not persist after the resolution of inflammation, while in contrast activated resident microglia proliferate locally, persist, and return to quiescence following remission. In another elegant model, investigators showed that pleural resident macrophages adopt "anti-inflammatory" phenotypes in host carrying helminths while passenger monocytes exhibit pro-inflammatory responses. These studies underlie the notion that the two cell types, monocytes and macrophages, can exert distinct functions within the same environment and in response to the same challenge, suggesting that tissue cues or polarizing signals may not suffice to account for their functions. Of note, it might not be always accurate, however, to describe tissue macrophages as being anti-inflammatory as opposed to passenger monocytes being proinflammatory. For example, in a model of chronic neurodegeneration proliferating resident microglial cells were proposed to contribute to neuronal damage during the development of the disease.

Models to study the pathophysiology of histiocytoses in vivo.

[0075] The data discussed above suggested (1) that somatic BRAF V600E mutations in EMPs may be causative of histiocytoses, and (2) that BRAF V600E somatic mutations in HSCs and EMPs may have different consequences at the clinical, cellular, and molecular levels, and may thus contribute to the clinical heterogeneity of the disease and influence its prognosis and clinical course. As EMPs are not self-renewing and disappear in the late embryo the mutation would be detectable only in their progeny, the resident macrophages, and not in HSCs. Therefore, modeling BRAF V600E mutations in EMPs in mice is a strategy of choice to understanding its role in resident macrophage biology, should provide insight into the role of macrophages in promoting neurological dysfunction as well as lung and liver fibrosis, and may thus illuminate the pathophysiology of histiocytoses.

[0076] In one embodiment therefore, the technology of the present disclosure relates to a method for preventing or treating BRAF V600E -associated neurodegeneration by administering to a subject in whom BRAF V600E+ resident macrophages have been identified in the neuronal microenvironment of the subject a therapeutically effective amount of a BRAF, MEK, PI 3- kinase, Ras, and/or CSF-1R inhibitor. Suitable BRAF inhibitors include, but are not limited to, vemurafenib, dabrafenib, encorafenib, PLX7904, PLX8394, GDC-0879, LGX818, and PLX4720. Suitable MEK inhibitors include, but are not limited to, AZD8330, refametinib, E6201, MEK 162 (binimetinib), PD0325901, pimasertib, R04987655, selumetinib, TAK-733, GDC-0623, WX-544, cobimetinib, and trametinib. Suitable PI 3 -kinase inhibitors include, but are not limited to, idelalisib, BKM120, GDC-0980, PF-04691502, XL147, IPI-145, BYL719, SF1126, BAY80- 6946, GSK2126458, NVP-BEZ235, GDC-0941, PX-866, XL765, and ZSTK474. Suitable Ras inhibitors include, but are not limited to, salirasib and TLN-4601. Suitable CSF-1R inhibitors include, but are not limited to, GW2580, BLZ945, pexidartinib (PLX3397), ARRY-382,

PLX7486, and JNJ-40346527.

Therapeutic Methods

[0077] The following discussion is presented by way of example only, and is not intended to be limiting.

[0078] One aspect of the present technology includes methods of treating BRAF V600E - associated neurodegenerative disease in a subject diagnosed as having, suspected as having, or at risk of having one or more BRAF V600E mutations comprising the administration of a BRAF, MEK, PI 3-kinase, Ras, and/or CSF-1R inhibitor. In some embodiments, the present technology includes methods of treating neurodegenerative disease in a subject in need thereof, wherein the neurodegenerative disease is characterized by cells expressing one or more BRAF V600E mutations. In some embodiments, the cells are resident macrophages in the central nervous system, or microglia. In some embodiments, the neurodegenerative disease is Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis (ALS), or multiple sclerosis. In some embodiments, treatment of a BRAF V600E -associated

neurodegenerative disease comprises relieving one or more symptoms selected from the group consisting of: impaired cognitive functions and dementia, ataxia, dysarthria, decreased motor coordination and synchrony, paralysis, microglia accumulation, astrogliosis, microglia phagocytosis, demyelination, neuronal loss, synaptic loss, and amyloid precursor protein (APP) expression.

[0079] One aspect of the present technology includes methods of treating phosphoinositide 3- kinases (PI 3-kinase)-associated neurodegenerative disease in a subject diagnosed as having, suspected as having, or at risk of having one or more mutations in one or more PI 3-kinases comprising the administration of a BRAF, MEK, PI 3-kinase, Ras, and/or CSF-IR inhibitor. In some embodiments, the present technology includes methods of treating neurodegenerative disease in a subject in need thereof, wherein the neurodegenerative disease is characterized by cells expressing one or more mutations in one or more PI 3-kinases. In some embodiments, the cells are resident macrophages in the central nervous system, or microglia. In some

embodiments, the neurodegenerative disease is characterized by at least a portion of the resident macrophages in the central nervous system of the subject being PIK3CA H1047R+ . In some embodiments, the neurodegenerative disease is Alzheimer's disease, Parkinson's disease, Huntington's disease, or amyotrophic lateral sclerosis (ALS). In some embodiments, treatment of a PI 3-kinase-associated neurodegenerative disease comprises relieving one or more symptoms selected from the group consisting of: ataxia, decreased motor coordination and synchrony, paralysis, microglia accumulation, astrogliosis, microglia phagocytosis,

demyelination, neuronal loss, synaptic loss, and amyloid precursor protein (APP) expression in the brain.

[0080] In therapeutic applications, compositions or medicaments comprising a BRAF inhibitor,

MEK inhibitor, Ras inhibitor, PI 3-kinase inhibitor, CSF-IR inhibitor, or a combination thereof, as disclosed herein, are administered to a subject suspected of, or already suffering from a neurodegenerative disorder, in an amount sufficient to cure, or at least partially arrest, the symptoms of the disease, including its complications and intermediate pathological phenotypes in development of the disease. In some embodiments, the BRAF inhibitor is selected from the group consisting of vemurafenib, dabrafenib, encorafenib, PLX7904, PLX8394, GDC-0879,

LGX818, and PLX4720. In some embodiments, the BRAF inhibitor is PLX4720. In some embodiments, the MEK inhibitor is selected from the group consisting of AZD8330, refametinib,

E6201, MEK162 (binimetinib), PD0325901, pimasertib, R04987655, selumetinib, TAK-733,

GDC-0623, WX-544, cobimetinib, and trametinib. In some embodiments, the Ras inhibitor is selected from the group consisting of salirasib and TLN-4601. In some embodiments, the PI 3- kinase inhibitor is selected from the group consisting of idelalisib, BKM120, GDC-0980, PF- 04691502, XL147, IPI-145, BYL719, SF1126, BAY80-6946, GSK2126458, NVP-BEZ235, GDC-0941, PX-866, XL765, and ZSTK474. In some embodiments the CSF-1R inhibitor is selected from the group consisting of GW2580, BLZ945, pexidartinib (PLX3397), ARRY-382, PLX7486, and JNJ-40346527.

Modes of Administration and Effective Dosages

[0081] Any method known to those in the art for contacting a cell, organ, or tissue with a BRAF, MEK, PI 3-kinase, Ras, and/or CSF-1R inhibitor of the present technology, may be employed. In some embodiments, the BRAF inhibitor is selected from the group consisting of vemurafenib, dabrafenib, encorafenib, PLX7904, PLX8394, GDC-0879, LGX818, and

PLX4720. In some embodiments, the BRAF inhibitor is PLX4720. In some embodiments, the MEK inhibitor is selected from the group consisting of AZD8330, refametinib, E6201, MEK162 (binimetinib), PD0325901, pimasertib, R04987655, selumetinib, TAK-733, GDC-0623, WX- 544, cobimetinib, and trametinib. In some embodiments, the Ras inhibitor is selected from the group consisting of salirasib and TLN-4601. In some embodiments, the PI 3-kinase inhibitor is selected from the group consisting of idelalisib, BKM120, GDC-0980, PF-04691502, XL147, IPI-145, BYL719, SF1126, BAY80-6946, GSK2126458, NVP-BEZ235, GDC-0941, PX-866, XL765, and ZSTK474. In some embodiments the CSF-1R inhibitor is selected from the group consisting of GW2580, BLZ945, pexidartinib (PLX3397), ARRY-382, PLX7486, and JNJ- 40346527. Suitable methods include in vitro, ex vivo, or in vivo methods. In vivo methods typically include the administration of BRAF, MEK, PI 3-kinase, Ras, and/or CSF-1R inhibitor, such as those described above, to a mammal, suitably a human. When used in vivo for therapy, the BRAF, MEK, PI 3-kinase, Ras, and/or CSF-1R inhibitor inhibitors are administered to the subject in effective amounts {i.e., amounts that have desired therapeutic effect). The dose and dosage regimen will depend upon the degree of the neurodegenerative disease in the subject, the characteristics of the particular BRAF, MEK, PI 3-kinase, Ras, and/or CSF-1R inhibitor used, e.g., its therapeutic index, the subject, and the subject's history.

[0082] The effective amount may be determined during pre-clinical trials and clinical trials by methods familiar to physicians and clinicians. An effective amount of a particular BRAF, MEK, PI 3-kinase, Ras, and/or CSF-1R inhibitor useful in the methods of the present technology may be administered to a mammal in need thereof by any of a number of well-known methods for administering pharmaceutical compounds. The BRAF, MEK, PI 3-kinase, Ras, and/or CSF-1R inhibitor may be administered systemically or locally.

[0083] Pharmaceutical compositions are typically formulated to be compatible with its intended route of administration. Examples of routes of administration include, but are not limited to orally, intranasally, parenterally (e.g., intravenously, intramuscularly,

intraperitoneally, intradermally, or subcutaneously), systemically, transdermally,

iontophoretically, intradermal, intraocularly, or topically.

[0084] Dosage, toxicity and therapeutic efficacy of any therapeutic agent can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for

determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds that exhibit high therapeutic indices are advantageous. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.

[0085] The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds may be within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the methods, the therapeutically effective dose can be estimated initially from cell culture assays. A dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC 50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to determine useful doses in humans accurately. Levels in plasma may be measured, for example, by high performance liquid chromatography.

[0086] Typically, an effective amount of the BRAF, MEK, PI 3-kinase, Ras, and/or CSF-1R inhibitor, sufficient for achieving a therapeutic or prophylactic effect, ranges from about

0.000001 mg per kilogram body weight per day to about 10,000 mg per kilogram body weight per day. In some embodiments, the dosage ranges are from about 0.0001 mg per kilogram body weight per day to about 100 mg per kilogram body weight per day. In some embodiments, the dosage ranges are from about 0.0001 mg per kilogram body weight per day to about 50 mg per kilogram body weight per day. For example dosages can be 1 mg/kg body weight, 10 mg/kg body weight, or 50 mg/kg body weight every day, every two days or every three days or within the range of 1-50 mg/kg every week, every two weeks, or every three weeks. In one

embodiment, a single dosage of a BRAF, MEK, PI 3-kinase, Ras, and/or CSF-1R inhibitor ranges from 0.001-10,000 micrograms per kg body weight. In one embodiment, BRAF, MEK, PI 3-kinase, Ras, and/or CSF-1R inhibitor concentrations in a carrier range from 0.2 to 2000 micrograms per delivered milliliter. An exemplary treatment regime entails administration once per day or once a week. In therapeutic applications, a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, or until the subject shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regimen.

[0087] The skilled artisan will appreciate that certain factors may influence the dosage and timing required to effectively treat a subject, including but not limited to, the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of the therapeutic compositions described herein can include a single treatment or a series of treatments.

[0088] The mammal treated in accordance with the present methods can be any mammal, including, for example, farm animals, such as sheep, pigs, cows, and horses; pet animals, such as dogs and cats; laboratory animals, such as rats, mice and rabbits. In some embodiments, the mammal is a human.

EXAMPLES

[0089] The following examples are provided to further illustrate the methods of the present disclosure. These examples are illustrative only and are not intended to limit the scope of the disclosure in any way.

Materials and Methods

[0090] Targeting of BRAF V600E mutation to EMPs and EMP-derived cells. EMPs appear in the yolk sac at embryonic day (E) 8.5 and express the Csfl receptor (Csflr). They colonize the fetal liver from E9.5 and give rise to macrophage precursors (pMacs) that distribute in embryonic tissues and differentiate into tissue-specific macrophage subsets such as microglia in the central nervous system. Therefore, to investigate the consequences of BRAF V600E expression in the EMP lineage, EMPs were genetically targeted by pulse labeling

CsfJt MenCreMer ;BRAF LSL-V600E . ^SL-YFP ^ embryos ^ 4_hydr OX ytam O xifen (4-OHT, Sigma). Embryonic development was estimated considering the day of vaginal plug formation as 0.5 days post-coitum (dpc). Cre recombination in C sflr Mer,CreMer ; Rosa26 LSL - YFP ; BRAF LSL~ V600E embryos was induced by single injection of 37.5 mg per kg (body weight) of OH- TAM into pregnant females. Short-lived 4-OHT leads to transient nuclear translocation of the estrogen receptor-Cre recombinase fusion protein (MeriCreMer) in cells expressing the Csfrl Mer,CreMer transgene and deletion of a floxed stop cassette (LSL) in the BRAF LSL - V600E and Rosa26L SL~YFP alleles. OH- TAM was supplemented with 18.75 mg per kg (body weight) progesterone (Sigma) to counteract the mixed oestrogen agonist effects of tamoxifen, which can result in fetal abortions.

[0091] Mice. Csfl^^ CsfJr^, CDll Cr \ Rosa26 YFp - LSL mice and BRAF^ L - V600E mice kindly provided by C. Pritchard (Leicester, UK) were maintained under SPF conditions. Animal procedures were performed in adherence to our project licenses issued by the Institutional Review Board (IACUC 15-04-006 and 13-04-003) from MSKCC. Genotyping was performed according to protocols described previously for Csflr ,Cre Csflr Mer,CreMer , CDll Cre and BRAF^™ r ce. Cre recombination in C sfl^ mCreMer - Rosa26 L ^ wp - BRAF^^ 60 ^ embryos was induced by single injection at E8.5 of 37.5 mg per kg (body weight) of 4-hydroxytamoxifen (OH- TAM, Sigma) into pregnant females. OH-TAM was supplemented with 18.75 mg per kg (body weight) progesterone (Sigma) to counteract the mixed oestrogen agonist effects of tamoxifen, which can result in fetal abortions. Embryonic development was estimated considering the day of vaginal plug formation as 0.5 dpc. For BRAF inhibition, animals were placed on ad libitum PLX4720 diet at 1 or 3 months of age (PLX4720-containing chow 417ppm, provided by Plexxikon Inc.). BRAF^ and BRAF 1 ^ male and female littermates were assigned randomly into the control or treated group. Scoring of mice was performed blinded and at least weekly by assessing hindlimb reflexes and other behavioral phenotypes such as axial rolling. The investigators were not blinded to allocation during experiments and outcome assessment.

[0092] Footprint analysis. Mice were given two trials to run down a runway before the experiment. Mouse forepaws and hindpaws were painted with red and blue ink, respectively. Mice were then allowed to run down a runway lined with white paper. At least four steps from the middle portion of each run were measured for (1) overlap between forepaw and hindpaw placement, (2) stride length, (3) front-base width (the distance between the right and left forelimb strides), and (4) hind-base width (the distance between the right and left hindlimb strides). Mean values were used for graphs and statistical analyses. [0093] Rotarod assay. The assay was conducted using a ROTO-ROD series 8 (IITC Life Sciences) with accelerating speed (accelerated from 4 to 40 rpm over 120 s). The mice were trained on the accelerating rotarod with 3 training session per day for 3 days. On the day of the experiment, the mean latency to fall off the rotarod recorded in the 3 trials was used in the analysis.

[0094] Preparation of cell suspensions, flow cytometry and cell sorting. Pregnant females were killed by exposure to C02. Embryos were removed from the uterus, washed in 4°C phosphate- buffered saline (PBS, Invitrogen) and dissected under a Leica M80 microscope. For blood phenotyping of adult mice, mice were anaesthetized and blood was collected by cardiac puncture. Bone marrow was collected by flushing one leg with 5 ml RPMI (Invitrogen). For flow cytometry experiments, organs were incubated in PBS containing lmg/ml collagenase D (Roche), lOOU/ml DNase I (Sigma), 2.4mg/ml of dispase (Invitrogen) and 3% FCS (Invitrogen) at 37°C for 30 min prior to mechanical disruption. For embryonic tissue incubation time at 37°C was reduced to 20 min. For cell sorting, tissues were digested for 30min at RT in PBS containing 2 mg/ml of collagenase D (Roche), 200U/ml DNase I (Sigma), 4.8 mg/ml of dispase (Invitrogen), 3% FCS (Invitrogen) and luM of flavopiridol (Sigma) followed by mechanical disruption under a l OOum filter. Cell suspensions were centrifuged at 320g for 7 min, resuspended in FACS buffer (PB S, 0.5% BSA and 2 mm EDTA) containing purified anti-CD16/32 (1 : 100 dilution) and 5 % normal mouse, 5 % normal rat and 5 % normal rabbit serum and incubated for 15min at 4°C. Samples were immunostained with antibodies mixes for 30 min at 4°C. For FMO (fluorescence minus one), brain cell suspensions from BRAF WT and BRAF VE mice were mixed prior to staining. The full list of antibodies used can be found in Supplementary Table 5. Cell sorting was performed using an Aria II BD cell sorter. Single live cells were gated on the basis of dead cell exclusion (DAPI), side (SSC-A) and forward scatter (FSC-A) gating, and doublet exclusion using forward scatter width (FSC-W) against FSC-A.

Macrophage populations were identified after gating on CD45 based on expression of F4/80, CD1 lb and YFP. 200 cells for each sample were directly sorted into a 96 well plate (Biorad) in 4ul of H20 containing 0.2% of triton-X (Sigma) and 0.8U/ul of RNAse inhibitor (Clontech), and processed as indicated below.

[0095] For intracellular stainings, cell suspensions were purified by a Percoll (Sigma) gradient (70/37/30%)), and cells were collected from the 70/37 interface. After washing twice with PBS, cells were stained with a viability dye (Ghost Dye Red 780, Tonbo Biosciences) according to the manufacturer's protocol. Cells were first stained with antibodies for surface markers {see Suppl ementary T abl e 5 ) , then proce s sed for intracel lul ar stai ni ng s u sing the Foxp3/Transcription Factor Staining Buffer Set (Affymetrix eBioscience) according to the manufacturer' s protocol. Cell numbers per organ or per gram of tissue were calculated as follows. For embryonic tissues, cell suspensions were prepared, stained, and acquired from whole organs, and the number of live cells per tissue was directly obtained from FCS files. In adult mice, organs were weighted, cell suspensions were prepared from 20 to 500 mg of tissue, and the number of cells per gram of tissue was determined using a cell counter (GUAVA easyCyte HT). For gating strategies used for different tissues, see Supplementary FIG. 2. Proportion of YFP + F4/80 + cells in tissues from BRAF VE and BRAF m littermates were analyzed by calculating the proportion of YFP among F4/80 + cells. Values from BRAF m littermates were then normalized and set to 1.

[0096] Generation and analysis of Kupffer cells and microglia RNA-seq in BRAF VE and

BRAF m littermate controls. Sorted cells underwent amplification (14 cycles) using the SMART - seq V4 (Clonetech) ultra low input RNA kit for sequencing. 10 ng of amplified cDNA was used to prepare Illumina hiseq libraries with the Kapa DNA library preparation chemistry (Kapa Biosystems) using 8 cycles of PCR. Samples were barcoded and run on a Hiseq 2500 IT in a 50bp/50bp Paired end run, using the TruSeq SBS Kit v3 (Illumina). An average of 54 million paired reads were generated per sample and the percent of mRNA bases was closed to 77% on average. FASTQ files were mapped to the Mouse genome mm 10 using the Star aligner that maps reads gnomically and resolves reads across splice junctions. Several QC metrics were used for the RNA-seq library, including intron-exon ratio, intragenic reads fraction, and GC bias. Exon and gene expression were quantified using Sailfish against the Mus musculus

transcriptome GRCh38. After merging technical replicates, differential expression tests were performed using the DESEQ2 algorithm. Genes that had a FDR<0.01 were considered to be significantly different between genotypes and ranked significant genes by fold-change with a cutoff of 1. Gene set enrichment analysis on KEGG pathway, GO term, Reactome, and

MSigDB gene set collection was done using the over-representation test with hypergeometric model to assess whether the number of selected genes associated with disease is larger than expected.

[0097] Generation and analysis of human brain RNA-seq. Snap frozen normal brain tissues were obtained from the MSKCC Medical Donation Program. RNA was extracted using the

Qiagen all prep DNA/RNA mini kit (Cat#80204) according to the manufacturer's instructions.

RNAs were submitted to ribogreen quantification and quality control on Agilent

BioAnalyzer. Average amount was 5.3ug, Average RIN was 8.9. 500ng of total RNA underwent polyA selection and Truseq library preparation according to instruction provided by Illumina (TruSeqTM RNA Sample Prep Kit v2), with 8 cycles of PCR. Samples were barcoded and run on a Hiseq 4000 in a 125bp/125bp Paired end run, using the TruSeq SBS Kit v3 (Illumina). An average of 75 million paired reads was generated per sample. At the most the ribosomal reads represented 7% and the percent of mRNA bases was closed to 45% on average. FASTQ files from control brains and previously sequenced Juvenile Xanthogranuloma (JXG) and Langerhans Cell Histiocytosis (LCH) brain samples (SJLCH13 and SJLCH14) were mapped to Human genome GRCh37 using Star aligner. Several QC metrics were used for the RNA-seq library, including intron-exon ratio, intragenic reads fraction, and GC bias. Exon and gene expression were quantified using Salmon against Homo sapiens transcriptome GRCh37. Differential expression tests, statistical tests and pathway analysis was performed as described above for murine RNA-seq.

[0098] Immunofluorescence, imaging and analysis (mouse). Embryos were imaged using a Leica M80 or Zeiss Axio Zoom. VI 6. Tissues were fixed for 1-3 days in 4% formaldehyde (Sigma). After fixation, tissues for cryosections were incubated overnight in 30% sucrose and embedded in OCT compound (Sakura Finetek). Cryoblocks were cut at a thickness of 16 μπι for liver and 50 μπι for spinal cord and brain and then blocked with PBS containing 10% normal goat serum (Invitrogen); 1% BSA (w/v); 0.3% Triton X-100 for 1 hour at room temperature. Livers were incubated overnight, brains and spinal cord for 48 hours at 4°C with rat anti-mouse F4/80 (1 :300, cat no: MCA497GA, Biorad), rabbit anti-mouse Ibal (1 :300; cat no: 019-19741, Wako), chicken anti-GFP for YFP detection (1 :500, cat no: A10262, Invitrogen), rabbit anti- pHis3 (1 : 100, cat no: sc-8656-R, Santa Cruz), goat anti-IL-lb (1 :40, cat no: AF-401-SP, R&D), rabbit anti-pERKl/2 (1 : 100, cat no: #4370, Cell signaling), rabbit anti-Collagen IV (1 : 100, cat no: 2150- 1470, Biorad), rabbit anti-Collagen VI (1 :200, cat no: ab6588, Abeam), rabbit anti- cleaved Caspase3 (1 :600, cat no: #9661, Cell Signaling), rat anti-Ki-67 (1 :200, cat no: 14-5698- 80, eBioscience). Secondary antibodies used were anti-rabbit Cy3 (1 :500, Invitrogen), anti- chicken Alexa Fluor 488 (1 :500, Invitrogen), anti-rat Alexa Fluor 647 (1 :500, Invitrogen), anti- goat Alexa Fluor 568. Samples were then mounted with Fluoromount mounting medium with DAPI (eBiosciences) and visualized using a LSM880 Zeiss microscope with 20x/0.5 (dry) and 40x/1.4 (oil) performing a tile scan and Z-stack on whole tissue. Image analysis and cell quantification was performed using Imaris (Bitplane) software. For paraffin sections, 3 μπι sections were prepared before staining with H&E, Trichrome, and luxol fast blue (LFB-PAS). Primary antibodies were rabbit anti-CD68 (5 μg/ml, Boster Biological Technology, cat. no: PA1518), rabbit anti-pERKl/2 (1 μg/ml, cat no: #4370, Cell signaling), chicken anti-GFP for YFP detection (1 :5000, cat no: AB 13970, Abeam), mouse anti-APP (1 :3000, cat no: MAB348, Millipore), rabbit anti-Iba-1 (1 :500, cat no: 019-19741, Wako), rabbit anti-GFAP (1 :200, cat. no. Z0334, Dako), mouse anti-NeuN (1 :200, cat. no. MAB377, Merck Millipore), rat anti-LAMP-2 (1 :250, abl3524, Abeam, Cambridge, UK), mouse anti-pERKl/2 (1 :200, sc-136521, Santa Cruz), rabbit anti-homerl (1 :200, cat. no. 160003, Synaptic Systems), mouse anti-synaptophysin (1 :500, cat. no.101 011, Synaptic Systems). Primary antibodies were incubated overnight at 4°C. Secondary Alexa- or HRP-conjugated antibodies were added at 1 :200 in antibody buffer for 2 hr at room temperature. For immunof uorescent stainings nuclei were counterstained with 4,6- diamidino-2-phenylindole (DAPI, cat. no. 236276, Boehringer). All slides were mounted in Vectashield (Vector Laboratories). Images were taken using a Zeiss Lab.Al or BZ-9000 Biorevo microscope (Keyence) and analyzed using the BZ-II Analyzer (Keyence). For quantification of cell numbers, at least three parasagittal sections per mouse were analyzed. In cases, where the cell density was too high and single cells could not be quantified, the immunolabeled area size per brain section was calculated. To determine synapse density, imaging was performed using an Olympus Fluoview 1000 confocal laser scanning microscope using a 20 x 0.95 NA objective. Z- stacks with 0.05-μιη steps in the z direction, 1,024 1,024 pixel resolution, were recorded and analyzed using Imaris software (Bitplane). Colocalization of the presynaptic marker synaptophysin with the postsynaptic marker Homerl was quantified.

[0099] Immunofluorescence, imaging and analysis (human). Written informed consent was obtained from histiocytosis patients for DNA/RNA sequencing and immunohistochemical studies according to Helsinki convention, and this study received approval from the Institutional Review Board at MSKCC (IRB Protocol 06-107). For all ECD patients, ECD was diagnosed in light of published guidelines for the diagnosis and clinical management of ECD42. Biopsy material was retrieved from the pathology archives at Memorial Sloan Kettering Cancer Center. Immunohistochemistry of human ECD and control tissue {see Supplementary Table 6) was carried out on 3-4 μπι thick paraffin sections, fixed with PFA. Immunohistochemical analysis was performed on paraffin sections with rabbit anti-Iba-1 (1 :500, cat no: 019-19741, Wako) and mouse anti-pERKl/2 (1 :200, sc-136521, Santa Cruz), or mouse anti-CD163 (0.06ug/mL, cat#760-4437, Cell Marque), rabbit anti-pERKl/2 (^g/ml, cat no: #4370, Cell Signaling) and mouse anti-BRAF V600E (VE1) (1 :800, cat no: #E19294, clone VE1, Spring). Secondary Alexa-or HRP-conjugated antibodies (Invitrogen) were added at 1 :200. Images were taken with using a Zeiss Lab.Al, Bondlll (Leica-Microsystems, Buffalo Groove, IL), BZ-9000 BIOREVO microscope (Keyence) and analyzed using the BZ-II Analyzer (Keyence), or with a LSM880 Zeiss microscope with 40x/1.4 (oil) performing a tile scan and Z-stack on whole tissue and manually analyzed using Imaris (Bitplane) software. [0100] Western blot. Tissues were homogenized using a cell lysis kit (Biorad) and a Precellys homogenizer. Primary antibodies were added overnight at a dilution 1 : 1000 for rabbit anti- pERKl/2 (cat no: #4370, Cell Signaling) and rabbit anti-ERKl/2 (cat no: #9102, Cell Signaling), and 1 :5000 mouse anti-beta actin (cat no: #ab6276, abeam). Secondary antibody anti-rabbit HRP (cat no: #7074, Cell Signaling) or anti-mouse AP (cat no: #AP-2000, Vector laboratories) was added for 1 hour at a dilution of 1 :2000. Detection of HRP was performed using Pierce ECL Western Blotting substrate (cat no: #32106). Detection of AP vas performed using BCIP/NBT substratekit (cat no: #SK-5400, Vector laboratories). For quantification, a ratio was made of the total integrated optical density (IOD) of pERK bands to the IOD of bands of the corresponding total ERK protein using ImageJ.

[0101] Cytokine analysis in spinal cords. Proteins were extracted from 25 mg of tissue using Bio-Plex cell lysis kit. Cytokine concentrations were measured using Bio-Plex Pro Mouse Cytokine 23-plex Assay according to manufacturer's protocol.

[0102] Serum analysis. 70-75uL of serum were analyzed for liver enzymes using Beckman Coulter AU680 Chemistry analyzer.

[0103] PLX4720 concentration measurement. Analysis of PLX4720 concentrations in brain, liver and serum was performed in collaboration with Plexxikon Inc. 25 μΐ serum and 20 mg of homogenized tissue were measured using a standard curve that was generated by adding known amounts of PLX4720 to an untreated serum or homogenized sample.

[0104] Statistical analysis and reproducibility . Data are shown as mean with individual values per mouse being represented as circles, unless stated otherwise. Statistical significance was analyzed with Graph Pad Prism by using Mann-Whitney tests, unpaired two-tailed t-tests, 1-way and 2-way ANOVA and Log-rank (Mantel-Cox) test as indicated in the figure legends.

[0105] The n value represents biological replicates. For RNA-seq statistical analysis R software was used {see Generation and analysis of Kupffer cells and microglia of RNA-seq in BRAFVE 548 and BRAFWT littermate controls and Generation and analysis of human brain RNA-seq). Significance was considered at p < 0.05. Animals that were labeled moribund by veterinarian services had to be euthanized and were therefore excluded from further longitudinal analyses as indicated in the figure legends. Kaplan-Meier survival analysis was used to estimate overall survival and cumulative incidence rate. Experiments were repeated to ensure

reproducibility of the observations. Equal variance was assumed for cell counting experiments. No statistical methods were used to predetermine sample size. Example 1: Constitutive expression of BRAF V600E in Csflr-expressing cells results in a leukemic phenotype.

[0106] BRAF V600E expression in hematopoietic progenitors in Vav Cre ; BRAF V600E mice results in a lethal and transplantable hematopoietic disorder characterized by splenomegaly, anemia, and thrombocytopenia. In an attempt to limit expression of the BRAF V600E mutation to myeloid cells and their progenitors (EMP and HSCs), a triple transgenic mouse model: Csflr ,Cre ; BRAF V600E ; Rosa26 LSL - YFP was established to target expression of BRAF V600E and a fluorescent YFP reporter to cells expressing Csflr (FIG. 3). The Rosa26 LSL~YFP transgene allows for facile identification of targeted cells in vivo and ex vivo. The resulting embryos develop normally until El 1.5, but show increased fetal liver size, weight, and accumulation of YFP + lineage-negative cells in the fetal liver and haemorrhagic foci from E12.5 onwards (FIG. 3C, and not shown) and all

BRAF V600E fetuses die between E13.5 - E14.5 with a phenotype reminiscent of the Vav Cre ;

BRAF V600E mice (30) (FIGs. 3B-3D).

[0107] In addition, expression of BRAF V600E in a large number of early hematopoietic precursors leads to a leukemic-like phenotype and an increase of early macrophages (not shown). This did not phenocopy histiocytoses, where a leukemic phenotype is rare. A limitation of this and previous murine models is that constitutive expression of Cre results in a very high frequency of cells expressing BRAF V600E within a target population, and this may not accurately model the behaviour of a limited number of BRAF V600E clonogenic progenitors. Therefore, an aim of the present technology is to target the BRAF V600E mutation to only a fraction of myeloid progenitors - either EMPs or HSCs -to investigate the contribution of the affected cell lineage to disease pathophysiology. To test this hypothesis, mouse models are utilized that will allow targeting of a small proportion of EMPs or HSCs, thereby obtaining mosaicism and more closely modelling a somatic mutation.

Example 2: Tamoxifen-inducible models to target resident macrophages or HSCs.

[0108] To target BRAF V600E expression specifically to a small number of EMPs or HSCs, BRAF V600E ;Rosa26 LSL~YFP and Rosa26 LSL~YFP mice were crossed either to a tamoxifen-inducible Csflj er,CreMer model, thereby allowing selective targeting of EMPs when pulsed with OH-TAM at E8.5, at time when EMP start to express Csflr, or to a Cxcr4 CreERT2 model allowing selective targeting of HSCs when pulsed with OH-TAM at E9.5 (FIG. 4). Pregnant females receive low doses of OH-TAM via i.p. injection, so that only a small proportion of EMPs express BRAF V600E and YFP. Analysis of 4-week-old Csfli er,CreMer ; Rosa26 LSL - YFP ; BRAF V600E Cre + mice indicates that YFP expression is not detected in bone marrow HSC (Lineage negative, Scal + Kit + or LSK) as expected (FIG. 4), while -10% of LSK cells were labelled in the bone marrow of Cxcr4

CreERT2 ; R D osa 2 ~.6 A LSL-YFP mi ce ffi (F T IG. 4 A ).

Example 3: The tamoxifen-inducible Csflr MeriCreMer ; BRAF V600E ; Rosa26 LSL YFP mouse model targets resident macrophages.

[0109] Littermates born from BRAF V600E ;Rosa26 LSL - YFP crossed to Csflr er,CreMer pulsed with OH-TAM at E8.5 are viable, and born at almost perfect Mendelian ratios (FIGs. 5A and 5B). At a young age (4 weeks old), Cre + BRAF V600E animals did not display any gross phenotypic abnormality. YFP expression in Csflr MenCreMer ; Rosa26 LSL - YFP ; BRAF V600E mice was restricted to small populations of F4/80 + tissue macrophages (FIG. 5C) that form clones within the liver tissue (FIG. 5D) as well as the nervous system, mostly brain stem, cerebellum and spinal cord (FIG. 5E). YFP expression was not observed in hepatocytes or neurons.

Example 4: Csflr MeriCreMer ; Rosa26 LSL YFP ; BRAF V600E mice develop clinical features of neurodegenerative disease.

[0110] In a cohort of 5-9 month-old Cre + BRAF V600E (n=20) mice and littermates (FIG. 6), Cre BRAF V0UUC mice began to develop impaired limb-clasping reflexes and ataxia, which was not detected in control littermates (FIGs. 6A and 6C). The cumulative incidence of these phenotypes reached -95% by 7 months of age, while BRAF WT littermates remained normal (FIG. 6B). Motor coordination and synchrony was assessed by footprint assay, measuring stride length, base width, and overlap between fore and hind paws (FIG. 6C). During the early stages of ataxia, the overlap of front and hind paws of Cre + BRAF V600E animals was significantly increased, while the other coordination measurements remained similar to Cre + BRAF WT littermates (FIG. 6D). Later, paralysis of the hind legs became apparent in the footprint assay, showing full loss of coordination (FIG. 6C). This observation is important, as the development of ataxia is a component of the clinical phenotype of histiocytosis patients that develop neurodegenerative disease. In patients with LCH, symptoms of neurodegenerative disease ( D- LCH) include cerebellar ataxia with dysarthria, dysdiadochokinesis, and associated with concentration deficits, psychomotor retardation, severe headaches, and psychosis, developing 3 to 15 years (median 6 years) after initial LCH diagnosis.

[0111] The pathophysiology of these neurological signs and symptoms are unknown, however, as biopsies are rarely performed in patients and when performed, pathological histiocytes are rarely identified. A preliminary histological analysis and flow cytometric analysis of the brain and spinal cord from Csfl ienCreMer ;BRAF V600E ;Rosa26 LSL - YFP mice presenting with ataxia and littermates, indicated the presence of foci of increased numbers of YFP + Ibal + microglia in the cerebellum and spinal cord in BRAF V600E+ littermates (FIG. 7), suggesting active involvement of pathological BRAF V600E histiocytes in the process of neurodegeneration. At the time of analysis, mice with a neurological phenotype already presented with extended histological lesions, such as a massive demyelination in the white matter of the spinal cord (FIG. 8A) associated with an inflammatory infiltrate rich in CD3 + T lymphocytes (FIG. 8B). These data are also reminiscent of MRI studies in patients, which show signal changes in cerebellar white matter. However, it is likely that the histological lesions started to develop long before clinical symptoms appeared. These preliminary results suggest that targeting BRAF V600E expression to a small number of EMPs gives rise to BRAF V600E tissue resident macrophages in adult mice that may be responsible for the most troubling clinical sequelae of LCH and ECD. This model may faithfully

recapitulate the disease in a manner, which has not been accomplished by BRAF V600E expression in other hematopoietic cellular compartments. These results warrant a detailed analysis of the neurological disease that develops in C sflr MenCreMer ; BRAF V600E ; Rosa26 LSL - YFP mice to elucidate the cellular and molecular mechanisms involved, justify investigations into other pathological consequences of BRAF V600E expression in macrophages in the liver, lung, and bones and determine whether these phenotypes can also result from mutated HSCs. These results also offer a unique opportunity to study the effects of therapeutic interventions and identify novel therapeutic targets.

Example 5: Determining the molecular and cellular mechanisms that underlie the development of brain neurodegenerative disease.

V600E .

[0112] At the onset of clinical symptoms, the nervous system of Csfli enCreMe ' " ; BRAF

Rosa26 LSL - YFP mice displays extensive damage. As outlined above, it is likely that the histological lesions in the nervous system started to develop long before clinical symptoms appeared. When limb-clasping reflexes and ataxia are detected by gross examination in 6 month old mice, histological evaluation of the spinal cord indicated extensive demyelination (FIG. 8A) and flow cytometry analysis indicated infiltration by CD3 + cells (FIG. 8B). Immunostaining for phosphorylated ERK (pERK), a downstream effector of the Ras/Raf pathway showed extensive areas of strong pERK staining in Cre + BRAF V600E microglia in comparison with littermates controls (FIG. 9). Clustering of YFP + CD68 + microglia in the cerebellum and spinal cord white matter was most prominent in the posterior columns and spinocerebellar tracts (FIG. 9, and data not shown), which are important for proprioception and fine touch of the hind limbs and lower trunk. Anatomical lesions involving these neurological tracts could explain the lack of coordination of the hind limbs with the forelimbs and may contribute to paralysis. However, the mice are paralyzed at this stage and the cellular damage is extensive, making it difficult to properly assess the course of the disease.

Example 6: Determining the onset of neurological disease in the Csflr MenCreMer ;

BRAF V600E ; Rosa26 LSL YFP mice.

[0113] As stated above, the onset of neurological defects that would impact locomotion, muscular strength or memory at early stages of the disease remain unknown. To identify the onset of the disease the occurrence of diabetes insipidus (DI), which corresponds to pituitary involvement and is associated with neurodegeneration in patients such that ~ 95% of clinical neurodegenerative syndrome cases have DI is investigated. Serum and urine sodium osmolarity are tested for a cohort of mice (n=10 per genotype) between 4 and 8 weeks, and if DI is suspected (high plasma/low urine sodium concentration), mice are tested in metabolic cages. A set of fine motor tests that will provide a good read-out of neurological function are also performed on a cohort of age matched Csfli er,CreMer ; BRAF V600E ; Rosa26 LSL - YFP mice and BRAF WT littermate controls (n=10 for each genotype) starting at the age of 4 weeks, and every two weeks initially until tests are positive. The rotarod assay is a sensitive assay that can detect subtle impairments in motor activity, and can be repeated over time in a cohort of animals, to monitor the onset and progression of neurological impairment or a motor coordination phenotype. The rotarod is a horizontally oriented cylinder that can rotate at a fixed or an accelerating speed. Normally, animals try to stay on the rotarod, and avoid falling to the ground. During the training period, each mouse is placed on the rotarod at a constant speed (24 rpm) for 60 seconds.

CsfJr MenCreMer . BRA pV600E . Rosa26 ^-ΥΡΡ ^ ^ BRA pWT Mermate contro l s at the age of 4 weeks receive four trials per day for three consecutive days, by which time they reach a steady performance baseline level. After the training trials, mice receive two trials for 60 seconds at eight increasing speed levels: 4, 8, 15, 20, 24, 31, 33 and 44 rpm. The mean latency to fall off the rotarod is used in subsequent analysis. If a mouse stays on the rod until the end of the trial, a time of 60 sec is recorded. The same mice are analyzed over a time period of 6 months (or until paralysis) every 2 weeks. The cohort of mice tested with rotarod above is also longitudinally studied with the footprint assay. The footprint assay measures motor coordination and

synchrony, and has been used to assess microglial inflammation in murine models, and it was found that 6-9 months old Csflr MenCreMer ; BRAF V600E ; Rosa26 LSL - YFP mice presented with ataxia in this assay {see FIG. 6). To obtain footprints, the front and hind paws of the mice are coated with red and blue ink, respectively. The animals are then allowed to walk along a 50-cm-long,

20-cm-wide runway (with 15-cm-high walls). All mice will have 2-3 training runs before ink is applied. A fresh sheet of white paper is placed on the floor of the runway before each run. The footprint patterns are analyzed for four parameters (see also FIG. 6): (1) Stride length, (2) hind- base width, and (3) front-base width are measured as the average distance between left and right hind footprints and left and right front footprints, respectively. These values are determined by measuring the perpendicular distance of a given step to a line connecting its opposite preceding and proceeding steps. (4) Distance from left or right front footprint/hind footprint overlap is used to measure uniformity of step alternation. If the center of the front footprint falls on the center of the preceding hind footprint, a value is recorded as 0 cm. When the footprints do not overlap, the distance between the centers of the footprints is measured. For each step parameter, at least three values are measured and averaged, excluding footprints made at the beginning of the run where the animal was initiating movement. Locomotion and exploratory behavior are also assessed using an open field analysis in a new environment (clear plexiglas 40 χ 40 χ 30 cm open-field arena). This test can only be performed once in a cohort to avoid the confounding effects of habituation to the open field, and is initially performed on the cohort at the time of the first symptoms in the rotarod or footprint assays. Activity in the open-field is quantified by a computer-operated Photobeam activity system (AccuScan Instruments, Columbus, OH). Mice are recorded for the total distance moved (cm), number of vertical episodes (rearing), and distance moved in the center of the arena (cm). The distance moved in the center (cm) is divided by the total distance moved (cm) to obtain center/total distance ratio values. Data is collected at 5-10 min intervals over 20-60 min test sessions. All results are analyzed comparing Cre + BRAF V600E and RAF m mice, for statistical analyses Student's t-test is performed and p-values of <0.05 are considered significant.

[0114] Results from this longitudinal approach characterize the onset of the motor coordination phenotype observed in the Csflr MenCreMer ; BRAF V600E ; Rosa26 LSL - YFP mice. These results guide the animal imaging histological and molecular analysis described below. To assure an unbiased analysis of behavioral assays the researcher is blind to the genotype of the mice during testing and in case of the open field study, also later during analysis of the tapes.

Example 7: Analyzing brain histology and neural cell populations in Csflr MenCreMer ;

BRAF V600E ; Rosa26 LSL"YFP mice during development and through progress of

neurodegeneration.

[0115] Microglia play important roles in the homeostasis of the brain and spinal cord, during development and in adults. They are involved in pruning synapses, modulating over-active neurons, promoting neuronal survival, organizing axonal projections and, in the adult, supporting synapse formation during learning. To uncover the consequences expression in microglia differentiation, proliferation, and activation during development and in post-natal animals, a detailed analysis of microglia proliferation and activation and its consequences on other neural cells populations is performed at different time points during development of C 7r er, re er BRAF V600E ; Rosa26 LSL - YFP mice and BRAF m littermate controls. Experiments are performed at E12.5, E14.5, E18.5, and after birth at postnatal week 1, week 2, at week 4 (before the onset of clinical symptoms), at the onset of symptoms, and in 5-6 month old mice. Embryos and brains are harvested for flow cytometry experiments or immediately fixed in a solution of 4% formaldehyde before processing for cryosections (for immunofluorescence) or paraffin sections (immunohistochemistry, IHC). Proliferation rate of BRAF V600E and BRAF WT microglia (F4/80 + CDl lb + CD45 l0 ) is quantitated in situ and by flow cytometry using phosphorylated histone H3 (pHis3) antibody. To analyze EMP-derived BRAF V600E and BRAF^ microglia and their impact on neurons, 30 μπι thick cryosections will be co-stained for YFP, Ibal, and pERK and the neuronal antigen NeuN. Automatic quantification of cell morphometry on three- dimensional reconstruction with Imaris will assess dendrite length, number of segments, branch points and terminal points as well as volume. To study how BRAF V600E expressing microglia may affect neurogenesis/neuronal loss, staining for Fluoro-Jade, a fluorochrome commonly used to label degenerating neurons, will be performed. The number of astrocytes is typically elevated when destruction of neurons occurs, and this is assessed by IHC of GFAP, a classic marker for reactive gliosis. Expression of maturation and activation markers Csflr, CD31, CD44, CD62L and MHC class II by microglia (F4/80 + CDl lb + CD45 l0 ) , and the presence and composition of an inflammatory infiltrate are determined by flow cytometry using a LSR Fortessa flow cytometer to identify T cells (CD3, CD4, CD8), B cells (CD19), eosinophils, monocytes and neutrophils (Ly6C, Ly6G, Siglec-F, CDl lb). In all experiments, immunostainings and flow cytometry analyses are performed on at least 3 embryos/brains per genotype and 3 imaged sections per embryo/brain, p-values of <0.05 obtained using student's t-test are considered significant.

[0116] Results from this longitudinal approach, which follows BRAF V600E expressing microglia throughout development to adulthood characterizes the disease onset and provides a better molecular understanding for the pathophysiology of central nervous system LCH and neurological manifestations in ECD. Example 8: Characterize the role of BRAF V600E expression on the transcriptional profile of microglia.

[0117] Raf-Mek-Erk signaling is involved in differentiation and cell identity, cell proliferation and activation via the control of gene transcription and chromatin accessibility. The

transcriptional consequences of BRAF V600E expression in macrophages have not been investigated. The transcriptional profiles of EMP-derived WT macrophages were identified in murine tissues, during development and in adults, using RNAseq analysis (FIG. 10). A transcriptional analysis of YFP + and YFP " macrophages obtained from the brain (microglia) is performed and for comparison from the liver (Kupffer cells) of Csflr MenCreMer ; BRAF V600E ;

Rosa26 LSL~YFP and BRAF m littermates, at 4 weeks, before the onset of disease, at the onset of disease and at 5 months, when the mice are sick. Comparison of YFP + cells from BRAF V600E mice and BRAF WT littermates will provide a first set of data, which will identify pathways and genes that characterize the presence of a BRAF V600E allele. Comparison between brain and liver macrophages, will inform the presence of tissue-specific BRAF signatures. Such tissue-specific BRAF-mutant macrophage signatures are likely to exist as the existence of tissue-specific transcriptional profiles of normal macrophages have been shown. Finally, comparison between the 3 time points should identify genes and pathways involved in macrophages as the disease progresses. For each time point, macrophages are FACS-sorted using an Aria II BD cell sorter from mouse tissues (liver, and brain) from 3 mice for each genotype. Gating of single live cells is performed using side (SSC-A) and forward scatter (FSC-A) gating, and doublet exclusion using forward scatter width (FSC-W) against FSC-A as well as dead cell exclusion with DAPI. Macrophages are identified after gating on CD45 + CD1 lb k>w F4/80 Wgl1 cells in liver and

CD45 low CDl lb + F4/80 + in the brain. 200 cells for each sample are sorted directly into a 96-well plate in 4μ1 of H 2 0 containing 0.2% of tritonX and 0.8υ/μ1 of RNAse inhibitor and processed for sequencing by the Integrated Genomics Operation (IGO) at MSKCC. Processing of RNAseq data is performed with the help of the bioinformatics core at the MSKCC. The raw count matrix generated by HTSeq are processed using the R/Bioconductor package DESeq, which is used to both normalize the full dataset and analyze differential expression between sample groups. Gene Ontology (GO) analysis is performed using the GO analysis function in GeneSpring GX 13.0 (Agilent), with the p-value calculated using a hypergeometric test with Benjamini-Yekutieli correction. For example, genes with a fold change difference of ± 2 between cells from

BRAF V600E and controls animals are selected. Significantly regulated genes (t-test p< 0.05; FDR <0.05) from this selection are grouped into GO terms. Heatmaps are generated using GeneSpring GX 13.0 (Agilent). All other analyses and plotting are performed in R. [0118] This analysis identifies molecular pathways and candidates genes involved in

BRAF V600E microglia activation. Following from these results, candidate genes and pathways involved for example in cytotoxicity, phagocytosis, or inflammation are selected for further study as they may represent mechanisms for tissue damage. RNASeq analysis in macrophages is performed in accordance with methods known in the art.

[0119] Determining the pathological consequences of targeting BRAF V600E outside the brain in vivo. The presence of BRAF V600E macrophages clones was not restricted to the nervous system of Cs Jr MenCreMer . BRA pV6 o E . Rosa26 LSL - YFP m [ ce ^ a nd was also documented particularly in the liver

(FIGs. 5 and 11)

Example 9: Determining if development of liver injury is accelerated by the presence of BRAF V600E -expressing Kupffer cells.

[0120] Some degree of liver involvement is frequent in histiocytosis and severe sclerosing cholangitis with liver fibrosis occurs in -5% of patients and has a high mortality. Our preliminary results showed that Csflr Mer,CreMer ; BRAF V600E ; Rosa26 LSL - YFP mice develop focal accumulation of histiocytes in the liver (FIGs. 5C and 5D;FIG. 11). Flow cytometric analysis shows that YFP and thus BRAF expressing Kupffer cell clones were bigger (12-13 % among F4/80 + ) when compared to BRAF WT littermates (1-2 %, FIG. 11 A). In histological analysis, focal collections of histiocytes were noted in multiple sections of liver in BRAF V600E mice (FIG. 11B). However, liver enzyme function assessed by serum analysis of alkaline phosphatase (ALP), alanine aminotransferases (ALT) and aspartate aminotransferases (AST), were normal (FIG. 11C), and a limited histopathological analysis did not observe evidence of cholestasis, interstitial fibrosis in the portal area, or reactive hyperplasia of small bile ducts, which are features of the disease. Therefore it is believed that the clonal Kupffer cell expansion observed in BRAF V600E mice in specific-pathogen free (SPF) conditions does not trigger portal fibrosis or cholangitis on its own. However, this provides an opportunity to manipulate the mice to investigate the impact of environmental factors that could promote the escape from a "silent mode" and trigger liver disease. Csflr Mer,CreMer ; BRAF V600E ; Rosa26 LSL - YFP mice housed under SPF conditions, and fed irradiated chow are protected from many harmful xenobiotics such as carcinogens, drugs, environmental pollutants, food additives, hydrocarbons, and pesticides. To phenocopy possible environmental factors that might accelerate or cause the liver

pathophysiology seen in histiocytosis patients, xenobiotic-induced cholangiopathy will be effected. Csflr MenCreMer ; BRAF V600E ; Rosa26 LSL - YFP mice and BRAF m littermates (n = 8 per genotype) will receive 0.1 % 3,5-diethoxycarbonyl- 1,4-dihydrocollidine (DDC)-chow over a period of 4 weeks, starting at 4 weeks of age. Wild-type mice exhibit sclerosing cholangitis and a biliary type of liver fibrosis in a highly reproducible manner at this time point. Morphological alterations of the liver are accompanied by a continuous increase in ALT levels as an indicator of hepatocyte injury followed by significant elevations of cholestasis parameters, such as AP and bilirubin. Thus, to follow onset and severity of disease, mice are bled from the facial vein, and serum is analyzed for ALT, AP, and bilirubin before the diet and weekly thereafter. Sclerosing cholangitis and fibrosis are analyzed histologically on paraffin sections as well as cryosections, thus livers oiBRAF WT and BRAF V600E animals are pre-fixed with 4 % PFA, or with a modified formalin/methanol/acetone fixation, depending on downstream procedure. H&E and Sirius red stain are the first readout for fibrosis severity. Morphometry of K19-positive cells indicates ductular reaction, and is compared in BRAF V600E mice and BRAF WT littermates by measuring bile duct mass and normalizing it to the size of portal veins. In addition, well-established markers for liver fibrosis such as osteopontin, a profibrogenic cytokine, and a-smooth muscle actin (SMA) staining, which indicates hepatic stellate cell activation, are used to quantify the excess of fibrosis. Resident Kupffer cells (F4/80 and YFP) in BRAF V600E ice and BRAF m littermates are also stained to assess the anatomical proximity of YFP + Kupffer cell clones to the lesion site. The inflammatory phenotype characterised by an infiltration of neutrophils (CD1 lb^yeG^ and monocytes (CD1 Vo^yeC*) is evaluated by flow cytometry. For statistical analyses Student's t- test is performed and all P-values of <0.05 will be considered significant.

[0121] The well-established carbon tetrachloride (CCU) model to induce liver fibrosis in BRAF V600E and BRAF WT littermate controls is used to confirm a possible acceleration of liver fibrosis with BRAF V600E expressing Kupffer cell clones. BRAF m and BRAF V600E littermates (4 week old, n = 8 per genotype) are injected with 1 μΐ/g body weight of a 1 :7 ratio CC1 4 : olive oil mix or just olive oil as control treatment every 5 days for 4 weeks to induce fibrosis. The grade of liver injury is assessed as described above.

Example 10: Assessing and monitoring the extent of organ involvement in Csflr MenCreMer ; BRAF V600E ; Rosa26 LSL YFP using whole animal imaging.

[0122] The following studies are used to characterize extra-neurological involvement and its pathological consequence more broadly. In patients, neurodegeneration as well as liver, lung, and bone disease are associated with MRI and PET/CT radiographic findings. To complete the analysis the presence of lung inflammatory or fibrotic lesions and of bone lytic lesions is investigated by whole animal imaging of a cohort (n = 8 per genotype) of 6 month old

CsfJr MenCreMer . BRA pV600E . Rosa26 ^-ΥΡΡ ^ BRA pWT Uttermates Mice are imaged under anesthesia at the MSKCC small imaging core facility. To identify skeletal lesions, microCT (microCAT II Imtek Corp, Oak Ridge, TN), which yields spatial resolution on the order of 100 microns, delivering a radiation dose on the order of 20 cGy, will be used. Soft tissue analysis (lung) will be carried out by whole body PET/CT using FDG. In addition, these experiments will provide an analysis of the liver, and will also image the brain of these mice using a 7 tesla MRI with gadolinium injection. Data analysis is performed in close collaboration with the MSKCC small imaging core facility. Histological and flow cytometry analysis of lung tissue and bone are performed on groups of BRAF V600E and BRAF WT littermates (n = 5 per genotype). Histology of bone tissue are performed after fixation with 4% paraformaldehyde for 3 days, decalcification for 4 days in 14% EDTA pH 7.1. and dehydration. Histology of lung is performed on paraffin sections, H&E and Sirius red stain are the first readout to analyze the presence of fibrosis. For flow cytometry analysis of the lung tissue, cell suspensions are prepared and stained with antibodies for alveolar and interstitial macrophages (F4/80, CD1 lb, CD45, Siglec-F, CDl lc), T cells (CD3, CD4, CD 8), B cells (CD19), eosinophils, monocytes and neutrophils (Ly6C, Ly6G, Siglec-F, CD1 lb) to identify and quantify an inflammatory infiltrate. Live-dead discrimination is performed by Hoechst or 7-AAD staining.

[0123] Results from these imaging studies complete the characterization of the model, providing a strategy to follow disease activity that could be used in aim 3, and may reveal the presence of bone and/or lung lesions in the Csflr er,CreMer ; Rosa26 LSL - YFP ; BRAF V600E mice. Soft tissue lesions that may correspond to inflammatory granuloma, demyelination, and bone defects are expected to be visible with MRI, FDG-PET/CT and microCT permitting observation of the progression of disease in live mice.

Example 11: Determining whether targeting of a small number of HSCs also results in the differentiation of BRAF V600E macrophages and features of histiocytoses (particularly neurodegenerative disease).

[0124] The strategy of targeting the BRAF V600E mutation to a small number of EMPs appears to model some aspects of histiocytic disorders, while targeting of HSCs in contrast leads to a leukemic disorder. However, it is possible that targeting a small proportion of HSCs, as has been done for EMPs, may also lead to histiocytic phenotype. BRAF V600E ;Rosa26 LSL~YFP crossed with Cxcr4 CreERT2 mice, which were recently characterized with the lab of Ralf Stumm (Jena

University, Germany) are analyzed. Cxcr4 is expressed in HSCs but not in EMPs, fetal macrophages, or adult tissue-resident macrophages. A low-dose (20mg/kg) pulse of OH-TAM at E9.5 allows transient and limited induction of Cre expression to pulse-label -10% of HSCs in adults (FIG. 4). If expression of BRAF V600E in LSK in embryos is lethal and embryos results in a leukemic phenotype in utero, the dose of OH-TAM is titrated down and mice are treated postnatally. Blood and bone marrow, brain, liver, spleen, and lung of Cxcr4 CreERT1 '; BRAF V600E ; Rosa26 LSL - YFP mice (n=5) and their littermates are then analyzed at 4 weeks by multiparameter flow cytometry analysis. Cell suspensions prepared from the indicated organs are stained using antibodies directed against hematopoietic progenitors (CD45, Kit, Seal, CD48, CD 150), myeloid cells (CD115, Ly6C, CDl lb, CDl lc, MHCII, F4/80, SiglecF) and lymphoid cells (CD3, CD4, CD8, CD 19). Results are analyzed with Flow Jo and with the PhenoGraph algorithm, which partitions high-dimensional single-cell data as obtained by multi-color flow cytometry into subpopulations without a priori gating. If flow cytometry suggests expansion of macrophages and/or the presence of an inflammatory infiltrate, histological analysis of bone marrow, brain, liver, spleen, and lung is performed to look for granulomas, inflammatory infiltrates, and fibrosis using H&E and Sinus red stains and immunostaining for YFP and macrophage antigens F4/80 and CD68. If Cxcr4 CreERT2 ;BRAF V600E ;Rosa26 LSL - YFP mice are viable and do not present with an overt phenotype at 4 weeks, a cohort oi Cxcr4 CreERT2 ;BRAF V600E ;Rosa26 LSL - YFP and littermates are monitored for the development of an histiocytic phenotype for >6 months and are then autopsied for histopathological and FACS analysis to examine for infiltration in hematopoietic tissues, liver, lung, and brain. In case of positive results, to ensure that a phenotype is due to cell- autonomous expression oiBRAF V600E in HSC-derived cells, BRAF V600E and BRAF V600E~ LSK from Cxcr4 CreERT2 ;BRAF V600E ;Rosa26 LSL - YFP mice are co-transplanted into lethally irradiated (2x450cGy) wild type recipients and these recipient mice are analyzed as described above.

Example 12: Testing whether the pathological changes associated with BRAF V600E expression in macrophages are reversible or can be prevented by BRAF inhibitors.

[0125] Whether PLX4720 can prevent the onset of neurological disease by continuous feeding of Csflr MenCreMer ; BRAF V600E ; Rosa26 LSL - YFP mice with PLX4720-impregnated chow (417mg/kg, Research Diets, Inc.) was investigated. A cohort of 3 month-old BRAF V600E and BRAF WT littermates were placed on PLX4720 or control diet. As a readout, hind limb clasping reflexes were examined every week and footprint assays were performed as described above before treatment and every 4 weeks during treatment. Preliminary results evaluating the cumulative incidence of the ataxic phenotype in this small group of mice suggested that the PLX4720-diet delays but does not totally prevent the onset of neurologic disease (FIG. 12). Example 13: Assessing the effects of BRAF inhibitor treatment in vivo on the incidence and severity of neurological disease and microglial activation.

[0126] Cohorts of age matched Csflr MenCreMer ; BRAF V600E ; Rosa26 LSL - YFP mice and BRAF m littermate controls (n=10 for each genotype and treatment group) are placed on PLX4720- impregnated (417mg/kg) or control chow from the age of 4 weeks and 3 months. Incidence and severity of the neurological phenotype are assessed by measurement of hind-limb clasping reflex every week and footprint assays every two weeks; rotarod analysis is performed monthly and the open field test and MRI (depending on results from Example 10) are performed on mice that do not exhibit neurological symptoms by 8 months. Surviving mice are sacrificed for pathological analysis (H&E, Luxol fast blue stainings, and IHC for GFP, Ibal, and pERK) and flow cytometry analysis as in Example 7 to assess the extent of microglial activation and the inflammatory infiltrate. On the same set of mice, RNAseq analysis is performed according to the protocol described in Example 8 to assess change of the BRAF V600E signature upon BRAF inhibitor treatment. Briefly, mice are sacrificed by 8 months and microglial cells FACS sorted (200 cell into 96-well plates, to assess those gene expression changes affected by the BRAF V600E activation (comparison of BRAF V600E to BRAF WT control treated mice) and those altered by PLX4720-treatment in each BRAF genotype. In addition to longitudinal experiments to assess for prevention of development of neurodegenerative disease, mice with established

neurodegenerative disease are treated. Cohorts of age matched c s f] ienCreMer ; BRAF V600E ;

Rosa26 LSL - YFP mice and BRAF WT littermate controls with established neurologic disease as assessed by the physiologic, behavioral, and radiographic assays described in Example 6 (n=10 for each genotype and treatment group) are placed on PLX4720-impregnated (417mg/kg) or control chow. Mice are evaluated using hind-limb clasping reflex, footprint assays, rotarod analysis, open field test, and MRI using the methods and schedule above. The effect of treatment on survival of these mice are evaluated using Kaplan-Meier estimates of survival (with log-ranked (Mantel-Cox) used to determine statistical significance).

Example 14: Conserved pathways and genes regulated by BRAF V600E expression in mouse and human histiocytosis lesions.

[0127] Prior and ongoing RNAseq analysis of human histiocyte tissue biopsy samples are utilized to correlate with the isogenic tissue-specific RNAseq data obtained from murine BRAF V600E and BRAF WT macrophages as described above. Currently this includes RNAseq patient tissue biopsies from brain, skin, and bone (n=10 distinct patients from each site, approximately half of whom are BRAF V600E mutant). Results from the murine RNAseq studies are compared with our human RNAseq data from patients to identify i) genes differentially expressed in the presence of the BRAF V600E mutation in mouse and human macrophages that are mutant versus WT for BRAF regardless of tissue source, and ii) BRAF V600E -dependent transcriptional changes that are tissue-specific. Also included in the analysis will be published microarray data from sorted CD207 + cells (presumably pathologic macrophages) from biopsies of LCH patients. Gene Set Enrichment Analysis (GSEA) is performed on the various human samples and the murine macrophages using the C7 collection (immunologic signatures) offered by the Broad institute, as has been done previously. Expression of candidate genes common to mouse and human cells are validated at the protein level using flow cytometry,

immunohistochemistry, and/or ELISA/Luminex technology (available in the laboratory) in murine and human tissues and cells.

[0128] Methods for the isolation of resident macrophages from the nervous system, and methods for determining the BRAF V600E status of those cells are well known. Cell suspensions prepared from brain or spinal cord of Csflr MenCreMer ; BRAF V600E ; Rosa26 LSL - YFP mice are stained using antibodies directed against macrophages (CD45, CD1 lb, F4/80). YFP expression allows to sort BRAF V600E+ cells. The mutation can be detected in the Braf locus after RNAseq or alternatively by digital PCR, if the sequenced reads do not cover the whole locus.

Example 15: Somatic mosaicism for a BRAF V600E allele and YFP in resident macrophage lineage using inducible genetic targeting in ( s jj t IerCreMer m i ce>

[0129] Somatic mosaicism was achieved for a BRAF V600E allele and yellow fluorescent protein (YFP) in the resident macrophage lineage using inducible genetic targeting in Csflr MerCreMer mice (FIG. 13A and FIG. 17). Mice reached weaning age in normal Mendelian ratio (n=342, FIG. 13B). YFP expression was absent from HSC-derived cells in the bone marrow and blood but was detected in tissue F4/80+ macrophages (FIGs. 13C-13E). RNA-seq analysis of sorted YFP+ F4/80+ macrophages confirmed expression of BRAF V600E transcripts in BRAF^ mice (FIG. 13F). The proportion of F4/80+ YFP+ macrophages were increased in tissues from BRAF^ mice in comparison to littermates (FIG. 13E). In situ Ki67, phospho-Hi stone H3, and cleaved caspase 3 staining of brain microglia indicated an increased proliferative index and decreased apoptosis in BRAF V600E YFP+ microglia (FIG. 13G and FIG. 17F). RNA-seq analysis of Kupffer cells and microglia from BRAF^ mice and littermates identified a mitotic gene expression signature, as well as expression of ERK target genes, inflammatory cytokines and lectins (FIGs. 13H and 131; FIG. 17G) Nevertheless, histological and flow cytometry analysis of liver, brain, lung, kidney, bone marrow, and spleen from young BRAF^ mice revealed no overt abnormalities, and in particular no tumoral or leukemic phenotypes (FIG. 17H). This is in contrast to results obtained when targeting BRAF V600E alleles in HSC using Vav Cre ; BRAF LSL"V600E mice and Csflr iCre ; BRAF LSL"V600E mice or in more mature HSC-derived myeloid precursors as achieved in CD1 lc Cre ; BRAF LSL"V600E mice (FIGs. 18 and 19). In each of these models, expression of BRAF V600E in HSCs or HSC-derived cells resulted in a highly penetrant (100%) leukemic or tumoral histiocytic phenotype in the bone marrow, spleen and lung. Altogether, these data show that targeted expression of a BRAF V600E allele in EMP does not lead to leukemic/tumoral transformation, in contrast to the targeting of HSC-derived progenitors, and results in otherwise healthy mice carrying clones of BRAF V600E resident macrophages endowed with a small proliferative advantage.

Example 16: Effects of BRAF V600E macrophage clones in adult tissues.

[0130] To determine the potential effects of BRAF V600E macrophage clones in adult tissues, a large cohort (n=155) of BRAF VE mice and littermate controls were analyzed. BRAF VE mice developed a slowly progressive neurologic impairment (FIGs. 14A-14C and FIG. 20). Young mice did not present with detectable neurological symptoms, as evaluated by the rotarod tes. However, from the age of 4-5 months, BRAF VE mice presented with axial rolling, a unilateral loss of hindlimb reflexes, and unilateral increase in overlap distance between hind and front paws followed by decreased stride length as measurd by footprint assays (score 1) (FIGs. 14A and 14D). BRAF VE mice later developed hindlimb paresis or loss of hindlimb reflexes (score 2), associated with weight loss. At a more advanced stage, BRAF VE mice presented in rapid succession with paralysis of one and subsequently both hindlimbs (score 3 and 4) and full loss of hindlimb reflexes (FIGs. 14A and 14B), at which time mice were flagged as moribund by veterinary staff and were sacrificed. Cumulative evidence of score 1 criteria indicated that -90% of male and female BRAF VE mice presented with a neurological disease by 7 months of age, and -60%) of BRAF VE mice had progressed to full paralysis by 9 months of age (FIGs. 14C-14F). To further investigate if the neurologic disease was dependent on constitutively active BRAF, BRAF VE and BRAF m mice were placed on an ad libitum diet containing a BRAF inhibitor (PLX4720, FIG. 20) at one and three months of age. PLX4720 treatment delayed the onset of the neurological phenotype when initiated early {e.g., at one month of age) and mitigated disease progression in both cases. By 9 months of age, 80% ofBRAF VE mice treated with PLX4720 were alive with an average clinical score of 2 (FIGs. 14D-14F). These results demonstrate that BRAF V600E mosaicism in tissue-resident macrophages causes a late-onset progressive

neurological disorder with features of cerebellar ataxia, also found in patients with cerebral histiocytoses. [0131] The pathological changes that may underlie neurologic impairment were investigated in asymptomatic one-month old BRAF VE mice, and early signs of microglial and astrocyte activation, without signs of neuronal damage, such as deposit of amyloid precursor protein (APP) in the brain or neuronal loss (FIG. 21) were found. However, in ~6 month-old BRAF VE mice, large clusters of amoeboid Ibal+ microglia were present in the cerebrum, preferentially located in the thalamus, brain stem, cerebellum, and spinal cord (FIGs. 14G and 14H; FIG. 22). In these areas, Lamp2+ phagocytic microglia, marked astrogliosis, synaptic and neuronal loss, and APP deposits (FIGs. 14G and 14H) were observed. Luxol Fast Blue/PAS staining also indicated demyelination, potentially secondary to axonal damage (FIG. 22). BRAF inhibition reduced microglia accumulation and astrogliosis, phagocytosis, demyelination, neuronal loss, and APP deposition (FIG. 14H and FIG. 22). Thus, mice presented with a neurodegenerative disease driven by constitutive active BRAF in microglia. Accordingly, this example

demonstrates that BRAF inhibitors, such as those described herein, are useful in methods of treating the symptoms of neurodegenerative disease.

Example 17: BRAF inhibition mitigates microglial accumulation.

[0132] Immunostaining for CD68, Ibal, YFP, and pERK demonstrated that microglial clusters represented the accumulation of EMP-derived (YFP + ) pERK + microglia (FIGs. 15A and 15B; FIG. 23). Microglial ERK-activation in brain and spinal cord were confirmed by western blot on whole tissue and by confocal microscopy (FIGs. 15C and 15D). Immunofluorescence analyses also indicated that pERK + YFP + amoeboid microglia had lost their ramifications (FIG. 15D). Flow cytometry analysis confirmed the preferential expansion of YFP + microglia (FIGs. 15E and 15F). Granulocytes, monocytes, and B-cells were absent or rare, but CD3 + CD8 + , CD4 + , and Foxp3 + T-cells were increased (FIG. 15F and FIG. 23). Finally, BRAF inhibition mitigated the accumulation of YFP + microglia, likely by decreasing their proliferation (FIGs. 15E and 15G), and prevented ERK phosphorylation and the amoeboid phenotype of microglia (FIGs. 15B-3D). Altogether, the data suggest that BRAF V600E somatic mosaicism in microglia drives a neurodegenerative disease mediated by microglial activation. Accordingly, this example demonstrates that BRAF inhibitors, such as those described herein, are useful in methods of treating symptoms of neurodegenerative disease. Example 18: Analysis of cellular and molecular mechanisms operating in microglia to drive neurodegenerative disease.

[0133] The cellular and molecular mechanisms that may operate in microglia to drive neurodegenerative disease were investigated. RNA-seq analysis of FACS-sorted YFP + microglia from paralyzed BRAF^ mice and control littermates identified -8000 differentially expressed genes (DEG), 80% of them being upregulated (data not shown). GSEA analysis of DEG identified Ras signaling, complement activation, and inflammatory response signatures, including expression of II lb, 1118, 116 and 1117, genes associated with phagocytosis, such as cathepsins and pattern recognition receptors, growth factors, and growth factor receptors usually expressed by fibroblasts, and matrix-associated genes laminins and collagens (FIGs. 16A and 16B). Expression of Dectin-1 (Clec7a), LFA-1 (CDl la), VLA-4 (CD49d), the Pdgf receptor, II lb, and 1117a, and their decrease following PLX4720 treatment was confirmed at the protein level and collagen IV and VI deposition was observed at sites of YFP + microglia accumulation (FIGs. 16C-16F). These results indicate that the pathological consequences of microglial ERK activation include the accumulation of amoeboid microglia producing inflammatory cytokines of the 111/18 and 116/17 families, which are mediators of neuronal loss and neurodegeneration and local matrix remodeling, including the production of collagen. Accordingly, these results demonstrate that BRAF inhibitors, such as those described herein, are useful in methods of treating neurodegenerative diseases.

[0134] Of note, apart from the hindbrain, midbrain and thalamus, expansion of YFP + F4/80 + pERK + macrophages was also detected in the forebrain and in organs outside of the central nervous system, such as the liver (FIGs. 24A-24C). However, histological or biological signs of liver, spleen, lung, or kidney damage in mice housed in SPF conditions in the course of this study (FIGs. 24D-24F) was not observed. Possible explanations for the intriguing finding that ERK-activated macrophages are better tolerated outside the posterior part of the brain may relate to cell-autonomous or cell-extrinsic homeostatic mechanisms that operate in various anatomical locations and tissue niches, such as the greater exposure to microbial and environmental stimuli in the liver.

[0135] Results from model described herein show that neurodegenerative diseases in histiocytoses patients may involve the accumulation of ERK-activated microglia, producing inflammatory cytokines and collagen at sites of neurodegeneration. Brain tissue from 5 patients with ECD-, LCH-, and Juvenile Xanthogranuloma- (JXG) associated neurodegenerative disease, carrying a BRAF mutation (FIG. 25A) was analyzed. In ECD patients, microglial activation, in the form of numerous amoeboid BRAF V600E+ CD163 + microglia with nuclear pERK + staining at sites of neuronal loss, astrogliosis, and demyelination (FIGs. 16G and 16H; FIGs. 25B and 25C) was observed. Comparison of the transcriptome from the JXG and LCH brain biopsies with control brain tissue also identified a MAPK-pathway activation signature, and indicated increased expression of ILlb, IL18, IL6 and IL17A, pattern recognition receptors and cathepsins, as well as PDGF receptor and collagen genes (FIG. 161). Therefore, brain tissues from human patients shared key histological, cellular and molecular features observed in the mouse.

Example 19: Analysis of somatic mutations in brain cells from human samples.

[0136] Microglia represent only -5% (2-10%) of brain cells, thus sequencing of whole brain tissue at current sequencing depths is unlikely to reliably detect somatic mutations in microglial clones. In addition, fresh brain tissue is in general not available for genetic studies. This example describes a protocol to extract and purify nuclei from microglia (Pu. l+ NeuN-), neurons (Pu. l- NeuN+), and double negative cells (Pu. l- NeuN-) from frozen brain samples by

immunotagging nuclei suspensions with antibodies (FIGs. 26A-26B).

[0137] Protocol for isolation of nuclei from individual brain cell nuclei in frozen samples.

Different nuclei sorting protocols were adapted to extract and FACS-sort brain cell nuclei from frozen samples (FIGs. 26A-26B). In brief, ~350mg of frozen tissue is homogenized in a sterile Dounce tissue grinder using a nonionic surfactant based buffer (250 mM sucrose, 25 mM KC1, 5 Mm MgC12, 10 mM Tris buffer, pH 8.0, Triton X-100 0.1% (v/v), DAPI 3μΜ and nuclease-free water) that lyses the cell membrane while releasing intact nuclei. To improve the efficiency and purity of the nuclei sorting, the homogenate preparation is followed with a sucrose-iodixanol gradient centrifugation to obtain clean nuclei suspensions. Samples are immunotagged first with anti-NeuN-PE (clone A60; Milli-Mark, 1 :500, 45 min), followed by fixation (15 min) and permeabilization (eBioscience Transcription Factor Staining Buffer Set, eBioscience) and immunotagging with anti-Pu. l-Alexa Fluor 647 (clone 9G7, Cell Signaling, 1 :50, 45 min). After fixation and permeabilization, centrifugations are carried out without breaks to reduce nuclei loss. Samples are FACS-sorted in a BD FACSAria with a 100-μπι nozzle and a sheath pressure 20 psi, operating at 1000 events per second. Microglia (Pu. l+ NeuN-), neuron (Pu. l- NeuN+) and double negative (DN) cells (Pu. l- NeuN-) are sorted in bulk into 1.5 ml tubes, pre-coated with 10% BSA (FIG. 26A). After sorting, nuclei are centrifuged for 20 minutes at 6000g. Nuclei pellets are processed for DNA extraction immediately using QIAamp DNA Mini Kit (QIAGEN) following manufacture recommendations. Samples are processed at all times in an Air Clean PCR Workstation. Criteria for sequencing: resulting DNA is analyzed for quality and quantity with Agilent 4200 TapeStation, and Quant-it (ThermoScientific), respectively. Samples with a DNA integrity number (DIN) >6 and a total quantity >200ng will proceed to library preparation. Based on extensive testing, it has been determined that isolation of brain nuclei from ~350mg (>150.000 nuclei p/population) of tissue using this protocol reproducibly allows DNA extraction with yield and quality for deep sequencing (data not shown, FIGs. 26A-26B and 27A-27B).

[0138] Protocol for library preparation, targeted sequencing using the MSKCC Heme-PACT platform. To explore the presence of mutations in the microglia genome targeted deep DNA sequencing of microglia, neurons, double negative, and blood was performed using the Heme- PACT sequencing panel. Matched samples obtained in (g) are sequenced in the Integrated Genomics Operation (IGO) at Memorial Sloan Kettering (MSKCC). Before sequencing, DNA samples are normalized to a yield 200ng of input (-150.000 cells) and diluted in 55 μΐ of TE buffer on the Biomek FXP Laboratory Automation Workstation (Beckman Coulter) before shearing on the Covaris instrument. Sequencing libraries are prepared on the Biomek FXP through a series of enzymatic steps including shearing, end-repair, A-base addition, ligation of barcoded sequence adaptors and low-cycle PCR amplification (Kapa Biosystems, Roche).

Libraries are analyzed for quantity and are combined in pools of 24-36 libraries for multiplexed capture using custom-designed biotinylated probes (NimbleGen). Captured DNA fragments are sequenced on an Illumina HiSeq 2500 as paired-end 100-bp reads. All DNA samples are sequenced to the same coverage (>400x), which will allow multiple comparisons during the analysis.

Example 20: Identification of BRAF somatic mutations in microglia from a human patient with LCH and neurodegenerative disease.

[0139] Brain, blood, and skin samples from a patient (patient A) from the French Histiocytosis Study Group database/biobank were analyzed. Prior written informed consent was obtained from the patient with histiocytosis for DNA sequencing and immunohistochemical studies according to the Helsinki convention. This study received approval from the Institutional Review Board at MSKCC (IRB Protocol XI 7-047). This patient presented with a BRAF V600E mutation in peripheral (skin) macrophages at 6 months of age (histiocytosis, an eczema type lesion that resolves by itself, with topical treatment, or in some case mild chemotherapy) and who later developed a lethal neurodegenerative disease over the following two decades.

[0140] The presence of BRAF V600E was analyzed using ddPCR and the MSKCC Heme-Pact sequencing panel in blood cells and post-mortem brain microglia, neurons, and glial cells from 8 different brain areas involved or not with neurodegenerative process after histolgical

examination. BRAF V600E was detected at allelic frequencies of 9%, 12% and 20% in Pu.1 + cells (microglia) from pons, cerebellum, and hippocampus, respectively (FIGs. 26A-26B and 27 A- 27B), which correspond with brain regions that present with severe histological signs of microgliosis, astrogliosis, and neurodegeneration (FIG. 28). Allelic frequencies of 0.2% and 0.3% were detected in amygdala and midbrain where discrete lesions were also observed. In contrast the BRAF V600E clones were undetectable in neurons and blood cells, and in Pu.1 cells from unaffected areas of the brain (F2, Tl, and medulla) (see FIGs. 26A-26B and 27A-27B).

[0141] Additionally, analysis of mosaicism in blood cells, neurons, and microglia from this patient indicate that microglia mosaicism is private to each brain area and not shared with blood or neurons, which is compatible with an early developmental origin and local clonal evolution of microglia, similar to what has been observed in mice (FIG. 29).

[0142] These results on the presence of BRAF V600E positive macrophages in brain areas with severe neuronal loss and skin, and the absence of BRAF V600E positive cells in blood of a patient with neurodegeneration, recapitulates what has been reported in mice. These results demonstrate that microglia somatic mosaicism for a BRAF V600E mutations is associated and likely causative of neurodegenerative disease.

Example 21: Effect of targeting PIK3CA H1047R to macrophage progenitors.

[0143] Mosaic expression of BRAF V600E mutation in microglia drives a neurodegenerative disease in mice and humans. Whether this phenotype is exclusive of BRAF mutations in the macrophage lineage or if mutations affecting other pathways may also drive a neurodegeneraive disorder remains unexplored. Approximately 10% of histiocytoses patients (17% for Erdheim- Chester disease) present with gain of function somatic mutations in the phosphoinositide-3- kinase-(PI3K)-AKT pathway, PIK3CA H1047R being the most frequent. The PIK3CA H1047R mutation is located in the kinase domain of the protein and leads to elevated kinase activity. Targeting PIK3CA H1074R expression specifically to a small number of EMPs is accomplished by crossing FVB.129S6-Gt RO&4 2^or imifft¾ca*ffl0¥7 ?;¾a J (R26-Pik3ca H1047R ) males to tamoxifen-inducible Csflr MenCreMer females, thereby allowing selective targeting of EMPs when pulsed with OH-TAM at E8.5. Pregnant females receive low doses of OH-TAM via

intraperitoneal injection so that only a small proportion of cells express PIK3CA H1074R

(PIKRCA HR ) (FIG. 30). Mice are born in Mendelian ratios (FIG. 30). Similar to what has been reported on BRAF mice, PIK3CA H1074R mice and not their wild-type littermates, present with neurological symptoms by the age of ~5 months of age as measured by cumulative incidence of behavioral abnormalities (FIG. 31A). Footprint assays showed a unilateral increase in overlap distance between hind and front paws in PIK3CA H1074R mice (FIG. 31B). These results demonstrate that activation of PI3K-AKT pathway in the macrophage lineage, and therefore in microglia, also drives a neurological phenotype as previously observed in BRAF V600E mice. Accordingly, this example suggests that PI 3-kinase inhibitors, such as those described herein, are useful in methods of treating the symptoms of neurodegenerative disease.

References

1. Berres ML, Lim KP, Peters T, Price J, Takizawa H, Salmon H, Idoyaga J, Ruzo A, Lupo PJ, Hicks MJ, Shih A, Simko SJ, Abhyankar H, Chakraborty R, Leboeuf M, Beltrao M, Lira SA, Heym KM, Bigley V, Collin M, Manz MG, McClain K, Merad M, Allen CE. BRAF-V600E expression in precursor versus differentiated dendritic cells defines clinically distinct LCH risk groups. The Journal of experimental medicine. 2014;211(4):669-83. doi: 10.1084/jem.20130977. PubMed PMID: 24638167; PMCID: PMC3978272.

2. Heritier S, Emile JF, Barkaoui MA, Thomas C, Fraitag S, Boudjemaa S, Renaud F, Moreau A, Peuchmaur M, Chassagne-Clement C, Dijoud F, Rigau V, Moshous D, Lambilliotte A, Mazingue F, Kebaili K, Miron J, Jeziorski E, Plat G, Aladjidi N, Ferster A, Pacquement H, Galambrun C, Brugieres L, Leverger G, Mansuy L, Paillard C, Deville A, Armari-Alla C, Lutun A, Gillibert-Yvert M, Stephan JL, Cohen-Aubart F, Haroche J, Pellier I, Millot F, Lescoeur B, Gandemer V, Bodemer C, Lacave R, Helias-Rodzewicz Z, Taly V, Geissmann F, Donadieu J. BRAF Mutation Correlates With High-Risk Langerhans Cell Histiocytosis and Increased Resistance to First-Line Therapy. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2016;34(25):3023-30. doi: 10.1200/JCO.2015.65.9508. PubMed PMID: 27382093.

3. Diamond EL, Durham BH, Haroche J, Yao Z, Ma J, Parikh SA, Wang Z, Choi J, Kim E, Cohen-Aubart F, Lee SC, Gao Y, Micol JB, Campbell P, Walsh MP, Sylvester B, Dolgalev I, Aminova O, Heguy A, Zappile P, Nakitandwe J, Ganzel C, Dalton JD, Ellison DW, Estrada- Veras J, Lacouture M, Gahl WA, Stephens PJ, Miller VA, Ross JS, Ali SM, Briggs SR, Fasan O, Block J, Heritier S, Donadieu J, Solit DB, Hyman DM, Baselga J, Janku F, Taylor BS, Park CY, Amoura Z, Dogan A, Emile JF, Rosen N, Gruber TA, Abdel-Wahab O. Diverse and Targetable Kinase Alterations Drive Histiocytic Neoplasms. Cancer discovery. 2015. doi: 10.1158/2159- 8290.CD-15-0913. PubMed PMID: 26566875.

4. Diamond EL, Abdel-Wahab O, Pentsova E, Borsu L, Chiu A, Teruya-Feldstein J, Hyman DM, Rosenblum M. Detection of an NRAS mutation in Erdheim-Chester disease. Blood. 2013;122(6): 1089-91. doi: 10.1182/blood-2013-02-482984. PubMed PMID: 23929840.

5. Emile JF, Diamond EL, Helias-Rodzewicz Z, Cohen-Aubart F, Charlotte F, Hyman DM, Kim E, Rampal R, Patel M, Ganzel C, Aumann S, Faucher G, Le Gall C, Leroy K, Colombat M, Kahn JE, Trad S, Nizard P, Donadieu J, Taly V, Amoura Z, Abdel-Wahab O, Haroche J. Recurrent RAS and PIK3CA mutations in Erdheim-Chester disease. Blood. 2014; 124(19):3016- 9. doi: 10.1182/blood-2014-04-570937. PubMed PMID: 25150293; PMCID: PMC4224196.

6. Badalian-Very G, Vergilio JA, Degar BA, MacConaill LE, Brandner B, Calicchio ML, Kuo FC, Ligon AH, Stevenson KE, Kehoe SM, Garraway LA, Hahn WC, Meyerson M, Fleming MD, Rollins BJ. Recurrent BRAF mutations in Langerhans cell histiocytosis. Blood. 2010;116(11): 1919-23. Epub 2010/06/04. doi: 10.1182/blood-2010-04-279083. PubMed PMID: 20519626; PMCID: 3173987.

7. Satoh T, Smith A, Sarde A, Lu HC, Mian S, Trouillet C, Mufti G, Emile JF, Fraternali F, Donadieu J, Geissmann F. B-RAF Mutant Alleles Associated with Langerhans Cell Histiocytosis, a Granulomatous Pediatric Disease. PloS one. 2012;7(4):e33891. Epub 2012/04/17. doi: 10.1371/journal.pone.0033891. PubMed PMID: 22506009; PMCID: 3323620.

8. Haroche J, Charlotte F, Arnaud L, von Deimling A, Helias-Rodzewicz Z, Hervier B, Cohen-Aubart F, Launay D, Lesot A, Mokhtari K, Canioni D, Galmiche L, Rose C, Schmalzing M, Croockewit S, Kambouchner M, Copin MC, Fraitag S, Sahm F, Brousse N, Amoura Z, Donadieu J, Emile JF. High prevalence of BRAF V600E mutations in Erdheim-Chester disease but not in other non-Langerhans cell histiocytoses. Blood. 2012; 120(13):2700-3. doi: 10.1182/blood-2012-05-430140. PubMed PMID: 22879539.

9. Hyman DM, Puzanov I, Subbiah V, Faris JE, Chau I, Blay JY, Wolf J, Raje NS, Diamond EL, Hollebecque A, Gervais R, Elez-Fernandez ME, Italiano A, Hofheinz RD, Hidalgo M, Chan E, Schuler M, Lasserre SF, Makrutzki M, Sirzen F, Veronese ML, Tabernero J, Baselga J. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. The New England journal of medicine. 2015;373(8):726-36. doi: 10.1056/NEJMoal502309. PubMed PMID: 26287849; PMCID: PMC4971773.

10. Hyman DM, Diamond EL, Vibat CR, Hassaine L, Poole JC, Patel M, Holley VR, Cabrilo G, Lu TT, Arcila ME, Chung YR, Rampal R, Lacouture ME, Rosen N, Meric-Bernstam F, Baselga J, Kurzrock R, Erlander MG, Janku F, Abdel-Wahab O. Prospective blinded study of BRAFV600E mutation detection in cell-free DNA of patients with systemic histiocytic disorders. Cancer discovery. 2015;5(1):64-71. doi: 10.1158/2159-8290.CD-14-0742. PubMed PMID: 25324352.

11. Charles J, Beani JC, Fiandrino G, Busser B. Major response to vemurafenib in patient with severe cutaneous Langerhans cell histiocytosis harboring BRAF V600E mutation. Journal of the American Academy of Dermatology. 2014;71(3):e97-9. doi: 10.1016/j .jaad.2014.03.038. PubMed PMID: 25128147.

12. Haroche J, Cohen-Aubart F, Emile JF, Arnaud L, Maksud P, Charlotte F, Cluzel P, Drier A, Hervier B, Benameur N, Besnard S, Donadieu J, Amoura Z. Dramatic efficacy of vemurafenib in both multisystemic and refractory Erdheim-Chester disease and Langerhans cell histiocytosis harboring the BRAF V600E mutation. Blood. 2013;121(9): 1495-500. doi: 10.1182/blood-2012-07-446286. PubMed PMID: 23258922. 13. Pettirossi V, Santi A, Imperi E, Russo G, Pucciarini A, Bigerna B, Schiavoni G, Fortini E, Spanhol-Rosseto A, Sportoletti P, Mannucci R, Martelli MP, Klein-Hitpass L, Falini B, Tiacci E. BRAF inhibitors reverse the unique molecular signature and phenotype of hairy cell leukemia and exert potent antileukemic activity. Blood. 2015; 125(8): 1207-16. doi: 10.1182/blood-2014- 10-603100. PubMed PMID: 25480661; PMCID: PMC4366655.

14. Perdiguero EG, Geissmann F. The development and maintenance of resident macrophages. Nature immunology. 2015; 17(l):2-8. doi: 10.1038/ni.3341. PubMed PMID: 26681456.

15. Schulz C, Gomez Perdiguero E, Chorro L, Szabo-Rogers H, Cagnard N, Kierdorf K, Prinz M, Wu B, Jacobsen SE, Pollard JW, Frampton J, Liu KJ, Geissmann F. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science. 2012;336(6077):86-90. doi: 10.1126/science.1219179. PubMed PMID: 22442384.

16. Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L, Garner H, Trouillet C, de Bruijn MF, Geissmann F, Rodewald HR. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature. 2015;518(7540):547-51. doi: 10.1038/naturel3989. PubMed PMID: 25470051.

17. Mass E, Ballesteros I, Farlik M, Halbritter F, Gunther P, Crozet L, Jacome-Galarza CE, Handler K, Klughammer J, Kobayashi Y, Gomez-Perdiguero E, Schultze JL, Beyer M, Bock C, Geissmann F. Specification of tissue-resident macrophages during organogenesis. Science. 2016;353(6304). doi: 10.1126/science.aaf4238. PubMed PMID: 27492475.

18. Kierdorf K, Erny D, Goldmann T, Sander V, Schulz C, Perdiguero EG, Wieghofer P, Heinrich A, Riemke P, Holscher C, Muller DN, Luckow B, Brocker T, Debowski K, Fritz G, Opdenakker G, Diefenbach A, Biber K, Heikenwalder M, Geissmann F, Rosenbauer F, Prinz M. Microglia emerge from erythromyeloid precursors via Pu. l- and Irf8-dependent pathways. Nat Neurosci. 2013; 16(3):273-80. doi: 10.1038/nn.3318. PubMed PMID: 23334579.

19. Gomez Perdiguero E, Geissmann F. Myb-independent macrophages: a family of cells that develops with their tissue of residence and is involved in its homeostasis. Cold Spring Harbor symposia on quantitative biology. 2013;78:91-100. doi: 10.1101/sqb.2013.78.020032. PubMed PMID: 24122769.

20. Yamasaki R, Lu H, Butovsky O, Ohno N, Rietsch AM, Cialic R, Wu PM, Doykan CE, Lin J, Cotleur AC, Kidd G, Zorlu MM, Sun N, Hu W, Liu L, Lee JC, Taylor SE, Uehlein L, Dixon D, Gu J, Floruta CM, Zhu M, Charo IF, Weiner HL, Ransohoff RM. Differential roles of microglia and monocytes in the inflamed central nervous system. The Journal of experimental medicine. 2014;211(8): 1533-49. doi: 10.1084/jem.20132477. PubMed PMID: 25002752; PMCID: PMC4113947. 21. Gosselin D, Link VM, Romanoski CE, Fonseca GJ, Eichenfield DZ, Spann NJ, Stender JD, Chun HB, Garner H, Geissmann F, Glass CK. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell. 2014; 159(6): 1327-40. doi: 10.1016/j .cell.2014.11.023. PubMed PMID: 25480297; PMCID: PMC4364385.

22. Lavin Y, Winter D, Blecher-Gonen R, David E, Keren-Shaul H, Merad M, Jung S, Amit I. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell. 2014; 159(6): 1312-26. doi: 10.1016/j .cell.2014.11.018. PubMed PMID: 25480296; PMCID: PMC4437213.

23. Bhatia S, Nesbit ME, Jr., Egeler RM, Buckley JD, Mertens A, Robison LL. Epidemiologic study of Langerhans cell histiocytosis in children. The Journal of pediatrics. 1997;130(5):774-84. PubMed PMID: 9152288.

24. Wnorowski M, Prosch H, Prayer D, Janssen G, Gadner H, Grois N. Pattern and course of neurodegeneration in Langerhans cell histiocytosis. The Journal of pediatrics. 2008; 153(1): 127- 32. doi: 10.1016/j jpeds.2007.12.042. PubMed PMID: 18571550.

25. Donadieu J, Chalard F, Jeziorski E. Medical management of langerhans cell histiocytosis from diagnosis to treatment. Expert opinion on pharmacotherapy. 2012; 13(9): 1309-22. doi: 10.1517/14656566.2012.688028. PubMed PMID: 22578036.

26. Diamond EL, Durham BH, Haroche J, Yao Z, Ma J, Parikh SA, Wang Z, Choi J, Kim E, Cohen-Aubart F, Lee SC, Gao Y, Micol JB, Campbell P, Walsh MP, Sylvester B, Dolgalev I, Aminova O, Heguy A, Zappile P, Nakitandwe J, Ganzel C, Dalton JD, Ellison DW, Estrada- Veras J, Lacouture M, Gahl WA, Stephens PJ, Miller VA, Ross JS, Ali SM, Briggs SR, Fasan O, Block J, Heritier S, Donadieu J, Solit DB, Hyman DM, Baselga J, Janku F, Taylor BS, Park CY, Amoura Z, Dogan A, Emile JF, Rosen N, Gruber TA, Abdel-Wahab O. Diverse and Targetable Kinase Alterations Drive Histiocytic Neoplasms. Cancer Discov. 2016;6(2): 154-65. doi: 10.1158/2159-8290.CD-15-0913. PubMed PMID: 26566875; PMCID: PMC4744547.

27. Pritchard C, Carragher L, Aldridge V, Giblett S, Jin H, Foster C, Andreadi C, Kamata T. Mouse models for BRAF -induced cancers. Biochemical Society transactions. 2007;35(Pt 5): 1329-33. doi: 10.1042/BST0351329. PubMed PMID: 17956344; PMCID: PMC2637606.

28. Gibney GT, Messina JL, Fedorenko IV, Sondak VK, Smalley KS. Paradoxical oncogenesis— the long-term effects of BRAF inhibition in melanoma. Nature reviews Clinical oncology. 2013; 10(7):390-9. doi: 10.1038/nrclinonc.2013.83. PubMed PMID: 23712190; PMCID: PMC3983565.

29. Tiacci E, Trifonov V, Schiavoni G, Holmes A, Kern W, Martelli MP, Pucciarini A, Bigerna B, Pacini R, Wells VA, Sportoletti P, Pettirossi V, Mannucci R, Elliott O, Liso A, Ambrosetti A, Pulsoni A, Forconi F, Trentin L, Semenzato G, Inghirami G, Capponi M, Di Raimondo F, Patti C, Arcaini L, Musto P, Pileri S, Haferlach C, Schnittger S, Pizzolo G, Foa R, Farinelli L, Haferlach T, Pasqualucci L, Rabadan R, Falini B. BRAF mutations in hairy-cell leukemia. The New England journal of medicine. 2011;364(24):2305-15. doi: 10.1056/NEJMoal014209. PubMed PMID: 21663470; PMCID: PMC3689585.

30. Chung SS, Kim E, Park JH, Chung YR, Lito P, Teruya-Feldstein J, Hu W, Beguelin W, Monette S, Duy C, Rampal R, Telis L, Patel M, Kim MK, Huberman K, Bouvier N, Berger MF, Melnick AM, Rosen N, Tallman MS, Park CY, Abdel-Wahab O. Hematopoietic stem cell origin of BRAFV600E mutations in hairy cell leukemia. Science translational medicine. 2014;6(238):238ra71. doi: 10.1126/scitranslmed.3008004. PubMed PMID: 24871132; PMCID: PMC4501573.

31. Chen MJ, Yokomizo T, Zeigler BM, Dzierzak E, Speck NA. Runxl is required for the endothelial to haematopoietic cell transition but not thereafter. Nature. 2009;457(7231):887-91. Epub 2009/01/09. doi: 10.1038/nature07619. PubMed PMID: 19129762; PMCID: 2744041.

32. Bertrand JY, Jalil A, Klaine M, Jung S, Cumano A, Godin I. Three pathways to mature macrophages in the early mouse yolk sac. Blood. 2005; 106(9):3004-11. Epub 2005/07/16. doi: 10.1182/blood-2005-02-0461. PubMed PMID: 16020514.

33. Palis J, Robertson S, Kennedy M, Wall C, Keller G. Development of erythroid and myeloid progenitors in the yolk sac and embryo proper of the mouse. Development. 1999;126(22):5073-84. Epub 1999/10/26. PubMed PMID: 10529424.

34. McGrath KE, Frame JM, Fegan KH, Bowen JR, Conway SJ, Catherman SC, Kingsley PD, Koniski AD, Palis J. Distinct Sources of Hematopoietic Progenitors Emerge before HSCs and Provide Functional Blood Cells in the Mammalian Embryo. Cell reports. 2015; 11(12): 1892- 904. Epub 2015/06/23. doi: 10.1016/j .celrep.2015.05.036. PubMed PMID: 26095363; PMCID: 4490098.

35. Bertrand JY, Giroux S, Golub R, Klaine M, Jalil A, Boucontet L, Godin I, Cumano A. Characterization of purified intraembryonic hematopoietic stem cells as a tool to define their site of origin. Proc Natl Acad Sci U S A. 2005; 102(1): 134-9. Epub 2004/12/30. doi: 10.1073/pnas.0402270102. PubMed PMID: 15623562; PMCID: 544043.

36. Sumner R, Crawford A, Mucenski M, Frampton J. Initiation of adult myelopoiesis can occur in the absence of c-Myb whereas subsequent development is strictly dependent on the transcription factor. Oncogene. 2000;19(30):3335-42. Epub 2000/08/05. doi: 10.1038/sj . one.1203660. PubMed PMID: 10918590.

37. Bartunek P, Kralova J, Blendinger G, Dvorak M, Zenke M. GATA-1 and c-myb crosstalk during red blood cell differentiation through GATA-1 binding sites in the c-myb promoter. Oncogene. 2003;22(13): 1927-35. Epub 2003/04/04. doi: 10.1038/sj .onc. l206281. PubMed PMID: 12673198.

38. Kieusseian A, Brunet de la Grange P, Burlen-Defranoux O, Godin I, Cumano A. Immature hematopoietic stem cells undergo maturation in the fetal liver. Development. 2012;139(19):3521-30. Epub 2012/08/18. doi: 10.1242/dev.079210. PubMed PMID: 22899849.

39. Bain CC, Bravo-Blas A, Scott CL, Gomez Perdiguero E, Geissmann F, Henri S, Malissen B, Osborne LC, Artis D, Mowat AM. Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice. Nature immunology. 2014;15(10):929-37. doi: 10.1038/ni.2967. PubMed PMID: 25151491; PMCID: PMC4169290.

40. Hashimoto D, Chow A, Noizat C, Teo P, Beasley MB, Leboeuf M, Becker CD, See P, Price J, Lucas D, Greter M, Mortha A, Boyer SW, Forsberg EC, Tanaka M, van Rooijen N, Garcia-Sastre A, Stanley ER, Ginhoux F, Frenette PS, Merad M. Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity. 2013;38(4):792-804. Epub 2013/04/23. doi: 10.1016/j .immuni.2013.04.004. PubMed PMID: 23601688.

41. Yona S, Kim KW, Wolf Y, Mildner A, Varol D, Breker M, Strauss-Ayali D, Viukov S, Guilliams M, Misharin A, Hume DA, Perlman H, Malissen B, Zelzer E, Jung S. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity. 2013;38(1):79-91. Epub 2013/01/01. doi: 10.1016/j .immuni.2012.12.001. PubMed PMID: 23273845.

42. Metzger D, Clifford J, Chiba H, Chambon P. Conditional site-specific recombination in mammalian cells using a ligand-dependent chimeric Cre recombinase. Proceedings of the National Academy of Sciences of the United States of America. 1995;92(15):6991-5. Epub 1995/07/18. PubMed PMID: 7624356; PMCID: 41457.

43. Qian BZ, Li J, Zhang H, Kitamura T, Zhang J, Campion LR, Kaiser EA, Snyder LA, Pollard JW. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature. 2011;475(7355):222-5. Epub 2011/06/10. doi: 10.1038/naturel0138. PubMed PMID: 21654748; PMCID: 3208506.

44. Hoeffel G, Chen J, Lavin Y, Low D, Almeida FF, See P, Beaudin AE, Lum J, Low I, Forsberg EC, Poidinger M, Zolezzi F, Larbi A, Ng LG, Chan JK, Greter M, Becher B, Samokhvalov IM, Merad M, Ginhoux F. C-Myb(+) erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity. 2015;42(4):665-78. Epub 2015/04/23. doi: 10.1016/j .immuni.2015.03.011. PubMed PMID: 25902481.

45. Del Rio-Hortega P. El "tercer elemento" de los centros nerviosus. I. La microglia en estado normal. II. Intervencion de la microglia en los procesos patologicos (Celulas en bastoncito y cuerpos granuloadiposos). III. Naturaleza probable de la microglia. Bol Soc Espan Biol. 1919;9:68-120.

46. Kupffer C. Ueber Sternzellen der Leber. Arch Fur Mikrosk Anat. 1876; 12:353-8.

47. Langerhans P. Ueber die Nerven der menschlichen Haut. Arch Fiir Pathol Anat Physiol Fur Klin Med. 1868;44(2-3):325-37.

48. Nimmerjahn A, Kirchhoff F, Helmchen F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005;308(5726): 1314-8. Epub 2005/04/16. doi: 10.1126/science. l l l0647. PubMed PMID: 15831717.

49. Chorro L, Sarde A, Li M, Woollard KJ, Chambon P, Malissen B, Kissenpfennig A, Barbaroux JB, Groves R, Geissmann F. Langerhans cell (LC) proliferation mediates neonatal development, homeostasis, and inflammation-associated expansion of the epidermal LC network. The Journal of experimental medicine. 2009;206(13):3089-100. Epub 2009/12/10. doi: 10.1084/jem.20091586. PubMed PMID: 19995948; PMCID: 2806478.

50. Stamatiades EG, Tremblay ME, Bohm M, Crozet L, Bisht K, Kao D, Coelho C, Fan X, Yewdell WT, Davidson A, Heeger PS, Diebold S, Nimmerjahn F, Geissmann F. Immune Monitoring of Trans-endothelial Transport by Kidney -Resident Macrophages. Cell. 2016;166(4):991-1003. doi: 10.1016/j .cell.2016.06.058. PubMed PMID: 27477514; PMCID: PMC4983224.

51. Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, Littman DR, Dustin ML, Gan WB. ATP mediates rapid microglial response to local brain injury in vivo. Nature neuroscience. 2005;8(6):752-8. Epub 2005/05/17. doi: 10.1038/nnl472. PubMed PMID: 15895084.

52. Nishibu A, Ward BR, Jester JV, Ploegh HL, Boes M, Takashima A. Behavioral responses of epidermal Langerhans cells in situ to local pathological stimuli. The Journal of investigative dermatology. 2006; 126(4):787-96. doi: 10.1038/sj Jid.5700107. PubMed PMID: 16439974.

53. Terpstra V, van Berkel TJ. Scavenger receptors on liver Kupffer cells mediate the in vivo uptake of oxidatively damaged red blood cells in mice. Blood. 2000;95(6):2157-63. PubMed PMID: 10706889.

54. Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, Giustetto M, Ferreira TA, Guiducci E, Dumas L, Ragozzino D, Gross CT. Synaptic pruning by microglia is necessary for normal brain development. Science. 2011;333(6048): 1456-8. Epub 2011/07/23. doi: 10.1126/science.1202529. PubMed PMID: 21778362.

55. Li MO, Sarkisian MR, Mehal WZ, Rakic P, Flavell RA. Phosphatidylserine receptor is required for clearance of apoptotic cells. Science. 2003;302(5650): 1560-3. Epub 2003/12/04. doi: 10.1126/science.1087621. PubMed PMID: 14645847. 56. Munoz-Espin D, Canamero M, Maraver A, Gomez-Lopez G, Contreras J, Murillo-Cuesta S, Rodriguez-Baeza A, Varela-Nieto I, Ruberte J, Collado M, Serrano M. Programmed cell senescence during mammalian embryonic development. Cell. 2013; 155(5): 1104-18. doi: 10.1016/j .cell.2013.10.019. PubMed PMID: 24238962.

57. Fantin A, Vieira JM, Gestri G, Denti L, Schwarz Q, Prykhozhij S, Peri F, Wilson SW, Ruhrberg C. Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF -mediated endothelial tip cell induction. Blood. 2010; 116(5):829-40. doi: Doi 10.1182/Blood-2009-12-257832. PubMed PMID: ISL000280596500025.

58. Ingman WV, Wyckoff J, Gouon-Evans V, Condeelis J, Pollard JW. Macrophages promote collagen fibrillogenesis around terminal end buds of the developing mammary gland. Developmental dynamics : an official publication of the American Association of Anatomists. 2006;235(12):3222-9. Epub 2006/10/10. doi: 10.1002/dvdy.20972. PubMed PMID: 17029292.

59. DeFalco T, Bhattacharya I, Williams AV, Sams DM, Capel B. Yolk-sac-derived macrophages regulate fetal testis vascularization and morphogenesis. Proceedings of the National Academy of Sciences of the United States of America. 2014; l l l(23):E2384-93. Epub 2014/06/10. doi: 10.1073/pnas. l400057111. PubMed PMID: 24912173; PMCID: 4060703.

60. Banaei-Bouchareb L, Gouon-Evans V, Samara-Boustani D, Castellotti MC, Czernichow P, Pollard JW, Polak M. Insulin cell mass is altered in Csflop/Csflop macrophage-deficient mice. Journal of leukocyte biology. 2004;76(2):359-67. Epub 2004/06/05. doi: 10.1189/jlb.1103591. PubMed PMID: 15178709.

61. Kubota Y, Takubo K, Shimizu T, Ohno H, Kishi K, Shibuya M, Saya H, Suda T. M-CSF inhibition selectively targets pathological angiogenesis and lymphangiogenesis. Journal of Experimental Medicine. 2009;206(5): 1089-102. doi: Doi 10.1084/Jem.20081605. PubMed PMID: ISL000266010000015.

62. Gordon EJ, Rao S, Pollard JW, Nutt SL, Lang RA, Harvey NL. Macrophages define dermal lymphatic vessel calibre during development by regulating lymphatic endothelial cell proliferation. Development. 2010; 137(22):3899-910. doi: Doi 10.1242/Dev.050021. PubMed PMID: ISL000283671100018.

63. Rymo SF, Gerhardt H, Sand FW, Lang R, Uv A, Betsholtz C. A Two-Way Communication between Microglial Cells and Angiogenic Sprouts Regulates Angiogenesis in Aortic Ring Cultures. PloS one. 2011;6(1). doi: ARTN el5846

DOI 10.1371/journal.pone.0015846. PubMed PMID: ISL000286513100013.

64. Yoshida H, Kawane K, Koike M, Mori Y, Uchiyama Y, Nagata S. Phosphatidyl serine- dependent engulfment by macrophages of nuclei from erythroid precursor cells. Nature. 2005;437(7059):754-8. Epub 2005/09/30. doi: 10.1038/nature03964. PubMed PMID: 16193055. 65. Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013;496(7446):445-55. Epub 2013/04/27. doi: 10.1038/naturel2034. PubMed PMID: 23619691; PMCID: 3725458.

66. Kierdorf K, Prinz M, Geissmann F, Gomez Perdiguero E. Development and function of tissue resident macrophages in mice. Seminars in immunology. 2015;27(6):369-78. doi: 10.1016/j .smim.2016.03.017. PubMed PMID: 27036090.

67. Ajami B, Bennett JL, Krieger C, Tetzlaff W, Rossi FM. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat Neurosci. 2007; 10(12): 1538- 43. Epub 2007/11/21. doi: 10.1038/nn2014. PubMed PMID: 18026097.

68. Ajami B, Bennett JL, Krieger C, McNagny KM, Rossi FM. Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nature neuroscience. 2011;14(9): 1142-9. Epub 2011/08/02. doi: 10.1038/nn.2887. PubMed PMID: 21804537.

69. Evans TA, Barkauskas DS, Myers JT, Hare EG, You JQ, Ransohoff RM, Huang AY, Silver J. High-resolution intravital imaging reveals that blood-derived macrophages but not resident microglia facilitate secondary axonal dieback in traumatic spinal cord injury. Exp Neurol. 2014;254: 109-20. doi: 10.1016/j.expneurol.2014.01.013. PubMed PMID: 24468477; PMCID: PMC3954731.

70. Jenkins SJ, Ruckerl D, Cook PC, Jones LH, Finkelman FD, van Rooijen N, MacDonald AS, Allen JE. Local macrophage proliferation, rather than recruitment from the blood, is a signature of TH2 inflammation. Science. 2011;332(6035): 1284-8. Epub 2011/05/14. doi: 10.1126/science.1204351. PubMed PMID : 21566158 ; PMCID : 3128495.

71. Gomez Perdiguero E, Geissmann F. Myb-Independent Macrophages: A Family of Cells That Develops with Their Tissue of Residence and Is Involved in Its Homeostasis. Cold Spring Harbor symposia on quantitative biology. 2013. Epub 2013/10/15. doi: 10.1101/sqb.2013.78.020032. PubMed PMID: 24122769.

72. Gomez -Nicola D, Fransen NL, Suzzi S, Perry VH. Regulation of microglial proliferation during chronic neurodegeneration. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2013;33(6):2481-93. Epub 2013/02/09. doi: 10.1523/JNEUROSCI.4440-12.2013. PubMed PMID: 23392676.

73. Croker B A, Metcalf D, Robb L, Wei W, Mifsud S, DiRago L, Cluse LA, Sutherland KD, Hartley L, Williams E, Zhang JG, Hilton DJ, Nicola NA, Alexander WS, Roberts AW. SOCS3 is a critical physiological negative regulator of G-CSF signaling and emergency granulopoiesis. Immunity. 2004;20(2): 153-65. PubMed PMID: 14975238.

74. de Boer J, Williams A, Skavdis G, Harker N, Coles M, Tolaini M, Norton T, Williams K,

Roderick K, Potocnik AJ, Kioussis D. Transgenic mice with hematopoietic and lymphoid specific expression of Cre. European journal of immunology. 2003;33(2):314-25. doi: 10.1002/immu.200310005. PubMed PMID: 12548562.

75. Carter RJ, Lione LA, Humby T, Mangiarini L, Mahal A, Bates GP, Dunnett SB, Morton AJ. Characterization of progressive motor deficits in mice transgenic for the human Huntington's disease mutation. J Neurosci. 1999; 19(8):3248-57. PubMed PMID: 10191337.

76. Donadieu J, Rolon MA, Thomas C, Brugieres L, Plantaz D, Emile JF, Frappaz D, David M, Brauner R, Genereau T, Debray D, Cabrol S, Barthez MA, Hoang-Xuan K, Polak M, French LCHSG. Endocrine involvement in pediatric-onset Langerhans' cell histiocytosis: a population- based study. The Journal of pediatrics. 2004; 144(3):344-50. doi: 10.1016/j .jpeds.2003.12.030. PubMed PMID: 15001940.

77. Brooks SP, Dunnett SB. Tests to assess motor phenotype in mice: a user's guide. Nat Rev Neurosci. 2009; 10(7):519-29. doi: 10.1038/nrn2652. PubMed PMID: 19513088.

78. Schaefer A, Sampath SC, Intrator A, Min A, Gertler TS, Surmeier DJ, Tarakhovsky A, Greengard P. Control of cognition and adaptive behavior by the GLP/G9a epigenetic suppressor complex. Neuron. 2009;64(5):678-91. doi: 10.1016/j .neuron.2009.11.019. PubMed PMID: 20005824; PMCID: PMC2814156.

79. Menalled LB, Sison JD, Wu Y, Olivieri M, Li XJ, Li H, Zeitlin S, Chesselet MF. Early motor dysfunction and striosomal distribution of huntingtin microaggregates in Huntington's disease knock-in mice. J Neurosci. 2002;22(18):8266-76. PubMed PMID: 12223581.

80. Ransohoff RM. A polarizing question: do Ml and M2 microglia exist? Nat Neurosci. 2016;19(8):987-91. doi: 10.1038/nn.4338. PubMed PMID: 27459405.

81. Schmued LC, Albertson C, Slikker W, Jr. Fluoro-Jade: a novel fluorochrome for the sensitive and reliable histochemical localization of neuronal degeneration. Brain research. 1997;751(l):37-46. PubMed PMID: 9098566.

82. Erny D, Hrabe de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, Keren- Shaul H, Mahlakoiv T, Jakobshagen K, Buch T, Schwierzeck V, Utermohlen O, Chun E, Garrett WS, McCoy KD, Diefenbach A, Staeheli P, Stecher B, Amit I, Prinz M. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci. 2015;18(7):965-77. doi: 10.1038/nn.4030. PubMed PMID: 26030851.

83. Mittheisz E, Seidl R, Prayer D, Waldenmair M, Neophytou B, Potschger U, Minkov M, Steiner M, Prosch H, Wnorowski M, Gadner H, Grois N. Central nervous system-related permanent consequences in patients with Langerhans cell histiocytosis. Pediatric blood & cancer. 2007;48(l):50-6. doi: 10.1002/pbc.20760. PubMed PMID: 16470521. 84. Grois N, Barkovich AJ, Rosenau W, Ablin AR. Central nervous system disease associated with Langerhans' cell histiocytosis. Am J Pediatr Hematol Oncol. 1993; 15(2):245-54. PubMed PMID: 8498649.

85. Grois N, Tsunematsu Y, Barkovich AJ, Favara BE. Central nervous system disease in Langerhans cell histiocytosis. Br J Cancer Suppl. 1994;23 :S24-8. PubMed PMID: 8075002; PMCID: PMC2149701.

86. Rouco I, Arostegui J, Canovas A, Gonzalez Del Tanago J, Fernandez I, Zarranz JJ. Neurological manifestations in Erdheim-Chester disease: Two case reports. Neurologia. 2016;31(6):426-8. doi: 10.1016/j .nrl.2014.01.010. PubMed PMID: 24735941.

87. Lachenal F, Cotton F, Desmurs-Clavel H, Haroche J, Taillia H, Magy N, Hamidou M, Salvatierra J, Piette JC, Vital-Durand D, Rousset H. Neurological manifestations and neuroradiological presentation of Erdheim-Chester disease: report of 6 cases and systematic review of the literature. J Neurol. 2006;253(10): 1267-77. doi: 10.1007/s00415-006-0160-9. PubMed PMID: 17063320.

88. Wright RA, Hermann RC, Parisi JE. Neurological manifestations of Erdheim-Chester disease. J Neurol Neurosurg Psychiatry. 1999;66(l):72-5. PubMed PMID: 9886456; PMCID: PMC1736172.

89. Lai CF, Chaudhary L, Fausto A, Halstead LR, Ory DS, Avioli LV, Cheng SL. Erk is essential for growth, differentiation, integrin expression, and cell function in human osteoblastic cells. The Journal of biological chemistry. 2001;276(17): 14443-50. doi: 10.1074/jbc.M010021200. PubMed PMID: 11278600.

90. Goke J, Chan YS, Yan J, Vingron M, Ng HH. Genome-wide kinase-chromatin interactions reveal the regulatory network of ERK signaling in human embryonic stem cells. Molecular cell. 2013;50(6):844-55. doi: 10.1016/j .molcel.2013.04.030. PubMed PMID: 23727019.

91. Zhang W, Liu HT. MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell research. 2002; 12(1):9-18. doi: 10.1038/sj .cr.7290105. PubMed PMID: 11942415.

92. Yamamoto T, Ebisuya M, Ashida F, Okamoto K, Yonehara S, Nishida E. Continuous ERK activation downregulates antiproliferative genes throughout Gl phase to allow cell-cycle progression. Current biology : CB. 2006;16(12): 1171-82. doi: 10.1016/j .cub.2006.04.044. PubMed PMID: 16782007.

93. Hatemi I, Bay sal B, Senturk H, Behzatoglu K, Bozkurt ER, Ozbay G. Adult Langerhans cell histiocytosis and sclerosing cholangitis: a case report and review of the literature. Hepatology international. 2010;4(3):653-8. doi: 10.1007/sl2072-010-9205-3. PubMed PMID: 21063491; PMCID: PMC2940001.

94. Fickert P, Stoger U, Fuchsbichler A, Moustafa T, Marschall HU, Weiglein AH, Tsybrovskyy O, Jaeschke H, Zatloukal K, Denk H, Trauner M. A new xenobiotic-induced mouse model of sclerosing cholangitis and biliary fibrosis. Am J Pathol. 2007; 171(2):525-36. doi: 10.2353/ajpath.2007.061133. PubMed PMID: 17600122; PMCID: PMC1934539.

95. Fickert P, Fuchsbichler A, Marschall HU, Wagner M, Zollner G, Krause R, Zatloukal K, Jaeschke H, Denk H, Trauner M. Lithocholic acid feeding induces segmental bile duct obstruction and destructive cholangitis in mice. Am J Pathol. 2006; 168(2):410-22. doi: 10.2353/ajpath.2006.050404. PubMed PMID: 16436656; PMCID: PMC1606500.

96. Constandinou C, Henderson N, Iredale JP. Modeling liver fibrosis in rodents. Methods Mol Med. 2005; 117:237-50. doi: 10.1385/1-59259-940-0:237. PubMed PMID: 16118456.

97. Levine JH, Simonds EF, Bendall SC, Davis KL, Amir el AD, Tadmor MD, Litvin O, Fienberg HG, Jager A, Zunder ER, Finck R, Gedman AL, Radtke I, Downing JR, Pe'er D, Nolan GP. Data-Driven Phenotypic Dissection of AML Reveals Progenitor-like Cells that Correlate with Prognosis. Cell. 2015; 162(l): 184-97. doi: 10.1016/j .cell.2015.05.047. PubMed PMID: 26095251; PMCID: PMC4508757.

98. Tsai J, Lee JT, Wang W, Zhang J, Cho H, Mamo S, Bremer R, Gillette S, Kong J, Haass NK, Sproesser K, Li L, Smalley KS, Fong D, Zhu YL, Marimuthu A, Nguyen H, Lam B, Liu J, Cheung I, Rice J, Suzuki Y, Luu C, Settachatgul C, Shellooe R, Cantwell J, Kim SH, Schlessinger J, Zhang KY, West BL, Powell B, Habets G, Zhang C, Ibrahim PN, Hirth P, Artis DR, Herlyn M, Bollag G. Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity. Proceedings of the National Academy of Sciences of the United States of America. 2008; 105(8):3041-6. doi: 10.1073/pnas.0711741105. PubMed PMID: 18287029; PMCID: PMC2268581.

99. Lito P, Pratilas CA, Joseph EW, Tadi M, Halilovic E, Zubrowski M, Huang A, Wong WL, Callahan MK, Merghoub T, Wolchok JD, de Stanchina E, Chandarlapaty S, Poulikakos PI, Fagin JA, Rosen N. Relief of profound feedback inhibition of mitogenic signaling by RAF inhibitors attenuates their activity in BRAFV600E melanomas. Cancer Cell. 2012;22(5):668-82. doi: 10.1016/j .ccr.2012.10.009. PubMed PMID: 23153539; PMCID: PMC3713778.

100. Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature. 2010;464(7287):427-30. doi: 10.1038/nature08902. PubMed PMID: 20179705; PMCID: PMC3178447. 101. Haroche J, Cohen-Aubart F, Emile JF, Donadieu J, Amoura Z. Vemurafenib as first line therapy in BRAF-mutated Langerhans cell histiocytosis. Journal of the American Academy of Dermatology. 2015;73(l):e29-30. doi: 10.1016/j .jaad.2014.10.045. PubMed PMID: 26089069.

102. Haroche J, Cohen-Aubart F, Emile JF, Maksud P, Drier A, Toledano D, Barete S, Charlotte F, Cluzel P, Donadieu J, Benameur N, Grenier PA, Besnard S, Ory JP, Lifermann F, Idbaih A, Granel B, Graffin B, Hervier B, Arnaud L, Amoura Z. Reproducible and sustained efficacy of targeted therapy with vemurafenib in patients with BRAF(V600E)-mutated Erdheim- Chester disease. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015;33(5):411-8. doi: 10.1200/JCO.2014.57.1950. PubMed PMID: 25422482.

103. Tzoulis C, Schwarzlmuller T, Gjerde IO, Softeland E, Neckelmann G, Biermann M, Haroche J, Straume O, Vintermyr OK. Excellent response of intramedullary Erdheim-Chester disease to vemurafenib: a case report. BMC Res Notes. 2015;8: 171. doi: 10.1186/sl3104-015- 1135-7. PubMed PMID: 25926131; PMCID: PMC4450489.

104. Vanden Borre P, Gunda V, McFadden DG, Sadow PM, Varmeh S, Bernasconi M, Parangi S. Combined BRAF(V600E)- and SRC-inhibition induces apoptosis, evokes an immune response and reduces tumor growth in an immunocompetent orthotopic mouse model of anaplastic thyroid cancer. Oncotarget. 2014;5(12):3996-4010. doi: 10.18632/oncotarget.2130. PubMed PMID: 24994118; PMCID: PMC4147301.

105. Chakravarty D, Santos E, Ryder M, Knauf JA, Liao XH, West BL, Bollag G, Kolesnick R, Thin TH, Rosen N, Zanzonico P, Larson SM, Refetoff S, Ghossein R, Fagin JA. Small- molecule MAPK inhibitors restore radioiodine incorporation in mouse thyroid cancers with conditional BRAF activation. The Journal of clinical investigation. 2011; 121(12):4700-11. doi: 10.1172/JCI46382. PubMed PMID: 22105174; PMCID: PMC3225989.

106. Rochet NM, Kottschade LA, Markovic SN. Vemurafenib for melanoma metastases to the brain. The New England journal of medicine. 2011;365(25):2439-41. doi: 10.1056/NEJMcl l l l672. PubMed PMID: 22188003.

107. Larkin J, Ascierto PA, Dreno B, Atkinson V, Liszkay G, Maio M, Mandala M, Demidov L, Stroyakovskiy D, Thomas L, de la Cruz-Merino L, Dutriaux C, Garbe C, Sovak MA, Chang I, Choong N, Hack SP, McArthur GA, Ribas A. Combined vemurafenib and cobimetinib in BRAF- mutated melanoma. The New England journal of medicine. 2014;371(20): 1867-76. doi: 10.1056/NEJMoal408868. PubMed PMID: 25265494.

108. Allen CE, Li L, Peters TL, Leung HC, Yu A, Man TK, Gurusiddappa S, Phillips MT, Hicks MJ, Gaikwad A, Merad M, McClain KL. Cell-specific gene expression in Langerhans cell histiocytosis lesions reveals a distinct profile compared with epidermal Langerhans cells. Journal of immunology. 2010; 184(8):4557-67. doi: 10.4049/jimmunol.0902336. PubMed PMID: 20220088; PMCID: PMC3142675.

109. Egeler RM, Favara BE, van Meurs M, Laman JD, Claassen E. Differential In situ cytokine profiles of Langerhans-like cells and T cells in Langerhans cell histiocytosis: abundant expression of cytokines relevant to disease and treatment. Blood. 1999;94(12):4195-201. PubMed PMID: 10590064.

110. Arnaud L, Gorochov G, Charlotte F, Lvovschi V, Parizot C, Larsen M, Ghillani-Dalbin P, Hervier B, Kahn JE, Deback C, Musset L, Amoura Z, Haroche J. Systemic perturbation of cytokine and chemokine networks in Erdheim-Chester disease: a single-center series of 37 patients. Blood. 2011; 117(10):2783-90. doi: 10.1182/blood-2010-10-313510. PubMed PMID: 21205927.

111. Sevcikova S, Kubiczkova L, Sedlarikova L, Rihova L, Kryukov F, Szturz P, Hajek R, Pour L, Adam Z. Impact of anakinra treatment on cytokine and lymphocytes/ monocytes profile of an Erdheim-Chester patient. Klin Onkol. 2014;27(4):276-82. PubMed PMID: 25115717.

EQUIVALENTS

[0144] The present technology is not to be limited in terms of the particular embodiments described in this application, which are intended as single illustrations of individual aspects of the present technology. Many modifications and variations of this present technology can be made without departing from its spirit and scope, as will be apparent to those skilled in the art. Functionally equivalent methods and apparatuses within the scope of the present technology, in addition to those enumerated herein, will be apparent to those skilled in the art from the foregoing descriptions. Such modifications and variations are intended to fall within the scope of the appended claims. The present technology is to be limited only by the terms of the appended claims, along with the full scope of equivalents to which such claims are entitled. It is to be understood that this present technology is not limited to particular methods, reagents, compounds compositions or biological systems, which can, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting.