This document relates to methods and materials for treating obesity-induced neuropsychiatric disorders. For example, one or more senotherapeutic agents can be administered to a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) to treat the mammal.
Obesity can be associated with a range of neurodegenerative and psychiatric disorders, including anxiety and depression in some cases (Gariepy et al., Int J Obes (Lond), 34:407-419 (2010); Hryhorczuk et al., Front Neurosci, 7:177 (2013); Stunkard and Wadden, Am J Clin Nutr, 55:524S-532S (1992)). Anxiety is a behavioral trait in some obese patients (Gariepy et al., Int J Obes (Lond), 34:407-419 (2010)), affecting 40% more obese patients and non-obese patients. Increased anxiety-like behavior also was reported in rodents genetically predisposed to develop obesity, e.g., db/db mice (Dinel et al., PLoS One 6:e24325 (2011)) and in high fat (HF) diet-induced obesity (Heyward et al., Neurobiol Learn Mem 98:25-32 (2012); and Mizunoya et al., Springerplus 2:165 (2013)). Processes such as inflammation (Capuron and Miller, Pharmacol Ther 130:226-238 (2011); and Lasselin and Capuron, 2014), altered hormone signaling (Ulrich-Lai and Ryan, Cell Metab 19:910-925 (2014)), and stem cell dysfunction (Anacker and Hen, Nat Rev Neurosci 18:335-346 (2017); and Gao et al., Neurochem Int 106:24-36 (2017)) have been speculated to underlie obesity-related anxiety, but the underlying mechanisms have not been identified.
This document provides methods and materials related to treating obesity-induced neuropsychiatric disorders. For example, this document provides methods and materials for using one or more senotherapeutic agents to treat a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety). In some cases, a mammal having, or at risk of developing, obesity-induced anxiety can be treated with a composition including one or more senotherapeutic agents (e.g., dasatinib and/or quercetin) to reduce or eliminate one or more symptoms of obesity-induced anxiety (e.g., anxiety-like behavior). In some cases, a mammal having, or at risk of developing, obesity-induced anxiety can be treated with a composition including one or more senotherapeutic agents to restore neurogenesis within the mammal.
As demonstrated herein, obesity can result in the accumulation of senescent glial cells in proximity to the lateral ventricle (LV), a region in which adult neurogenesis occurs, and these senescent glial cells can exhibit an accumulation of lipids in senescence (ALISE; e.g., excessive fat accumulation). Also as demonstrated herein, reducing the level of cells with an ALISE phenotype from obese mammals (e.g., high fat-fed and leptin receptor-deficient (db/db) obese mice) can restore neurogenesis and alleviated anxiety-related behavior. The ability to decrease the number of senescent glial cells in the LV of an obese mammal (e.g., by administering one or more senotherapeutic agents to the mammal) can be used to treat the mammal having an obesity-induced neuropsychiatric disorder such as anxiety and depression.
In general, one aspect of this document features methods for treating an obesity-induced neuropsychiatric disorder. The methods can include, or consist essentially of, administering a composition including a senolytic agent to a mammal identified as having an obesity-induced neuropsychiatric disorder. The mammal can be a human. The obesity-induced neuropsychiatric disorder can be obesity-induced anxiety. The obesity-induced neuropsychiatric disorder can be obesity-induced depression. The composition can be effective to clear senescent cells from within the brain of the mammal. The senescent cells can include an ALISE phenotype. The senescent cells can be cleared from in proximity to the lateral ventricle of the brain of the mammal. The composition can be effective to decrease a level of one or more senescence-associated secretory phenotype (SASP) factor polypeptides in the mammal.
In another aspect, this document features methods for increasing neurogenesis. The methods can include, or consist essentially of, administering a composition including a senolytic agent to a mammal identified as having an obesity-induced neuropsychiatric disorder under conditions wherein neurogenesis within the mammal is increased. The mammal can be a human. The obesity-induced neuropsychiatric disorder can be obesity-induced anxiety. The obesity-induced neuropsychiatric disorder can be obesity-induced depression. The neurogenesis can be increased in the brain of the mammal. For example, the neurogenesis can be increased in the subventricular zone of the brain of the mammal. For example, the neurogenesis can be increased in the olfactory bulbs of the mammal. The composition can be effective to decrease a level of one or more SASP factor polypeptides in the mammal.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. Although methods and materials similar or equivalent to those described herein can be used to practice the invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.
This document provides methods and materials related to treating obesity-induced neuropsychiatric disorders. For example, this document provides methods and materials for using one or more senotherapeutic agents (e.g., dasatinib and/or quercetin) to treat a mammal having an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety). In some cases, one or more senotherapeutic agents (e.g., dasatinib and/or quercetin) can be used as described herein to treat a mammal at risk of developing an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety).
In some cases, a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder can be treated with a composition including one or more senotherapeutic agents (e.g., dasatinib and/or quercetin) to alleviate (e.g., to reduce or eliminate) one or more (e.g., one, two, three, four, five, or more) symptoms of the obesity-induced neuropsychiatric disorder. An obesity-induced neuropsychiatric disorder can be any type of obesity-induced neuropsychiatric disorder. Examples of obesity-induced neuropsychiatric disorder include, without limitation, obesity-induced anxiety, obesity-induced depression, obesity-induced fearfulness, obesity-related suicide, and obesity-induced stress. A symptom of an obesity-induced neuropsychiatric disorder can be any appropriate symptom. For example, examples of symptoms of obesity-induced anxiety include, without limitation, anxiety-related behaviors such as feeling nervous, feeling restless, feeling tense, feeling stressed, having a sense of impending danger, increased heart rate, hyperventilation, sweating, trembling, feeling weak or tired, trouble concentrating or thinking about anything other than the present worry, having trouble sleeping, and gastrointestinal problems. Each of these symptoms of obesity-induced anxiety can be identified, staged, and/or monitored using clinical techniques as described elsewhere (see, e.g., Practice Guidelines for Psychiatric Evaluation of Adults, Third Edition, American psychiatric association, 2016; Lykouras et al., Psychiatriki 22:307-13 (2011); and Locke et al., Am Fam Physician 91:617-24 (2015)). For example, examples of symptoms of obesity-induced depression include, without limitation, feelings of sadness, feelings of tearfulness, feelings of hopelessness, feelings of worthlessness, angry outbursts, irritability or frustration, loss of interest or pleasure normal activities hobbies or sports, sleep disturbances, lack of energy, fixating on past failures or self-blame, frequent or recurrent thoughts of death, suicidal thoughts, suicide attempts, and unexplained physical problems such as back pain or headaches. Each of these symptoms of obesity-induced depression can be identified, staged, and/or monitored using clinical techniques as described elsewhere (see, e.g., Practice Guidelines for Psychiatric Evaluation of Adults, Third Edition, American psychiatric association, 2016; and Clinical Practice Guidelines, American Psychiatric Association, available at psychiatry.org/psychiatrists/practice/clinical-practice-guidelines) In some cases, administering one or more senotherapeutic agents to a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder can be effective to alleviate one or more anxiety-related behaviors in the mammal.
In some cases, a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) can be treated with a composition including one or more senotherapeutic agents (e.g., dasatinib and/or quercetin) to clear one or more senescent cells from within the mammal. A senescent cell can be any type of cell. In some cases, a senescent cell can exhibit excessive fat accumulation (e.g., can have an ALISE phenotype). Examples of senescent cells that can be cleared as described herein include, without limitation, a senescent glial cell, an ependymal cell, a neural progenitor cell, a neuron, and an endothelial cell. A senescent cell can be cleared from any location within the mammal. In some cases, a senescent cell can be cleared from the brain of a mammal. Examples of locations from which a senescent cell cleared include, without limitation, in proximity to the LV of the brain of the mammal, in the LV of the brain of the mammal, in proximity to the subventricular zone (SVZ) of the brain of the mammal, in the SVZ of the brain of the mammal, and cerebral blood vessels. A location in proximity to the LV of the brain of a human can be the region within about 10 mm (within about 9 mm, within about 8 mm, within about 7 mm, within about 6 mm, within about 5 mm, within about 4 mm, within about 3 mm, within about 2 mm, or within about 1 mm) of the LV. A location in proximity to the SVZ of the brain of a human can be the region within about 10 mm (within about 9 mm, within about 8 mm, within about 7 mm, within about 6 mm, within about 5 mm, within about 4 mm, within about 3 mm, within about 2 mm, or within about 1 mm) of the SVZ. In some cases, administering one or more senotherapeutic agents to a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder can be effective to clear one or more senescent cells having an ALISE phenotype from a location in proximity to the LV of the brain of the mammal.
In some cases, a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) can be treated with a composition including one or more senotherapeutic agents (e.g., dasatinib and/or quercetin) to increase (e.g., restore) neurogenesis within the mammal. In some cases, a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) can be treated with a composition including one or more senotherapeutic agents (e.g., dasatinib and/or quercetin) to alleviate (e.g., to reduce or eliminate) obesity-related impairment of neurogenesis in the mammal. Neurogenesis of any appropriate type of cell can be increased. Examples of cells for which neurogenesis can be increased as described herein include, without limitation, neuronal precursor cells, immature neurons, ependymal cells, and developing neurons. Neurogenesis can be increased in any location within the mammal. Examples of locations in which a neurogenesis can be increased include, without limitation, in the SVZ of the brain of the mammal, and in the olfactory bulbs of the mammal. In some cases, administering one or more senotherapeutic agents to a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder can be effective to restore neurogenesis in the mammal.
In some cases, a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) can be treated with a composition including one or more senotherapeutic agents (e.g., dasatinib and/or quercetin) to alleviate (e.g., to reduce or eliminate) inflammation in the mammal. A level (e.g., a systemic level) of any appropriate inflammatory factor (e.g., cytokines, chemokines, and matrix proteases) can be altered (e.g., increased or decreased) to alleviate inflammation in a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder. In cases where an inflammatory factor is a pro-inflammatory factor (e.g., SASP factor polypeptides such as G-Csf, Il-1α and Il-1β, Kc/Cxcl1, Mcp-1, Mig, Il-6, Tnf-α; and IL-8) the pro-inflammatory factor can be decreased. In cases where an inflammatory factor is an anti-inflammatory factor, the anti-inflammatory factor can be increased. Inflammation at any appropriate location within the mammal can be alleviated. Examples of locations from which inflammation can be alleviated as described herein include, without limitation, the brain, blood vessels, adipose tissue, the lungs, kidneys, the liver, bone, bone marrow, and skin. In some cases, a systemic inflammatory factor (e.g., systemic SASP factor polypeptides) can cross the blood-brain barrier to alleviate brain inflammation. In some cases, administering one or more senotherapeutic agents to a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder can be effective to alleviate brain inflammation within the mammal.
In some cases, when a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) is treated with a composition including one or more senotherapeutic agents (e.g., dasatinib and/or quercetin), the mammal's body weight is not affected (e.g., is not altered).
In some cases, when a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) is treated with a composition including one or more senotherapeutic agents (e.g., dasatinib and/or quercetin), the mammal's body composition is not affected (e.g., is not altered).
In some cases, when a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) is treated with a composition including one or more senotherapeutic agents (e.g., dasatinib and/or quercetin), the mammal's activity is not affected (e.g., is not altered).
When treating a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) as described herein (e.g., by administering one or more senotherapeutic agents such as dasatinib and/or quercetin), the mammal can be any appropriate mammal. In some cases, a mammal can be an obese mammal (e.g., a mammal that is overweight). Examples of mammals that can be treated using a composition containing one or more senotherapeutic agents as described herein include, without limitation, humans, non-human primates such as monkeys, dogs, cats, horses, cows, pigs, sheep, mice, and rats. In some cases, a composition containing one or more senotherapeutic agents can be administered to a human having an obesity-induced neuropsychiatric disorder to treat the human. In some cases, a composition containing one or more senotherapeutic agents can be administered to a human at risk of developing an obesity-induced neuropsychiatric disorder to slow the onset or progression of an obesity-induced neuropsychiatric disorder within the human.
In some cases, the methods described herein also can include identifying a mammal as having, or as being at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety). Examples of methods for identifying a mammal as having, or as being at risk of developing, an obesity-induced neuropsychiatric disorder include, without limitation, psychological evaluation, physical examination, and/or laboratory tests such as stress hormone levels. Once identified as having, or as being at risk of developing, an obesity-induced neuropsychiatric disorder, a mammal can be administered or instructed to self-administer one or more senotherapeutic agents (e.g., dasatinib and/or quercetin).
A composition containing one or more (e.g., one, two, three, four, five, or more) senotherapeutic agents can include any appropriate senotherapeutic agent(s). A senotherapeutic agent can be any type of molecule (e.g., small molecules or polypeptides). In some cases, a senotherapeutic agent can be a senolytic agent (i.e., an agent having the ability to induce cell death in senescent cells). In some cases, a senotherapeutic agent can be a senomorphic agent (i.e., an agent having the ability to suppress senescent phenotypes without cell killing). Examples of senotherapeutic agents that can be used as described herein (e.g., to treat a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder such as obesity-induced anxiety) can include, without limitation, dasatinib, quercetin, navitoclax, A1331852, A1155463, fisetin, luteolin, geldanamycin, tanespimycin, alvespimycin, piperlongumine, panobinostat, FOX04-related peptides, nutlin3a, ruxolitinib, metformin, and rapamycin.
In some cases, a composition containing one or more (e.g., one, two, three, four, five, or more) senotherapeutic agents (e.g., dasatinib and/or quercetin) can include the one or more senotherapeutic agent(s) as the sole active ingredient(s) in the composition that is effective to treat an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety). In some cases, a composition containing one senotherapeutic agent (e.g., fisetin) can include that one senotherapeutic agent as the sole active ingredient in the composition that is effective to treat an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety).
In some cases, a composition containing one or more (e.g., one, two, three, four, five, or more) senotherapeutic agents (e.g., dasatinib and/or quercetin) can include one or more (e.g., one, two, three, four, five, or more) additional active agents (e.g., therapeutic agents) in the composition that are effective to treat an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety).
In some cases, a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) being treated as described herein (e.g., by administering one or more senotherapeutic agents such as dasatinib and/or quercetin) also can be treated with one or more (e.g., one, two, three, four, five, or more) additional therapeutic agents. A therapeutic agent used in combination with one or more senotherapeutic agents described herein can be any appropriate therapeutic agent. Examples of therapeutic agents that can be used in combination with one or more senotherapeutic agents described herein include, without limitation, benzodiazepines (e.g., alprazolams such as XANAX™, chlordiazepoxides such as LIBRIUIM®, clonazepams such as KLONOPIN®, diazepams such as VALIUM®, and lorazepams such as ATIVAN®), buspirone, and antidepressants including selective serotonin reuptake inhibitors (SSRIs; e.g., escitaloprams such as LEXAPRO, fluoxetines such as PROZAC®, paroxetines such as PAXIL®, and sertralines such as ZOLOFT®). In some cases, the one or more additional therapeutic agents can be administered together with the one or more senotherapeutic agents (e.g., in a composition containing one or more senotherapeutic agents and containing one or more additional therapeutic agents). In some cases, the one or more (e.g., one, two, three, four, five, or more) additional therapeutic agents can be administered independent of the one or more senotherapeutic agents. When the one or more additional therapeutic agents are administered independent of the one or more senotherapeutic agents, the one or more senotherapeutic agents can be administered first, and the one or more additional therapeutic agents administered second, or vice versa.
In some cases, a composition containing one or more senotherapeutic agents (e.g., dasatinib and/or quercetin) can be formulated into a pharmaceutically acceptable composition for administration to a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety). For example, one or more senotherapeutic agents can be formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents. Pharmaceutically acceptable carriers, fillers, and vehicles that can be used in a pharmaceutical composition described herein include, without limitation, saline, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol (PEG; e.g., PEG400), sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, and wool fat.
In some cases, when a composition containing one or more senotherapeutic agents (e.g., dasatinib and/or quercetin) is administered to a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety), the composition can be designed for oral or parenteral (including subcutaneous, intramuscular, intravenous, and intradermal) administration to the mammal. Compositions suitable for oral administration include, without limitation, liquids, tablets, capsules, pills, powders, gels, and granules. Compositions suitable for parenteral administration include, without limitation, aqueous and non-aqueous sterile injection solutions that can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient.
A composition containing one or more senotherapeutic agents (e.g., dasatinib and/or quercetin) can be administered to a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) in any appropriate amount (e.g., dose). Effective amounts can vary depending on the route of administration, the age and general health condition of the subject, excipient usage, the possibility of co-usage with other therapeutic treatments such as use of other agents, and the judgment of the treating physician. An effective amount of a composition containing one or more senotherapeutic agents can be any amount that can treat a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder without producing significant toxicity to the mammal. For example, an effective amount of dasatinib (D) can be from about 1 milligram per kilogram body weight (mg/kg) to about 20 mg/kg (e.g., about 5 mg/kg). For example, an effective amount of quercetin (Q) can be from about 10 mg/kg to about 200 mg/kg (e.g., about 50 mg/kg). The effective amount can remain constant or can be adjusted as a sliding scale or variable dose depending on the mammal's response to treatment. Various factors can influence the actual effective amount used for a particular application. For example, the frequency of administration, duration of treatment, use of multiple treatment agents, route of administration, and severity of the obesity-induced neuropsychiatric disorder in the mammal being treated may require an increase or decrease in the actual effective amount of senotherapeutic agent(s) administered.
A composition containing one or more senotherapeutic agents (e.g., dasatinib and/or quercetin) can be administered to a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) in any appropriate frequency. The frequency of administration can be any frequency that can treat a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder without producing significant toxicity to the mammal. For example, the frequency of administration can be from about twice a day to about once every 6 months, from about once a day to about once a week, or from about once a week to about once every 6 months. In some cases, a composition containing one or more senotherapeutic agents can be administered once a day. The frequency of administration can remain constant or can be variable during the duration of treatment. As with the effective amount, various factors can influence the actual frequency of administration used for a particular application. For example, the effective amount, duration of treatment, use of multiple treatment agents, and route of administration may require an increase or decrease in administration frequency.
A composition containing one or more senotherapeutic agents (e.g., dasatinib and/or quercetin) can be administered to a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) for any appropriate duration. An effective duration for administering or using a composition containing one or more senotherapeutic agents can be any duration that can treat a mammal having, or at risk of developing, an obesity-induced neuropsychiatric disorder without producing significant toxicity to the mammal. For example, the effective duration can vary from several days, to several weeks, to several months, or to a lifetime. In some cases, the effective duration can range in duration from about several months to about 10 years. Multiple factors can influence the actual effective duration used for a particular treatment. For example, an effective duration can vary with the frequency of administration, effective amount, use of multiple treatment agents, and route of administration.
In certain instances, a course of treatment can be monitored. In some cases, methods described herein also can include monitoring the severity of an obesity-induced neuropsychiatric disorder (e.g., obesity-induced anxiety) in a mammal. Any appropriate method can be used to monitor the severity of an obesity-induced neuropsychiatric disorder in a mammal. In some cases, methods described herein also can include monitoring a mammal being treated as described herein for toxicity. The level of toxicity, if any, can be determined by assessing a mammal's clinical signs and symptoms before and after administering a known amount of a particular composition. It is noted that the effective amount of a particular composition administered to a mammal can be adjusted according to a desired outcome as well as the mammal's response and level of toxicity.
The invention will be further described in the following examples, which do not limit the scope of the invention described in the claims.
This example shows that during obesity, glial cells show increased markers of cellular senescence in the periventricular region of the lateral ventricle (LV), a region in close proximity to the neurogenic niche. Senescent glial cells in obese mice show excessive fat accumulation, a phenotype termed accumulation of lipids in senescence (ALISE). Importantly, specific clearance of senescent cells alleviates the obesity-related impairment in adult neurogenesis, and decreases obesity-induced anxiety-like behavior. This work suggests that targeting senescent cells can be used as a therapeutic avenue for treating obesity-induced anxiety.
In order to investigate the relationship between obesity and anxiety, eight month old C57Bl/6 mice were fed a high fat (60% of calories from fat) or standard chow diet for 2 months. It was found that body weight and body fat content were increased in high fat diet (HFD) mice in comparison to chow-fed controls (
As an additional measurement of anxiety-like behavior, the elevated plus maze (EPM) test was used. The EPM is based on the animal's natural fear of heights and open spaces. Increased anxiety-like behavior in the EPM test is manifested as a decrease in the number of head-pokes and entries into the open arms. It was found that animals on a HFD had decreased entries into the open arms of the EPM (frequency and time) compared to lean animals (
It was investigated if senescent cells could contribute to anxiety-like behavior during obesity by using the INK-ATTAC mouse model, which allows the induction of suicide gene-mediated ablation of p16Ink4a-expressing senescent cells upon administration of the drug AP20187 (AP) (Baker et al., Nature 479:232-236 (2011); Xu et al., Elife 4:e12997 (2015)).
Chow- and HFD-fed 10 month old mice were repeatedly treated with AP or vehicle (
Next, anxiety-like behavior was assayed with the elevated plus maze (EPM). As previously observed, obese animals avoided entries into the open arms of the EPM (frequency and time) compared to lean animals (
To exclude off-target effects of the drug AP, wild-type C57Bl/6 mice were treated with the drug and tested for anxiety-like behavior. Wild-type mice showed a significant difference between chow and HFD in the OF test before the start of the treatment (
In addition to HFD fed mice, complementary experiments were conducted in db/db mice in which obesity is caused by a point mutation in the leptin receptor gene lepr, leading to spontaneous type 2 diabetes (Wang et al., Current Diabetes Reviews 10:131-145 (2014)). These mice were treated intermittently for two months with the senolytic drug cocktail, Dasatinib and Quercetin (D+Q) (Zhu et al., Aging Cell 14:644-658 (2015)). Db/db mice have significantly increased body weights and adipose depot weights when compared to lean db+/− heterozygous littermates, but interestingly body weight did not change over the course of D+Q treatment (
Similarly to HFD fed mice, db/db mice exhibited increased anxiety-like behavior as assessed by the OF test (
Finally, these results were confirmed in a cohort of double-transgenic, INK-ATTAC;db/db mice. Similarly to treatment with D+Q, genetic clearance of p16Ink4a-positive senescent cells in INK-ATTAC;db/db mice did not alter body weight, body composition, or activity (
These data show that pharmacological or pharmacogenetic clearance of senescent cells in two different models of obesity significantly alleviates anxiety-like behavior.
To investigate the effectiveness of senescent cell-clearance in HFD mice, senescent markers were measured in the perigonadal adipose tissue, a tissue previously shown to exhibit a marked increase in the number senescent cells with age (Schafer et al., Nature Commun. 8:14532 (2017); Tchkonia et al., Aging Cell 9:667-684 (2010); Xu et al., Elife 4:e12997 (2015)).
It was found that the senescence markers SA-β-Gal, p16Ink4a, and telomere-associated DNA damage foci (TAF) were increased in INK-ATTAC mice on HFD and were significantly reduced upon administration of AP (
Given that the senolytic approaches applied act systemically, it is possible that they reduce SASP factors which can penetrate the blood-brain-barrier and therefore impact on the brain. To investigate that, blood plasma was analyzed for a large array of SASP factors.
Evaluation of circulating cytokines in the bloodstream of chow- and HFD-fed INK-ATTAC mice revealed that HFD resulted in the up-regulation of known SASP factors such as G-Csf, Il-1β, Kc/Cxcl1, Mcp-1, Mig, and Tnf-α which were down-regulated upon AP treatment (
Recently, it has been shown that transplantation of relatively low numbers of senescent cells in young animals resulted in physical dysfunction measured by Rotarod performance, grip strength, or endurance when compared to transplantation of young cells (Xu et al., Nature Medicine 24:1246-1256 (2018)). This study showed that transplantation of senescent cells resulted in long-lasting systemic effects in tissues located distantly from where senescent cells were injected.
To test if senescent cells could induce anxiety-like behavior via systemic effects, young or senescent cells were transplanted into lean mice and assessed behavior and physical function 6 and 12 weeks later. It was confirmed the previous observations that transplanted senescent cells reduced physical function, as measured by Rotarod (
It was next examined if obesity could induce senescence specifically in the brain, thereby contributing to anxiety. Markers of senescence were first assessed in these regions of the brains of obese and lean INK-ATTAC mice treated with and without AP. No differences were found in the senescent markers p21, p16, γ-H2A.X, and TAF between any of the experimental groups (
Interestingly, assessment of senescent cells in the amygdala, a brain region associated with emotional responses including anxiety and fear (Adhikari et al., Nature 527:179-185 (2015)), exhibited a significant increase in the number of p16Ink4a-positive cells in HFD-fed mice (
Together, these data indicate that HFD does not induce senescence in regions of the brain implicated in learning, memory, and motor-neuron control such as the cortex, cerebellum, and hippocampus. However, HFD induces senescence in the hypothalamus and amygdala, which may contribute to its effects on anxiety-like behavior and treatment with AP reduced senescent cell abundance and attenuated these behavioral changes.
A connection between senescence and fat accumulating in the brain was investigated.
Analysis of Perilipin 2 (Plin2) expression (a protein which surrounds lipid droplets) in the brain of HF diet mice revealed a significant increase in Plin2+ cells (
In order to investigate if Plin2+ cells show features of senescence, the senescence marker TAF was analyzed in combination with immunostaining against Plin2. Higher mean values and higher frequencies of TAF in Plin2+ cells (
To further investigate the impact of senescent cells on the build-up of fat in the brain, the INK-ATTAC mouse model (Baker et al., Elife. 4:e12997 (2015); and Baker et al., Nature 479:232-236 (2011)) was used. Treatment of HFD INK-ATTAC mice with AP resulted in a significant reduction of Plin2+ cells (
Lastly, anxiety markers in HFD animals, such as distance travelled in the central zone and entries into the central zone, showed a strong negative correlation with the abundance of Plin2+ cells detected in the lateral ventricle (
To further investigate the impact of fat accumulation on cell senescence, mouse adult fibroblasts (MAF) were used and senescence was induced by X-ray irradiation as described elsewhere (see, e.g., Jurk et al., Nat Commun 2 (2014); Ogrodnik et al., Nat Commun. 8:1569 (2017)). Senescent cells were cultured in the presence or absence of external sources of lipids. It was found that in the absence of extracellular lipids, the ALISE phenotype cells (assessed by lipophilic dye, Nile Red) was suppressed (
These data show that excessive lipid accumulation during senescence (ALISE) may be a contributor of genomic instability, resulting in release of chromatin fragments and activation of the SASP.
In HFD mice, it was observed that Plin2+ senescent glial cells are frequently found in close proximity to cells expressing doublecortin (Dcx), a marker of neuronal precursor cells and immature neurons (
Lean and obese INK-ATTAC mice were treated with or without AP as previously described. Following organ harvesting, single-cell suspensions were obtained from one brain hemisphere and analyzed them by Cytometry by Time Of Flight (CyTOF), which allows mapping and discriminating between different brain cells including astrocytes, oligodendrocytes, microglia, neurons, ependymal cells, pericytes, and endothelial cells. The second brain hemisphere was reserved for histological analyses (
It was found that brains of mice fed a HFD did not exhibit significant changes in the frequencies of oligodendrocytes (CNPase+ or OSP+), microglia (CD11b+, CD45), mature neurons (NeuN+), or endothelial cells (CD31+ or CD146+) (
These findings were validated by performing immunostaining for Dcx (
In summary, these data indicate that senescent cells play a causal role in the decreased neurogenesis induced by HFD. Targeting senescent cells in obese mice alleviates obesity-related anxiety-like behavior related to clearance of periventricular fat accumulation and restoration of adult neurogenesis.
Experimental procedures were approved by the Institutional Animal Care and Use Committee at Mayo Clinic (protocol A26415). INK-ATTAC+/− transgenic mice were generated and genotyped as described elsewhere (see, e.g., Baker et al., Nature 479:232-236 (2011)). Briefly, INK-ATTAC mice were produced and phenotyped at Mayo Clinic. Controls for the INK-ATTAC experiments were INK-ATTAC-null C57BL/6 background mice raised in parallel. C57BL/6 db/db and db/− mice were purchased from Jackson Laboratories.
Mice were housed 2-5 mice per cage, at 22+/−0.5° C. on a 12-12 hour day-night cycle and provided with food and water ad libitum. For high fat diet-induced obesity studies, mice were randomly assigned to chow or high fat diet groups. Mice were fed the high fat diet for 2-4 months before experiments started. High fat food was purchased from Research Diets (cat no #D12492). 60% of calories in this high-fat diet are from fat. Standard mouse chow diet was obtained from Lab Diet (cat no #5053).
INK-ATTAC mice were injected intraperitoneally (i.p.) with AP20187 (10 mg/kg) or vehicle for 3 days every 2 weeks for a total of 8-10 weeks.
Senolytic-treated db/db mice were gavaged with Dasatinib (D; 5 mg/kg) and quercetin (Q; 50 mg/kg) or vehicle for 5 days every 2 weeks for 8 weeks.
For off target effect measurements db/db and HDF mice (fed with HFD for 2 months prior treatment) were injected intraperitoneally (i.p.) with AP21087 at 10 mg/kg or vehicle for 3 days every 2 weeks for 8 weeks.
Recombinant CXCL1 (Peprotech, #250-11) or vehicle (PBS) was administered to lean C57BL/6 via i.p. injection (5 μg/kg in PBS) daily for 7 days. 2 hours after the last injection mice were tested in open field and elevated plus maze and afterwards dissected.
Reparixin L-lysine salt (MedChemExpress, #HY-15252) or L-Lysine hydrochloride (MedChemExpress, #HY-N0470) was dissolved in H2O was administered to obese C57BL/6 mice (fed for 2 months with high-fat diet) via subcutaneous injection (30 mg/kg) twice per day for 2 weeks. 2 hours after the last injection mice were tested in open field and elevated plus maze and afterwards dissected.
Tissues from mice sacrificed at the indicated time points were snap-frozen in liquid nitrogen for biochemical studies or fixed in 4% PFA for 24 hours prior to processing and paraffin embedding. Paraffin-embedded tissues were cut at 3 μm or 10 μm intervals.
Wild-type C57BL/6 mice were obtained from the National Institute on Aging (NIA) and maintained in a pathogen-free facility at 23-24° C. under a 12 hours light, 12 hours dark regimen with food and water ad libitum. Cell transplantation was done as previously described (Xu et al., Nature Medicine 24:1246-1256 (2018)). Briefly, when mice were 18 months of age, they were anesthetized using isoflurane and were injected intraperitoneally with 150 μl PBS through a 22-G needle, containing 106 control or senescent mouse preadipocytes cells, or only PBS. Preadipocytes were obtained from inguinal fat from young Luciferase transgenic C57BL/6 mice from The Jackson Laboratory (Bar Harbor, Me.; stock no. 025854). Senescence was induced by 10 Gy of cesium radiation. Open field testing was carried out at 2 and 6 weeks after transplantation and Rotarod performance was tested 2 and 12 weeks after transplantation.
Lean and fat mass of individual mice were determined by quantitative nuclear magnetic resonance using an EchoMRT analyser (Houston, Tex.) and expressed as a function of body weight. Un-anesthetized animals were placed in a plastic tube that was introduced into the EchoMRT instrument. Body composition, comprising fat mass and lean mass, was determined in approximately 90 seconds per animal.
Locomotor activity and anxiety-like behavior of mice were assessed in sound-insulated, rectangular activity chambers (Med Associates Inc., St Albans, Vt., USA: W×L×D=27 cm×27 cm×20 cm with continually running fans, infrared lasers, and sensors). Beam breaks were assessed in 2-minute bins over 30 minutes, converted automatically to current mouse location and distance travelled (cm), and recorded on a computer with Med-PC software Version 4.0. Before the test, mice were acclimatized to the room for 1-1.5 hours before being introduced into the chambers. Mice were habituated for 5 minutes in the Open Field chamber (without recording) then placed for another 5 minutes in the home cage. Afterwards, mice were introduced back to the chambers and all mouse movements were recorded for 30 minutes. Anxiety was quantified by the distance mice travelled in the central 25% of the chamber (zone 1) as a function of the total distance mice travelled and by frequencies of entries into zone 1.
A grey colored elevated plus maze apparatus was used. Two open arms (25×5 cm) and two closed arms (25×5 cm) were attached at right angles to a central platform (5×5 cm). The apparatus was set 40 cm above the floor. Mice were first acclimatized to the room for 1-1.5 hours. Mice were then placed individually on the central platform with their back to one of the open arms. Before the test, mice were habituated for 1 minute to the maze, then placed back in the home cage for 5 minutes. Mice were tested for 5 minutes during which they could freely explore the apparatus. Tracking software (Ethovision) recognizes mouse head, central body point, and the base of the tail. Anxiety was quantified by frequency of and time spent during head pokes/dips toward open arms. Higher anxiety is indicated by a lower frequency of movement into open arms and less time spent there.
Rotarod performance test evaluates mouse balance and motor coordination. Mice were brought to the test room a day before testing and habituated overnight. For the baseline tests, mice were trained on Rotarod (3375-M5; TSE systems) first for three consecutive days. Mice were placed (having their back turned towards the experimenter) on the rotating rod of 4.0 cm diameter. Mice trained to stay on the rod for 200 seconds at one constant speed per day, incrementing speed each day from 4 rpm, 6 rpm, and 8 rpm. If a mouse fell during training, it was put back on the rod. For the test on the fourth day, the Rotarod started at 4 rpm and steadily accelerates to 40 rpm over a 5 minute interval. The speed at which mice dropped was recorded, in four consecutive trials. 2 and 12 weeks after baseline measurements mice were tested again, habituating overnight prior test day. The average was normalized to the baseline and taken as an indicator of mouse balance and motor coordination.
A water-motivated version of the Stone's T-maze was used to measure parameters of cognition. A straight run (for pre-training) or Stone's T-maze were placed into a steel pan filled with water to a depth of approximately 3 cm so that half the height of the interior walls of the maze were under water. The ceilings of both the straight run and maze were covered with clear acrylic to prevent mice from rearing out of the water. These dimensions created a situation that enables the mice to maintain contact with the floor while keeping their heads above water. The mice were placed into a start box and were pushed into the maze using a sliding panel. At the end of the straight run or maze there was a goal box that contains a ramp to a dry floor, which allows the mice to escape from the water upon successful completion of the straight run or maze. On day one, mice underwent straight run training to establish the concept that moving forward allows them to escape the water by reaching a water-free goal box. Successful completion of this phase requires the mice to reach the goal box in 10 seconds or faster in 8 out of 10 trials. Mice that did not reach this criterion were excluded from further testing. Maze training commenced the following day. Mice had to complete 9 maze acquisition trials in a single day. All mice per group performed one trial before performing the next one. Runs using between 6 and 8 mice resulted in inter-trial intervals (ITI) of approximately 5-12 minutes. During ITI, mice were placed in a holding cage containing a dry towel that was additionally heated by a red heat lamp. Primary measures of learning and memory were the latency to reach the goal box and the numbers of errors committed. An error was defined as complete entry of the mouse's head or the whole body into an incorrect path. During the acquisition phase, if any mouse failed to reach the goal box within 5 minutes, the trial was terminated and scored as a failure. Any mouse having 3 failures was removed from further trials. No mouse was excluded from this study.
Total RNA was extracted from white adipose tissue and brain using Trizol (Life Technologies, Carlsbad, Calif.) and reverse transcribed to cDNA with a M-MLV Reverse Transcriptase kit (Life Technologies). Real-time PCR was performed in a 7500 Fast Real Time PCR System (Applied Biosystems, Foster City, Calif.) using TaqMan Fast Universal PCR Master Mix (Life Technologies) and predesigned primers and probes from Applied Biosystems (Assay ID: Mm00494449_m1 [CDKN2A]; Mm04205640_g1 [CDKN1A]; Mm00446191_m1 [IL6]). Target gene expression was expressed as 2-ΔΔCT by the comparative CT method and normalized to the expression of TATA-binding protein (TBP) (Assay ID: Mm01277042 ml [TBP]).
On the day of the sacrifice, a small piece of adipose tissue was fixed with 2% PFA and 0.5% glutaraldehyde (Sigma) for 15 minutes at room temperature before being incubated overnight in SA-β-Gal solution (150 mM NaCl (Sigma), 2 mM MgCl2 (Sigma), 40 mM Citric Acid (Sigma), 12 mM NaPO3 (Sigma), 400 μg/ml X-gal (Thermofisher), 2.1 mg/ml potassium hexacyanoferrat(II)trihydrate, and 1.65 mg/ml Potassium hexacyanoferrat(III)trihydrate (Sigma), pH 6.0) at 37° C. overnight. Fat chunks were washed with PBS three times and stored in PBS at 4° C. protected from light. Within 3 days, adipose tissue was stained with Hoechst solution (1:5000; Thermofisher), lightly squashed between two 1×3 inch glass slides, and imaged using a light microscope. 10-20 random visual fields were captured at 20× magnification at light exposure identical for all the samples. Images were quantified by manual counting of SA-β-Gal positive cells by a blinded assessor and the data were expressed as percent of total DAPI-positive cells.
This technique uniquely combines time-of-flight mass spectrometry with metal-labelling technology to enable detection of up to 40 protein targets per cell. A panel of antibodies based on surface markers, transcription factors, and cytokines (see Table 1) was designed for brain mass cytometry/cytometry by time of flight (CyTOF). Each antibody was tagged with a rare metal isotope and its function verified by mass cytometry according to the factory manual (Multi Metal labelling Kits, Fluidigm, CA). A CyTOF-2 mass cytometer (Fluidigm, South San Francisco, Calif.) was used for data acquisition. Acquired data were normalized based on normalization beads (Ce140, Eu151, Eu153, Ho165, and Lu175). A single brain hemisphere was dissociated into a single-cell suspension using brain tissue dissociation kits (Adult Brain Dissociation Kit, Miltenyi Biotec Inc., CA). Collected cells were incubated with metal-conjugated antibodies and, for testing intracellular proteins, including transcription factors and cytokines, fixation and permeabilization was conducted according to the manufacturer's instructions (Transcription Factor Staining Buffer Set, eBioscience, San Diego, Calif.). CyTOF data were analyzed by Cytobank (Santa Clara, Calif.).
Serum levels of cytokines: Eotaxin, G-Csf, Tnf-α, Il-6, Ifn-γ, Il-1α, Il-1β, Il-17, Il-2, Kc/Cxcl1, Mcp-1, M-Csf, Mig, Mip-1α, and Mip-1β were determined using a Multiplexing LASER Bead Assay (Mouse Cytokine Array/Chemokine Array 31-Plex (MD31), Eve Technologies; Canada). Blood was withdrawn from mice by punctuation of the sub-mandibular vein at the day of dissection before an animal was sacrificed. 50 μL of serum were shipped to Eve Technologies on dry ice. Due to high variability of data an unbiased elimination of outliers was performed using ROUT's method (Graphpad 7 Prism). The same panel was used to detect SASP in MAF and 50 μl of media was shipped to Eve Technologies on dry ice.
MAF were extracted from 3-5 month old male C57BL/6 male mice. Ear clippings were transported and stored (not longer than 1 hour) in serum-free DMEM on ice. Punches were washed three times with serum-free media, finely cut, and incubated for 2-3 hours at 37° C. in DMEM containing 2 mg/ml collagenase A. A single-cell suspension was obtained by repeated pipetting and passing through a 24-G fine needle. Cells were centrifuged for 10 min at 1,000 r.p.m. and cultured in Advanced D-MEM/F-12 (DMEM, Invitrogen) plus 10% FBS (Sigma) in 3% 02 and 5% CO2. Each cell strain was derived from a separate donor mouse and expanded until enough cells are generated for freezing aliquots. For each experiment, MAFs were defrosted, seeded and allowed to grow for 24 hours and then X-ray irradiated with 10 Gy using a PXI X-Rad 225 (RPS Services Ltd) to induce cellular senescence. Media were changed twice a week. The last medium change was performed at day 20 after senescence induction (IR) and cells were fixed in 2% PFA the next day.
For cytokine measurements media from the last 24 hours of culture (before cell fixation) were sent to Eve Technologies for SASP assessment (Mouse Cytokine Array/Chemokine Array 31-Plex (MD31)).
Under normal conditions MAFs were kept in Advanced DMEM/F-12 (DMEM, Invitrogen) supplemented with 10% fetal bovine serum (FBS) (Sigma), 100 IU/ml penicillin/streptomycin, and 2 mM L-glutamine. In order to reduce content of lipids in tissue culture media, lipid-deprived FBS (Biowest) was used. Therefore, media containing standard FBS (with lipids) were designated “LIPID” and media containing lipid-deprived FBS were designated as “NO LIPID”. Young (control) cells were kept for at least 7 days under “NO LIPID” conditions before they were collected or senescence was induced.
Astrocytes were extracted from 16-day-old embryo brains of either sex. At 16th day of pregnancy mice were sacrificed and brains of embryos were dissected. Neocortex was isolated and homogenized by pipetting through a fire-polished, FBS-coated Pasteur pipette. Bigger pieces of the neocortex were isolated by sedimentation and supernatant was centrifuged to isolate astrocytes. Astrocyte cultures were seeded at a density of 0.5×106 cells/ml on culture dishes that had been coated previously with 15 μg/ml poly-1-ornithine overnight and subsequently washed with H2O and PBS. Astrocytes were maintained in DMEM/F12 medium supplemented with 5 mM HEPES, 33 mM glucose, 13 mM sodium bicarbonate, 10% fetal bovine serum, 2 mM glutamine 100 U/ml penicillin and 100 μg/ml streptomycin (all from Invitrogen). Cells were cultured at 37° C. in a humidified atmosphere of 5% CO2 and 3% 02. Induction of senescence and assessment of senescence markers and the ALISE phenotype were performed as for MAFs.
MAFs and astrocytes were plated on 19 mm (diameter) coverslips and at the end of experiments washed briefly with PBS and fixed for 10 minutes with 2% paraformaldehyde dissolved in PBS. Cells were permeabilized for 5 minutes with 0.5% TRITON X-100 dissolved in PBS. Cells were incubated with blocking buffer (5% normal goat serum (S-1000, Vector Laboratories) in PBS) for 60 minutes at room temperature. Plin2 (PROGEN #GP46, 1:250) 53BP1 (Novus Biologicals, #NB100-304, 1:250) and GFAP (Synaptic Systems, #173 004, 1:1000) antibodies were diluted in blocking buffer and applied overnight at 4° C. The next day, cells were washed three times with PBS and incubated for 60 minutes with secondary Alexa Fluor 594, goat, anti-guinea pig antibody (1:1000) for Plin 2 staining; Alexa Fluor, goat, anti-guinea pig (1:1000) for 53BP1 staining or Alexa Fluor, goat, anti-guinea pig (1:1000) for GFAP staining. For quantification of senescence markers coverslips were washed 3 times in PBS, then mounted in Vectashield, DAPI-containing mounting media. For assessment of lipid accumulation cells were washed 3 times with PBS before and after DAPI solution (PARTEC) was added for 30 minutes at room temperature. 2 μl of Nile red solution (Nile red (Sigma N3013) 150 μg ml−1 in acetone) were added to 1 ml 80% glycerol (in Milli-Q water) and mixed thoroughly. 20 μl of Nile Red/glycerol were directly added to each cell sample and mounted on a glass microscope slide. Images were taken immediately after mounting using a Leica DM5500 widefield fluorescence microscope with a 20× objective lens. Area of lipid droplets was quantified using ImageJ (“Analyze particles” tool) in >50 cells in ≥10 images.
For EdU experiments, HFD and control chow-fed INK-ATTAC mice treated with AP were injected with EdU (Life Technologies) at a dose of 123 mg/kg with the final concentration of 6.15 mg/mL, dissolved in sterile PBS (pH 7.4, Fisher Scientific), 2 hours before perfusion. 15 minutes intervals were allowed between mice injections to consider time needed for perfusing each mouse. The animals were deeply anesthetized with 90 mg/kg ketamine and 10 mg/kg of xylazine in sterile PBS prior perfusion. Transcardiac perfusion with PBS was followed by perfusion with 4% paraformaldehyde in PBS chilled on ice. Brains were harvested and postfixed overnight in 4% paraformaldehyde in PBS at 4° C., washed with PBS, and stored at 4° C. for vibratome sectioning. Sagittal brains sections of 50 μm were cut on a vibratome and collected sequentially in 6 different plate wells, total of 13 sections, 250 μm apart in each well, representing ⅙ of the brain hemisphere. Sections were stained free-floating in 12 well-plates, all procedures performed at room temperature at a volume of 500 μL for each well. Sections were initially permeabilized in 4% Triton X-100 (Sigma-Aldrich) in PBS for 1 hour with subsequent PBS washing for three times. Click reaction was performed for EdU visualization including 20 mM (+)-sodium L-ascorbate (Sigma-Aldrich), 10 μM Alexa 555-azide (Life Technologies), and 4 mM copper sulfate (Sigma-Aldrich) in PBS. Sections were incubated with gentle shaking for 15 minutes followed by PBS washing. Brain sections were collected and placed on gelatinized glass slides. All preparations were mounted with fluorescent mounting medium (DAKO) and coverslipped.
For assessment of hippocampal neurogenesis, EdU+ cells slides were imaged using a Leica DM5500B fluorescence microscope in depth Z stacking was used. Cells were manually counted in the basal layer of dentate gyrus in all 13 sections and multiplied by 6 to obtain as estimate of the number of dividing cells per hemisphere.
For assessment of neurogenesis in the subventricular zone (SVZ), 13 sagittal, 50 μm-thick sections were imaged using a Leica DM5500B fluorescence microscope. In depth Z stacking was used (images were captured as stacks separated by 4 μm with a 10× objective). Quantity of positive cells was manually counted in the ventral SVZ using ImageJ and the total number of cells was normalized to the number of images taken.
Paraffin sections were deparaffinized with Histoclear and hydrated in an ethanol gradient followed by water and PBS. Antigen was retrieved by incubation in 0.01M citrate buffer (pH 6.0) at 95° C. for 10 minutes. Slides were placed in blocking buffer (1:60 normal goat serum [S-1000, Vector Laboratories] in 0.1% BSA/PBS) for 60 minutes at room temperature. For TAF staining, slides were additionally blocked with Avidin/Biotin (Vector Lab, #SP-2001) for 15 minutes each. Primary antibodies used (Table 2) were diluted in blocking buffer and applied overnight at 4° C. The next day, slides were washed 3 times with PBS and incubated for 30 minutes with secondary goat, anti-rabbit antibody (1:200; Vector Laboratories #BA-1000) for TAF staining or for 60 minutes with secondary Alexa antibody (Table 1). For TAF staining, Fluorescein-Avidin in PBS (1:500; #A-2011, Vector Lab) was applied to each sample for 20 minutes. Slides were washed 3 times in PBS, which was followed by FISH for TAF detection. Briefly, tissues were crosslinked with 4% paraformaldehyde for 20 minutes and dehydrated in graded ethanol. Sections were denatured for 10 minutes at 80° C. in hybridization buffer (70% formamide (Sigma), 25 mM MgCl2, 0.1 M Tris (pH 7.2), and 5% blocking reagent [Roche]) containing 2.5 μg ml−1 Cy-3-labelled telomere-specific (CCCTAA) peptide nucleic acid probe (Panagene), followed by hybridization for 2 hour at room temperature in the dark. Slides were washed twice with 70% formamide in 2×SSC for 15 minutes, followed by washes in 2×SSC and PBS for 10 minutes. Sections were mounted in Vectashield, DAPI-containing mounting media and imaged.
A single, 3 μm-thick section per mouse was used for TAF staining, while for Dcx and Plin2 staining was performed on three 10 μm-thick sections 80 μm-apart. To quantify periventricular lipid accumulation 10-30 images in the periventricular region were taken using the DM5500 widefield fluorescence microscope from Leica with a 10× (for frequency of periventricular glia) or ×40 (for ALISE phenotype of ependymal cells) objective lens. Number of Plin2-positive cells (for frequency of ALISE-positive periventricular glia) or area of Plin2-positive vesicles (for ALISE phenotype of ependymal cells) was assessed using ImageJ software. For identity assessment of Plin2+ cells, 10 μm-thick sections were stained with combination of antibodies for Iba1 (secondary antibody conjugated with Alexa Fluor 488), Plin2 (secondary antibody conjugated with Alexa Fluor 594), and Vimentin (secondary antibody conjugated with Alexa Fluor 647) and quantified for frequency of Plin2+ astrocytes (Iba1−, Vim+) and microglia (Iba1+) in the periventricular region. A separate staining for Plin2 (secondary antibody conjugated with Alexa Fluor 594) and NeuN (secondary antibody conjugated with Alexa Fluor 647) was used to determine frequency of Plin2+ neurons in periventricular region. For TAF quantification in depth Z stacking was used (images were captured as stacks separated by 0.4 μm with ×63 objective) followed by ImageJ analysis.
Tissue distribution glial fibrillary acidic protein (GFAP) in the brain was assessed by immunohistochemistry using an image analysis workstation after staining with antibodies. The brain sections were pretreated with 0.3% H2O2 methanol for 1 hour at room temperature and with normal goat serum for 1 hour at room temperature. Each specimen was incubated with the primary antibody overnight at 4° C. The primary antibody used in this study and the dilutions were as follows: rabbit anti-cow glial fibrillary acidic protein (GFAP) [1:800, DAKO, Denmark]. Immunohistochemistry was performed using the VECTASTAIN ABC System (Vector Laboratories Inc., Burlingame, Calif.) with the avidin/biotin peroxidase complex (ABC) method. Negative controls included replacement of the primary antibodies with normal rabbit serum [1:200, DAKO]. The immunoreactivity to rat positive control specimen of the primary antibodies was determined before use.
RNA-ISH was performed after RNAscope protocol from Advanced Cell Diagnostics Inc. (ACD). Paraffin sections were deparaffinized with Histoclear, rehydrated in graded ethanol (EtOH) and H2O2 was applied for 10 minutes at RT followed by two washes in H2O. Sections were placed in hot retrieval reagent and heated for 15 minutes. After washes in H2O and 100% EtOH sections were air dried. Sections were treated with protease plus for 30 minutes at 40° C., washed with H2O and incubated with target probe (p16) for 2 hours at 40° C. Afterwards, slides were washed with H2O followed by incubation with AMP1 (30 minutes at 40° C.) and next washed with wash buffer (WB) and AMP2 (15 minutes at 40° C.), WB and AMP3 (30 minutes at 40° C.), WB and AMP4 (15 minutes at 40° C.), WB and AMP5 (30 min at RT) and WB, and, finally, AMP6 (15 minutes at RT). RNAscope 2.5 HD Reagent kit-RED was used for chromogenic labelling. After counterstaining with haematoxylin, sections were mounted.
For analysis of cytokines (Il-6 and Cxcl1) sections were co-stained with antibodies for Plin2 and S100β. Briefly, following chromogenic labelling for cytokines, sections were washed 3 times in TBS for 5 minutes each followed by blocking in 0.1% BSA in PBS for 30 minutes at RT. Sections were incubated overnight with primary antibodies at 4° C. Next, sections were washes 3 times in TBS for 5 minutes each followed by secondary antibody incubation for 1 hour at RT. After 3 TBS washes sections were mounted using ProLong Gold mounting media containing DAPI. Probes used: Cdkn2a: 411011, Il-6: 315891, Cxcl1:407721 (all from ADC).
For all RNA-ISH experiments data was analyzed by quantifying the % of positive cells (which means each cell containing at least 1 focus was counted as positive).
Data are presented as mean±SEM for all data. All statistical analyses including testing the normality of data distribution were performed using GraphPad Prism 7.01 and a P value <0.05 was considered as significant. The study was designed to compare change in parameters between lean and obese animals and between obese and obese treated animals. All data were assessed for normality using D'Agostino & Pearson normality test (for n>7) or Shapiro-Wilk normality test (for n 7≥n>3). For 2-group comparisons and planned comparison 2-group comparisons (were used where appropriate when main effects were significant without significant interactions) data was further tested for equality of variances using F test. For non-normally distributed datasets (p<0.05 in D'Agostino & Pearson or Shapiro-Wilk normality tests) Mann-Whitney U test was used. For normally distributed datasets Welch's t-test (if p<0.05 in F test) or Student's t-test was used. For 2> groups comparisons one-way ANOVA with Tukey's multiple comparison test was used. For datasets split on two independent factors two-way ANOVA was used. Correlations were assessed using Pearson's (for datasets of normal distribution) or Spearman's (for datasets of non-normal distribution) rank correlation test.
It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.
This application claims priority to U.S. Application Ser. No. 62/782,995, filed on Dec. 20, 2018. The disclosure of the prior application is considered part of the disclosure of this application, and is incorporated in its entirety into this application.
Filing Document | Filing Date | Country | Kind |
---|---|---|---|
PCT/US2019/067147 | 12/18/2019 | WO | 00 |
Number | Date | Country | |
---|---|---|---|
62782995 | Dec 2018 | US |