The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×

Abstract

Objective

It is well established that personal experiences of trauma, adversity, and discrimination can “get under the skin” and increase risk for a whole host of negative mental and physical health outcomes. The aim of this article is to review emerging research on transgenerational epigenetic inheritance which suggests that negative exposures in one generation, can also be passed down to affect the health and well‐being of future generations.

Method

This paper reviews key concepts in transgenerational epigenetic inheritance research, select animal and human studies examining the role of epigenetic mechanisms in transmitting the impact of ancestral stress and trauma, poor nutrition, and toxicant exposure across generations, and factors that can mitigate the effects of these experiences.

Results

The animal studies provide compelling evidence for a role for these mechanisms in the transmission of the negative effects associated with ancestral adversities. Animal and clinical studies also suggest that the negative impact of personal and ancestral traumas can be prevented, with a role for in humans for evidence‐based trauma treatments, culturally adapted prevention and intervention programs, and enrichment opportunities strongly indicated.

Conclusions

Although comparable definitive data is lacking in multigenerational human cohorts, preliminary data supports a potential role for transgenerational epigenetic mechanisms in explaining persistent health disparities in the absence of personal exposures, and further elucidation of these mechanisms may guide the design of novel interventions. In addressing ancestral traumas, however, true change and healing will require acknowledgement of the harms that were done, and broader systemic policy level changes.

Highlights

  • Ancestral traumas can negatively impact the well‐being of grandchildren and great grandchildren.

  • While most research on transgenerational epigenetic inheritance has been conducted in animals, studies in humans have shown ancestor's exposure to trauma, poor nutrition, and toxic chemicals can impact the health of descendants across several generations.

  • A role for evidence‐based trauma treatments, culturally adapted prevention and intervention programs, and enrichment opportunities is strongly indicated to reduce and prevent the impact of personal and ancestral adversities.

  • In addressing ancestral traumas, however, true healing will require acknowledgement of the harms that were done, and broader systemic level changes.

The pandemic and the recent murders of George Floyd, Ahmaud Arbery, and others have shined a spotlight on health disparities and systemic racism in this country. Since Africans were first brought to this continent against their will by the Spanish in 1526 (1), government and societally sanctioned atrocities against African descendants have continued (2, 3). Historical trauma and ongoing systemic racism has a toll, not just on the psyche of African Americans, but on their physical health as well (4, 5, 6, 7). It is well established that personal experiences of trauma, adversity, and discrimination can “get under the skin” and increase risk for a whole host of negative psychiatric (8, 9, 10) and medical health problems (11, 12, 13, 14, 15, 16, 17) through stress, brain, epigenetic, and immune system mechanisms (7, 18, 19). Emerging data now also suggests that adversities and traumas in one generation, can be passed down to affect the health and well‐being of future generations. Through transgenerational epigenetic mechanisms, grandchildren and great grandchildren can be negatively impacted by ancestral traumas – even when they have not been directly exposed to any harm themselves (20, 21).

The concept of generational trauma was first introduced in 1967 by Vivian Rakoff who recorded markedly elevated rates of psychological distress among children of Holocaust survivors (22, 23). Since that initial publication, multiple investigators have reported elevated rates of psychological distress in the children (24, 25) and grandchildren (26, 27, 28) of Holocaust survivors. There have also been several epidemiological studies which suggest parental exposure to trauma and stress, inadequate nutrition, and toxicants can impact the health of descendants across several generations (29, 30, 31).

Systemic racism is associated with experiences of persistent discrimination and elevated rates of exposure to inadequate nutrition and toxicants (e.g., lead, air population) that impact an individual's health and well‐being. While economic success may prevent offspring exposure to inadequate nutrition and harmful toxicants, it cannot eliminate all experiences of discrimination. Enduring health disparities, therefore, are likely to be due to both personal and ancestral adversities.

This paper reviews key concepts in transgenerational epigenetic inheritance research, select studies examining the role of epigenetic mechanisms in transmitting the impact of ancestral stress and trauma, poor nutrition, and toxicant exposure across generations, and factors that can mitigate the effects of these experiences. Culturally adapted interventions that address historical trauma and systemic racism are also briefly discussed.

While it is beyond the scope of this manuscript to elaborate on the societal factors that perpetuate adversity and impact the health and well‐being of African Americans in the United States, the authors acknowledge that trauma did not end for this population with the Emancipation Proclamation. It continued through Jim Crow, lynchings, (3) and redlining practices (32, 33), and persists today via the well‐documented systemic biases in the criminal justice system (34). As depicted in Figure 1 (21), the current discriminatory criminal justice system policies not only negatively impact the person behind bars – but the entire family. Children whose fathers are incarcerated tend to have minimal contact with their parent while he is in prison (32, 33), are apt to experience food insecurity (35), and are more likely to live in neighborhoods that are socioeconomically disadvantaged (36). These neighborhoods are associated with poorer quality schools, a concentration of environmental hazards, including lead and air pollution (37), fewer safe outdoor spaces for children to play, and higher rates of crime and community violence (38). And as we know from countless examples (39), even when African American families transcend the cycle of disadvantage propagated by government and societal policies and practices, privilege does not guarantee one can keep their loved ones safe.

image

FIGURE 1. Mass incarceration and the cycle of disadvantage.a

aThe current discriminatory criminal justice system negatively impacts the entire family and perpetuates a cycle of disadvantage that affects the health and well‐being of African Americans in this country. Reprinted from Kaufman et al. (21).

TRANSGENERATIONAL EPIGENETIC INHERITANCE: KEY CONCEPTS

Genetic inheritance occurs through the DNA passed from parents to their offspring through the gametes, with the DNA from the male carried in the sperm, and the DNA from the female is carried in the egg. When the egg and the sperm unite, they form a single cell which will have to multiply and make all the different cell types required for life. Every cell in the body has the same DNA, but different genes are turned on in different cells, making, for example, a neuron different than a cardiac muscle cell. The development of each different cell type is programmed through epigenetic mechanisms – chemical modifications to the DNA that change its three‐dimensional shape and the likelihood of a given gene product being turned on or off. Blood, kidney, and brain cells begin to develop by the fifth week in utero, with normal development proceeding through a predictable order, although the full range of molecular mechanisms responsible for normal embryonic and fetal development are not fully known (40).

Epigenetic mechanisms are also one of the ways experiences of trauma and adversity get “under the skin.” Methylation – the addition of a carbon atom with three hydrogen atoms to DNA – is known to shut off genes when added to the beginning of the gene sequence, and to increase expression when occurring elsewhere on the gene. One of the most highly replicated findings in the field of epigenetics research is that experiences of early life stress can lead to DNA methylation of the glucocorticoid receptor (GR) gene. First noted in 2004, by 2016 this finding had been replicated in 40 independent investigations, 13 animal and 27 human studies. The glucocorticoid receptor helps to turn off the stress response, and methylation of the GR gene is associated with reduced number of glucocorticoid receptors and heightened stress reactivity (41).

Whether or not these environmentally induced, epigenetic modifications can be inherited and transmitted across generations is an active area of research. Studies of transgenerational epigenetic inheritance are hard to execute in humans as it is difficult to obtain multigenerational cohorts and exclude psychosocial (e.g., poverty) and cultural (e.g., racism) confounders that may lead to common epigenetic, behavioral, and health outcomes across generations (29, 42). However, as reviewed in the following section, a growing body of animal research suggests the effects of traumatic stress and other negative exposures can be transmitted to subsequent generations through epigenetic mechanisms (21, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55).

For the environmentally induced epigenetic modifications to be inherited across non‐successive generations, they must be contained in the germline – the sperm or the egg – as these are the only two cells used to create subsequent life. Transgenerational epigenetic inheritance of traumatic stress and other negative exposures (e.g., lead) requires: 1) epigenetic modifications in the exposed animal/individual be present in the germline (e.g., sperm, egg); 2) epigenetic modifications in the exposed animal/individual be causally linked to the negative outcomes associated with the exposure; 3) the negative outcomes associated with the exposure be evident in subsequent generations with no history of exposure; and 4) the presence of the negative outcomes in the subsequent generations be causally linked to the epigenetic modifications initiated in the first exposed generation.

Figure 2 (56), depicts the experimental paradigm used to investigate transgenerational epigenetic inheritance in animals (56). The male or female that has the initial negative environmental exposure is labeled the F0 generation. As the impact of exposure can lead to epigenetic modifications to the germline (e.g., sperm, egg) of the F0 generation that creates the F1 generation, the F1 generation is considered ‘exposed’ as well. Transgenerational inheritance cannot be examined until the F2 generation. If as depicted on the right side of the diagram, the female is pregnant at the time of exposure, she (F0) is exposed; her developing baby (F1) is exposed, and its germline and the subsequent offspring (F2) is also considered exposed. The F3 generation (e.g., great grand offspring) would then be the first unexposed offspring in which transgenerational inheritance could be examined.

image

FIGURE 2. Transgenerational epigenetic inheritance: Experimental paradigm.a

aTransgenerational epigenetic inheritance requires the negative outcomes associated with the exposure in the original generation (F0) be evident in subsequent generations with no history of exposure; the F2 generation if the F0 was male or female and not gestating; the F3 generation if the F0 was female and pregnant at time of exposure. See text for full explanation. Code: F0 = first exposed generation; F1 = Offspring; F2 = Grand offspring; F3 = Great grand offspring (Figure reprinted with permission from Nilsson et al. (56)).

The three forms of epigenetic modifications linked most frequently to transgenerational inheritance include: DNA methylation, as discussed previously; histone modifications; and the action of non‐coding RNA molecules (ncRNA) (57). Histones are the proteins that act as spools to wind DNA, and chemical modifications to histones can also affect gene regulation. While RNA molecules are best known for their role in coding proteins; ncRNAs can also act as epigenetic factors and impact gene regulation (56).

Although evidence of transgenerational inheritance has been reported through the female germline (58), most transgenerational inheritance studies have focused on examining epigenetic factors in sperm due to the relative ease of obtaining large numbers of sperm cells for analyses. Eggs cannot be readily obtained or acquired in large enough quantity for traditional molecular analysis, and there are other factors that confound the testing of oocyte transmission. Currently more research is needed using evolving single‐cell analytic techniques and cross‐fostering designs to fully elucidate the role of the female germline (e.g., eggs) in epigenetic inheritance (56).

Because sperm develop behind a protective barrier (59), there was skepticism about the capacity for environmental exposures to elicit epigenetic modifications in sperm. Recently three independent teams were able to demonstrate that extracellular vesicles could transmit information about environmental stress and other adverse exposures to sperm, leading to epigenetic modifications that could be transmitted intergenerationally (59, 60, 61).

Skepticism about transgenerational epigenetic inheritance also flourished as DNA methylation and histone epigenetic marks are known to be erased after fertilization so the cells of the evolving embryo can be totipotent – capable of transforming into all the different cell types required for life (21, 30, 31). It has since been established that erasure and reprogramming is not complete, (53, 56, 59, 62, 63) and as discussed in the following section, DNA methylation, ncRNAs, and histone epigenetic marks appear to be involved in facilitating experience‐dependent transgenerational inheritance.

TRANSGENERATIONAL EPIGENETIC INHERITANCE: RESEARCH REVIEW

Rodent studies have reported epigenetic modifications in the germline (e.g., sperm) which have been causally linked to the inheritance of negative outcomes associated with stress, (50, 51, 59, 64, 65, 66) a high fat diet (67, 68, 69), and multiple chemical exposures (44, 46, 47, 53, 57, 58, 70, 71, 72, 73). Many of these studies, however, only provide evidence of multigenerational transmission (see Figure 2). Select studies that provide evidence of true transgenerational inheritance – with the impact of ancestral exposures evident in subsequent generations with no history of exposure – are reviewed here.

Dias and Ressler used fear conditioning to study the transgenerational transmission of traumatic stress (64). They subjected F0 mice to odor fear conditioning before conception by pairing an odor, an innocuous stimulus, with a shock, so that with time the odor alone elicited fear. They then found that subsequently conceived F1 (e.g., children/offspring) and F2 (e.g., grandchildren/grand offspring) generations had an increased behavioral sensitivity (e.g., startle response) to the F0‐conditioned odor, but not to other odors, despite no prior exposure to the odor or shocks. F0 mice subjected to fear conditioning and their F1 offspring were also found to have epigenetic marks in their sperm in the odorant receptor gene (Olfr151). Enhanced behavioral response to the F0‐conditioned odor was also associated with alterations in the olfactory epithelium and olfactory bulb in the F1 and F2 generation offspring of F0 fear conditioned mice. These same neuroanatomical alterations were also observed in odor naïve mice generated using in vitro fertilization (IVF) with sperm from the F0 fear conditioned mice, suggesting the neuroanatomical changes to the olfactory system were transmitted through the male germline (e.g., sperm). Due to animal quarantine issues, however, behavioral studies could not be conducted with the IVF‐generated offspring.

To determine if behavioral sensitivity to the conditioned stimuli could be transmitted via the female, Dias and Ressler conducted a cross‐fostering study using the design depicted in Figure 3 (64). Sexually naive female mice were conditioned with the odor (i.e., fear conditioned) or left in their home cage (control). They were then mated with odor‐naive males. Immediately after birthing their offspring were then divided into the following groups: (A) offspring of the fear‐conditioned mothers raised by the fear‐conditioned mothers; (B) offspring of the control mothers raised by the fear‐conditioned mothers; (C) offspring of the fear‐conditioned mothers raised by the control mothers; and (D) offspring of the control mothers raised by the control mothers. The females were only exposed to the odor conditioning before mating, and never while pregnant, precluding in utero exposure. Offspring of the fear‐conditioned mice, whether raised by fear‐conditioned mothers, or raised by the control mothers, exhibited increased behavioral sensitivity to the F0‐conditioned odor, suggesting conditioned fear responses can be transmitted via the female germline as well.

image

FIGURE 3. Cross‐fostering model.a

aTo test the transmission of fear conditioning through the female germline, Dias and Ressler (64) conducted a cross‐fostering study. Immediately after birth the offspring were divided into the following groups: A. offspring of the fear‐conditioned mothers raised by the fear‐conditioned mothers; B. offspring of the control mothers raised by the fear conditioned mothers; C. offspring of the fear‐conditioned mothers raised by the control mothers; and D. offspring of the control mothers raised by the control mothers. Offspring of the fear‐conditioned mice, whether raised by fear‐conditioned mothers, or raised by the control mothers, exhibited increased behavioral sensitivity (e.g., fear) to the F0‐conditioned odor, suggesting conditioned fear (e.g., psychological distress) can be transmitted via the female germline as well.

Yao and colleagues examined the impact of ancestral and multigenerational stress on maternal weight gain, gestational length, maternal blood glucose levels, and offspring weight in a four generation study (66). Pregnant rats in the first (F0) generation were exposed to stress from gestational days 12 to 18. In this study, “stress” involved being put in a confined environment that limited movement and a brief forced swim challenge. The pregnant female offspring (F1) and grand female offspring (F2) of the F0 moms were either exposed to stress or left undisturbed (e.g., non‐stressed). Outcomes were examined in each generation, including in great grand offspring (F3). Stress reduced maternal weight gain in the F0 cohort and each successive generation, decreased gestational length beginning in the F1 cohort, and increased maternal blood glucose levels by the F2 cohort. Decreased offspring weight was evident by the F1 cohort and greatest in the F3 offspring of transgenerationally stressed mothers. As depicted in Figure 4 (66), transgenerational and multigenerational prenatal stress resulted in low birth weight among F3 offspring. In the diagram NNNN indicates there was no stress across the four generations; SNNN indicates only F0 transgenerational stress, and SSNN and SSSN are indicative of multigenerational stress.

image

FIGURE 4. Transgenerational and multigenerational prenatal stress effects on birthweight.a

aTransgenerational and multigenerational prenatal stress is associated with significantly reduced birthweight in offspring. Codes: NNNN = no stress across the four generations; SNNN = only F0 transgenerational stress; SSNN and SSSN = multigenerational stress. Figure adapted with permission from Yao et al. (66).

In addition to the impact on birth weight, offspring of prenatally, multigenerationally, and transgenerationally stressed mothers were reported to exhibit developmental delays. Yao and colleagues also conducted brain frontal cortex, uterus, and placenta ncRNA and gene expression analyses in F0‐N, F0‐S, and F2‐SSS animals, with results in F2 stressed animals demonstrating that a multigenerational history of prenatal stress is associated with changes in genes implicated in brain plasticity (e.g., miR‐200 family genes), parturition/childbirth (e.g., Zeb2), and preterm birth (e.g., miR‐181a) (66). The documentation of an impact of transgenerational and multigenerational stress on preterm birth is particularly interesting given racial disparities in rates of preterm birth, and recent findings that adequate prenatal care does not reduce racial disparities, with African American women who engage in adequate prenatal care still at elevated risk for preterm birth (74).

de Castro Barbosa and colleagues showed that a high‐fat diet could reprogram the epigenome of sperm and transgenerationally affect metabolism in the offspring (68). In this study, F0 male rats were fed either a high‐fat or normal chow‐diet for 12 weeks and then mated to normal chow‐fed females to create F1 and F2 generation offspring. Sperm were isolated from F0 and F1 males. The F0 male rats fed the high‐fat diet had increased body weight and impaired glucose tolerance. The F1 (e.g., offspring) and F2 (e.g., grand offspring) offspring of the F0 males fed the high‐fat diet had reduced birth‐weight when compared to the offspring of chow‐fed F0 males; with low birth‐weight a documented risk factor for obesity and type 2 diabetes (75, 76). F0 male rats fed the high‐fat diet and their F1 male offspring had common sperm DNA methylation and small ncRNA expression signatures – with several of the epigenetic sites identified in genes implicated in the regulation of glucose homeostasis, insulin sensitivity, and a predisposition to Type 2 diabetes (e.g., let‐7c) (68). Consistent with these data demonstrating the role of a high‐fat diet in programming the epigenome of sperm to affect the metabolism of the offspring, Grandjean and colleagues showed that microinjection of either testis or sperm ncRNA of male mice fed a high‐fat diet into naive one‐cell embryos lead to the establishment of the high‐fat diet‐induced metabolic phenotype (e.g., insulin resistance, impaired glucose tolerance) in the resulting progenies, whereas ncRNAs prepared from healthy controls did not (69).

To the best of our knowledge, no rodent transgenerational studies have examined the impact of lead exposure, a major public health hazard for African American urban children, with profound and well‐characterized developmental and behavioral implications across the lifespan (77). Meyer and colleagues used zebrafish to study the transgenerational repercussions of lead exposure (77). F0 embryos were exposed for 24 h to waterborne lead. The F0 generation zebrafish were then raised to adulthood and F1 and F2 generation offspring, who had no direct lead exposure, were then studied. The dosage of lead exposure used in this investigation was previously found to generate learning impairments in zebrafish, and similar learning impairments were found to be present in the F2 offspring of F0 lead exposed zebrafish. RNA was extracted from the brains of the F2 grand offspring of control and lead‐exposed F0 zebrafish. Significant expression differences were found in 648 genes, with path analyses revealing altered expression in genes involved in brain development (e.g., synaptic function and plasticity, neurogenesis), endocrine homeostasis, and epigenetic processes – genes which may be involved in lead‐induced neurobehavioral deficits and/or their inheritance. These data provide an initial step in demonstrating the potential transgenerational health effects of lead exposure (77).

The observation by Dias and Ressler that in vitro fertilization with sperm from F0 fear conditioned mice generated offspring that had the same olfactory perception brain changes that were observed in the F1 and F2 generation offspring of F0 fear conditioned mice provides strong support that transgenerational epigenetic inheritance is transmitted through the male germline (e.g., sperm) (64). Comparable support for the role of the male germline in epigenetic inheritance has been reported in multiple other studies using similar methodology across fewer generations. For example, Gapp and colleagues injected sperm ncRNAs from males subjected to an early stress paradigm into eggs and produced offspring with the behavioral and metabolic alterations associated with their early stress experimental paradigm (65); Rodgers and colleagues generated offspring with patterns of stress dysregulation observed in mice subjected to their chronic stress paradigm by microinjecting a zygote with sperm ncRNAs altered by the chronic stress protocol (52); and Chan and colleagues produced offspring with neurodevelopment and stress reactivity indices similar to their stress‐treated animals using assisted reproductive technology with sperm from naïve adult male mice that was incubated with extracellular vesicles from stress‐treated animals (59).

The biological relevance of the genes regulated by the epigenetic marks identified in the studies of transgenerational epigenetic inheritance also provides compelling support for the role of these mechanisms in the transmission of experience‐dependent traits and health problems. To review, the F0 mice subjected to fear conditioning using an odor and their F1 offspring were found to have epigenetic marks in their sperm in a gene critical to olfactory perception (64). A multigenerational history of prenatal stress which promoted reduced gestational length and developmental delays in the offspring was associated with changes in genes implicated in brain plasticity, parturition/childbirth, and preterm birth (66). F0 male rats fed the high‐fat diet and their F1 male offspring had epigenetic alterations in genes implicated in the regulation of glucose homeostasis, insulin sensitivity, and a predisposition to Type 2 diabetes (68); and the F2 offspring of lead‐exposed F0 zebrafish had significant expression differences in genes involved in brain development (e.g., synaptic function and plasticity, neurogenesis) which may be relevant in understanding lead‐induced cognitive and neurobehavioral deficits.

There is also emerging data suggesting the relevance of this research for understanding the transgenerational transmission of the effects of adversity and other negative exposures in human cohorts. Beyond the epidemiological studies which suggest parental exposure to trauma and stress, inadequate nutrition, and toxicants can impact the health of descendants across several generations (29, 30, 31), several investigators have documented the presence of the epigenetic marks noted in the rodents in human samples. For example, alterations in the ncRNAs (e.g., miR‐16, miR‐37) reported in the sperm of mice subjected to maternal separation have been observed in the serum of children aged 7–12 years of age who experienced paternal loss and maternal separation, the serum of adult men aged 18–25 years of age who likewise experienced paternal loss and maternal separation at a young age, and the sperm of adult men aged 21–50 years of age who experienced two or more significant traumatic events in childhood (65, 78). In another study, male adults with a history of early life stress exhibited reduction in a ncRNA (e.g., miRNA‐434) in sperm that was also reported to be altered in a mouse model of early life stress (79). The finding of alterations in this particular ncRNA in the sperm of adults with histories of early life stress was also replicated in an independent sample (78). Experiences of recurrent stress in healthy adult males was also found to be associated with changes in ncRNAs detected in sperm that were identical to the ncRNA changes reported in a mouse study of chronic stress in adult animals (80).

While more work is needed to fully elucidate the mechanisms by which experience can alter the epigenome and impact health and developmental trajectories in subsequent generations, the accumulating body of animal research is quite compelling. The role of the female germline in transgenerational epigenetic inheritance requires further investigation, but the cross‐fostering study by Dias and Ressler (64), and the four‐generation pregnancy stress investigation by Yao and colleagues (66), suggest a role for the female germline in epigenetic inheritance (56). Elucidation of the molecular mechanisms involved in environmentally induced epigenetic transgenerational inheritance is essential to fully understand disease etiology (53), and has important implications for the development of novel prevention and treatment interventions to mitigate the negative impact of deleterious ancestral exposures. The relevance of this preclinical research in understanding human disease, however, requires carefully designed multigenerational studies.

FACTORS THAT MITIGATE THE EFFECTS OF HISTORICAL AND PERSONAL TRAUMA

The transgenerational negative effects demonstrated in the animal studies reviewed in the prior section can be prevented. Animal studies showing ways to mitigate the deleterious effects of the various exposures on the F0 and subsequent generations are highlighted in this section, with parallel and other promising human interventions also discussed.

In the initial study by Dias and Ressler (64), the pairing of an odor with a shock (e.g., fear conditioning) was used to model the effect of traumatic stress in the F0 and subsequent generations. In a follow‐up experiment, the same procedures were used, but a subset of the animals were provided “treatment” to eliminate the elicitation of fear by the odor (81). “Treatment” was comprised of extinction training – the gradual elimination of the conditioned response (e.g., fear when presented with the odor) by repeat presentation of the odor without any shocks. Animals that were initially conditioned to fear the odor and then provided “treatment” stopped exhibiting fear when exposed to the odor. Their offspring (F1) also did not show increased behavioral sensitivity (e.g., startle response) to the F0‐conditioned odor. In addition, the epigenetic changes observed in the sperm in the gene critical to olfactory perception was only evident in the mice conditioned to fear the odor and not provided any “treatment”; the mice that received extinction training (e.g., treatment) did not have these epigenetic marks in their sperm. It appears “treatment” can prevent the transgenerational transmission of the negative effects associated with ancestral traumatic stress.

Extinction training is at the core of all evidence‐based psychotherapeutic approaches for treating Posttraumatic Stress Disorder (PTSD) in children, adolescents, and adults (e.g., Exposure Therapy, Trauma‐Focused Cognitive Behavioral Therapy), with talking about and visualizing the traumatic events (e.g., repeat exposure) paired with relaxation training and cognitive processing (82, 83, 84). These interventions are highly effective in diverse populations for a broad range of traumatic experiences (e.g., sexual abuse, intimate partner violence, community violence, traumatic loss of a loved one).

de Castro Barbosa and colleagues showed that a high‐fat diet could reprogram the epigenome of sperm and transgenerationally affect metabolism in the offspring (68). An independent group using a similar mouse model demonstrated that diet or exercise interventions for 8 weeks in obese males prior to conception prevented the development of metabolic problems (e.g., insulin sensitivity, excess adipose tissue) in the offspring (85). Paternal diet and exercise also prevented changes to sperm ncRNAs. We are not aware of comparable multigenerational obesity interventions in humans, but these animal studies suggest preconception diet and exercise programs may help to break the transmission of obesity and associated negative health outcomes (e.g., diabetes, cardiovascular disease, cancer, and premature mortality). There is, however, a plethora of data that suggests adopting a healthy lifestyle can diminish an individual's risk for obesity and these other health problems. Reducing intake of red meat (86, 87), consuming plant protein over animal protein (88), having regular portions of fruit (86, 87), eating foods rich in antioxidants or taking antioxidant supplements (89, 90), refraining from excessive alcohol use (86, 87), avoiding smoking (87, 91), doing physical exercise, (87, 91, 92) and engaging in mindfulness‐based stress reduction activities (89, 90) are all associated with longevity and reduced risk of these stress‐related health problems. As food deserts – areas lacking in affordable healthy foods – are concentrated in minority neighborhoods (93), federal efforts to enhance access to quality foods through the Healthy Food Financing Initiative, which provides incentives for healthy food retailers to open stores in areas lacking access to nutritious fresh food, may be an important first step in addressing the obesity epidemic in the Black community (94). Available data, however, suggests that access alone is not always sufficient to improve residents' diets (94, 95, 96), indicating additional targeted interventions are required.

Many of the other deleterious transgenerational effects reviewed in the prior section were found to be prevented when, after the initial negative exposures, the F0 cohort was provided enrichment experiences (e.g., living in enhanced spaces that included toys to provide rich social, physical, and sensory experiences). For example, female mice subjected to prenatal stress who were subsequently provided enrichment experiences did not experience preterm birth and their offspring did not show any developmental delays (97, 98), male mice subjected to early stress who were provided enrichment experiences did not exhibit the sperm epigenetic changes associated with the early stress paradigm and there was no transmission of any stress‐related behaviors to their offspring, (99) and providing enrichment experiences to female rats exposed to lead while gestating prevented the development of lead exposure‐related deficits in the cognitive performance of their offspring (100).

Studies in children, adolescents, and young adults also suggest a role for enrichment experiences in mitigating the impact of personal and ancestral traumas. The Harlem Children's Zone programs and the Carolina Abecedarian Project, which provided educational enrichment programs to low income African American youth, have been found to promote resilience and a range of long‐term positive developmental outcomes which are sustained across time and generations (101, 102, 103). Participation in team sports is also associated with resilience, specifically, reduced mental health problems among youth with histories of significant childhood adversities (104). The Stanford Medical Youth Science Program, which provides academic enrichment in the sciences and college admissions support to very low‐income minority high school students, most with poor academic preparation, has also been associated with very positive outcomes (103). Of the more than 400 youth who completed the program, 99% have been admitted to college, 81% earned a four‐year college degree, and among four‐year college graduates, 52% are attending or have graduated from medical or graduate school.

CULTURAL ADAPTATIONS OF PREVENTION AND INTERVENTION PROGRAMS

The Strong African American Families (SAAF) intervention is an evidence‐based intervention developed for 11‐year‐old youth which is designed to enhance the parent‐child relationship and address issues unique to African American youth (e.g., racial socialization, racism). The developers of SAAF have also created programs for older youth, and programs to enhance parenting relationships (105, 106, 107, 108, 109). SAAF was initially developed for youth from low‐income families from disadvantaged neighborhoods in rural Georgia; however, it is currently being implemented in urban communities around the nation (110, 111), including 24 social services agencies in Harlem (112). SAAF consists of seven consecutive 2.5‐h weekly family group meetings held at community facilities, with separate skill‐building curricula for youths and their primary caregivers. The caregiver sessions emphasize positive parenting skills, including the consistent provision of instrumental and emotional support, high levels of monitoring and control, adaptive racial socialization strategies, and methods for communicating about sex and alcohol use. Youth sessions focus on forming goals for the future and making plans to attain them, resistance efficacy skills, and adaptive behaviors to use when encountering racism. At SAAF meetings, families eat a meal together and then divide into small parent and child discussion groups. For the final hour of each session, the caregivers and youth reunite for a two‐generation group meeting.

The SAAF program has been associated with positive outcomes on child behavioral problems, health risk behaviors, health problems, and number of physiological indices. Specifically, SAAF participation has been associated with decreased rates of conduct problems in youth two years after the intervention (113); reduced rates of smoking (114), drinking (115), drug use, (116) and risky sexual behaviors (117) in late adolescence and early adulthood; and reduced risk of obesity (118) and prediabetes in young adulthood (119). In the latter study, adverse childhood experiences were not associated with risk for prediabetes in young adults who participated in the SAAF intervention, but among the youth in the control intervention, each additional experience of adversity was associated with a 37% increase in risk for prediabetes (119). In terms of physiological indices, the SAAF intervention was associated with reducing the impact of family risk factors on stress system (e.g., adrenaline, norepinephrine) (120), inflammation (121), and epigenetic (122, 123) markers. The parenting‐focused SAAF intervention was also associated with diminishing the impact of poverty on hippocampal and amygdala brain volumes measured in adulthood—key brain regions affected by stress (124). The investigators note their findings are consistent with a possible role for supportive parenting in brain development, and appear to suggest a strategy for narrowing social disparities (124). The programs focus on positive racial socialization and equipping youth to deal with racism likely also greatly contributes to the success of the intervention.

Over the past two decades there have been over a dozen meta‐analyses examining the effectiveness of culturally adapted psychotherapeutic interventions (125). Positive findings have been reported for individual, group, and family culturally adapted treatments (126), with culturally adapted interventions associated with better outcomes when compared to the same intervention without the adaptations (g = 0.52, medium effect size). In a large meta‐analysis with nearly 14,000 participants, culturally adapted interventions had a 4.68 times greater likelihood of producing remission from psychopathology than the non‐adapted version of the intervention (125). The success of cultural adaptations of prevention and intervention programs in producing positive treatment outcomes and mitigating the impact of personal adversity highlights the importance of utilizing these approaches to address health disparities and promote resilience and recovery.

CONCLUSIONS

Research in animals strongly suggests that ancestral traumas can impact the health and well‐being of generations to come through transgenerational epigenetic transmission. Ongoing work is required, however, to fully delineate the mechanisms involved. Although comparable definitive data is lacking in multigenerational human cohorts, epidemiological studies in humans have shown ancestors' exposure to trauma, poor nutrition, and toxic chemicals can impact the health of descendants across several generations, and some of the epigenetic modifications noted in animal studies have also been reported in humans. These data provide preliminary support for a potential role for transgenerational epigenetic mechanisms in explaining persistent health disparities in the absence of personal exposures, and further elucidation of these mechanisms may guide the design of novel interventions. The review of animal and clinical studies also highlighted that the negative impact of ancestral traumas can be prevented, with a role for evidence‐based trauma treatments, culturally adapted prevention and intervention programs, and enrichment opportunities strongly indicated. In addressing ancestral traumas, however, true change and healing will require acknowledgement of the harms that were done, and broader systemic policy level changes (3, 20).

Center for Child and Family Traumatic Stress, Kennedy Krieger Institute and Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD (J. Kaufman, M. Khan, J. Shepard Payne, J. Mancini, Y. Summers White)
Send correspondence to Dr. Kaufman
( or )

Supported by a grant from the National Coalition of Blacks for Reparations in America (N’COBRA, Dr. Kaufman) and a grant from the National Institute of Minority Health and Health Disparities (NIMHD; R01 MD011746; Dr. Kaufman).

Dr. Kaufman has received grant funding from NIH, has served as a consultant for Pfizer and Otsuka Pharmaceuticals, and has a proprietary financial interest in the computer‐administered KSADS (KSADS‐COMP, LLC). Dr. Payne has a proprietary financial interest as the developer of a culturally‐tailored Acceptance and Commitment Therapy intervention called POOF®. The remaining authors have no financial relationships with commercial interests.

REFERENCES

1 Guasco M. The misguided focus on 1619 as the beginning of slavery in the U.S. damages our understanding of American history. Smithsonian Magazine; 2017. https://www.smithsonianmag.com/history/misguided‐focus‐1619‐beginning‐slavery‐us‐damages‐our‐understanding‐american‐history‐180964873/Google Scholar

2 Alexander M. The New Jim Crow: mass incarceration in the age of colorblindness. New York, NY: New Press; 2020.Google Scholar

3 Stevenson B. Lynching in America: confronting the legacy of racial terror. Lynching in America; 2017.Google Scholar

4 Williams DR, Yan Y, Jackson JS, Anderson NB. Racial differences in physical and mental health: socio‐economic status, stress and discrimination. J Health Psychol. 1997;2(3):335–51. https://doi.org/10.1177/135910539700200305Google Scholar

5 James SA. The strangest of all encounters: racial and ethnic discrimination in US health care. Cad Saúde Pública. 2017;33(Suppl 1):e00104416. https://doi.org/10.1590/0102‐311X00104416Google Scholar

6 Bailey ZD, Krieger N, Agenor M, Graves J, Linos N, Bassett MT. Structural racism and health inequities in the USA: evidence and interventions. Lancet. 2017;389(10077):1453–63. https://doi.org/10.1016/S0140‐6736(17)30569‐XGoogle Scholar

7 Lockwood KG, Marsland AL, Matthews KA, Gianaros PJ. Perceived discrimination and cardiovascular health disparities: a multisystem review and health neuroscience perspective. Ann N Y Acad Sci. 2018;1428(1):170–207. https://doi.org/10.1111/nyas.13939. Epub 12018 Aug 13938Google Scholar

8 Teicher MH, Samson JA. Childhood maltreatment and psychopathology: a case for ecophenotypic variants as clinically and neurobiologically distinct subtypes. Am J Psychiatr. 2013;170(10):1114–33. https://doi.org/10.1176/appi.ajp.2013.12070957Google Scholar

9 Kendler KS, Bulik CM, Silberg J, Hettema JM, Myers J, Prescott CA. Childhood sexual abuse and adult psychiatric and substance use disorders in women: an epidemiological and cotwin control analysis. Arch Gen Psychiatry. 2000;57(10):953–9. https://doi.org/10.1001/archpsyc.57.10.953Google Scholar

10 Fisher HL, Jones PB, Fearon P, Craig TK, Dazzan P, Morgan K, et al. The varying impact of type, timing and frequency of exposure to childhood adversity on its association with adult psychotic disorder. Psychol Med. 2010;40(12):1967–78. https://doi.org/10.1017/S0033291710000231. Epub 0033291710002010 Feb 0033291710000224Google Scholar

11 Dong M, Giles WH, Felitti VJ, Dube SR, Williams JE, Chapman DP, et al. Insights into causal pathways for ischemic heart disease: adverse childhood experiences study. Circulation. 2004;110(13):1761–6. https://doi.org/10.1161/01.cir.0000143074.54995.7fGoogle Scholar

12 Felitti VJ, Anda RF, Nordenberg D, Williamson DF, Spitz AM, Edwards V, et al. Relationship of childhood abuse and household dysfunction to many of the leading causes of death in adults. The Adverse Childhood Experiences (ACE) Study. Am J Prev Med. 1998;14(4):245–58. https://doi.org/10.1016/s0749‐3797(98)00017‐8Google Scholar

13 Romans S, Belaise C, Martin J, Morris E, Raffi A. Childhood abuse and later medical disorders in women. An epidemiological study. Psychother Psychosom. 2002;71(3):141–50. https://doi.org/10.1159/000056281Google Scholar

14 Anda RF, Brown DW, Dube SR, Bremner JD, Felitti VJ, Giles WH. Adverse childhood experiences and chronic obstructive pulmonary disease in adults. Am J Prev Med. 2008;34(5):396–403. https://doi.org/10.1016/j.amepre.2008.02.002Google Scholar

15 Dube SR, Fairweather D, Pearson WS, Felitti VJ, Anda RF, Croft JB. Cumulative childhood stress and autoimmune diseases in adults. Psychosom Med. 2009;71(2):243–50. https://doi.org/10.1097/psy.0b013e3181907888Google Scholar

16 Brown DW, Anda RF, Felitti VJ, Edwards VJ, Malarcher AM, Croft JB, et al. Adverse childhood experiences are associated with the risk of lung cancer: a prospective cohort study. BMC Publ Health. 2010;10(20):20. https://doi.org/10.1186/1471‐2458‐10‐20Google Scholar

17 Thompson E, Kaufman J. Prevention, intervention, and policy strategies to reduce the individual and societal costs associated with adverse childhood experiences (ACEs) for children in Baltimore City. Baltimore, MD: Abell Foundation; 2019.Google Scholar

18 Aroke EN, Joseph PV, Roy A, Overstreet DS, Tollefsbol TO, Vance DE, et al. Could epigenetics help explain racial disparities in chronic pain? J Pain Res. 2019;12:701–10. https://doi.org/10.2147/JPR.S191848. eCollection 192019Google Scholar

19 Danese A, McEwen BS. Adverse childhood experiences, allostasis, allostatic load, and age‐related disease. Physiol Behav. 2012;106(1):29–39.Google Scholar

20 Kaufman J, Khan M, Mancini J, Summers White Y. Transgenerational epigenetic inheritance and systemic racism in the United States: a report prepared for the National Coalition of Blacks for Reparations in America (N’COBRA); 2021. Published on National Coalition of Blacks for Reparations in America (N’COBRA) website. Author Affiliations: Center for Child and Family Traumatic Stress, Kennedy Krieger Institute; Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD. https://www.ncobraonline.org/harmreport/Google Scholar

21 Ben Maamar M, Nilsson EE, Skinner MK. Epigenetic transgenerational inheritance, gametogenesis and germline development. Biol Reprod. 2021;105(3):570–92. https://doi.org/10.1093/biolre/ioab085Google Scholar

22 Rakoff VJJS, Epstein N. Children and families of concentration camp survivors. Can Ment Health. 1967;14:24–6.Google Scholar

23 Gillespie C. What is generational trauma? Here's how experts explain it. Explore Health Web site. Published 2020. Available from: https://www.health.com/condition/ptsd/generational‐traumaGoogle Scholar

24 Flory JD, Bierer LM, Yehuda R. Maternal exposure to the holocaust and health complaints in offspring. Dis Markers. 2011;30(2‐3):133–9. https://doi.org/10.1155/2011/250470Google Scholar

25 Danieli Y, Norris FH, Engdahl B. A question of who, not if: psychological disorders in Holocaust survivors' children. Psychol Trauma. 2017;9(Suppl 1):98–106. https://doi.org/10.1037/tra0000192Google Scholar

26 Sigal JJ, DiNicola VF, Buonvino M. Grandchildren of survivors: can negative effects of prolonged exposure to excessive stress be observed two generations later? Can J Psychiatry. 1988;33(3):207–12. https://doi.org/10.1177/070674378803300309Google Scholar

27 Iliceto P, Candilera G, Funaro D, Pompili M, Kaplan KJ, Markus‐Kaplan M. Hopelessness, temperament, anger and interpersonal relationships in Holocaust (Shoah) survivors' grandchildren. J Relig Health. 2011;50(2):321–9. https://doi.org/10.1007/s10943‐009‐9301‐7Google Scholar

28 Greenblatt‐Kimron L, Shrira A, Rubinstein T, Palgi Y. Event centrality and secondary traumatization among Holocaust survivors' offspring and grandchildren: a three‐generation study. J Anxiety Disord. 2021;81:102401. https://doi.org/10.1016/j.janxdis.2021.102401Google Scholar

29 Jawaid A, Jehle KL, Mansuy IM. Impact of parental exposure on offspring health in humans. Trends Genet. 2021;37(4):373–88. https://doi.org/10.1016/j.tig.2020.10.006Google Scholar

30 Ambeskovic M, Roseboom TJ, Metz GAS. Transgenerational effects of early environmental insults on aging and disease incidence. Neurosci Biobehav Rev. 2020;117:297–316. https://doi.org/10.1016/j.neubiorev.2017.08.002Google Scholar

31 Liberman N, Wang SY, Greer EL. Transgenerational epigenetic inheritance: from phenomena to molecular mechanisms. Curr Opin Neurobiol. 2019;59:189–206. Epub 2019 Oct 1018. https://doi.org/10.1016/j.conb.2019.1009.1012Google Scholar

32 Jan T. Redlining was banned 50 years ago: it's still hurting minorities today. The Washington Post; 2018.Google Scholar

33 Mitchell B, Franco J. HOLC “redlining” maps: the persistent structure of segregation and economic inequality; 2018.Google Scholar

34 The_Sentencing_Project . Report to the United Nations on racial disparities in the U.S. criminal justice system; 2018. Washington, D.C. https://www.sentencingproject.org/reports/report‐to‐the‐united‐nations‐on‐racial‐disparities‐in‐the‐u‐s‐criminal‐justice‐system/Google Scholar

35 de Vuono‐powell S, Schweidler C, Walters A, Zohrabi A. Who pays? The true cost of incarceration on families. Oakland, CA: Ella Baker Center, Forward Together, Research Action Design; 2015.Google Scholar

36 Scommegna . When a parent is incarcerated, partners and children also pay a price; 2020. https://www.prb.org/resources/when‐a‐parent‐is‐incarcerated‐partners‐and‐children‐also‐pay‐a‐price/Google Scholar

37 Mathiarasan S, Hüls A. Impact of environmental injustice on children's health‐interaction between air pollution and socioeconomic status. Int J Environ Res Public Health. 2021;18(2):795. https://doi.org/10.3390/ijerph18020795Google Scholar

38 Sacks V. 5 ways neighborhoods of concentrated disadvantage harm children. Child Trends; 2018. https://www.childtrends.org/publications/5‐ways‐neighborhoods‐of‐concentrated‐disadvantage‐harm‐children. Published February 14, 2018. Accessed 30 May 2021.Google Scholar

39 Bash D, Nolan B. Rep. Lucy McBath is living her son’s legacy. CNN; 2021. Published U.S. Edition. Published May 24, 2021 https://www.cnn.com/2021/05/23/politics/badass‐women‐lucy‐mcbath/index.htmlGoogle Scholar

40 Belmonte‐Mateos C, Pujades C. From cell States to cell fates: how cell proliferation and neuronal differentiation are coordinated during embryonic development. Front Neurosci. 2021;15:781160.Google Scholar

41 Turecki G, Meaney MJ. Effects of the social environment and stress on glucocorticoid receptor gene methylation: a systematic review. Biol Psychiatry. 2016;79(2):87–96. Epub 2014 Dec 1013. https://doi.org/10.1016/j.biopsych.2014.1011.1022Google Scholar

42 Scorza P, Duarte CS, Hipwell AE, Posner J, Ortin A, Canino G, et al. Research review: intergenerational transmission of disadvantage: epigenetics and parents' childhoods as the first exposure. J Child Psychol Psychiatry Allied Discip. 2019;60(2):119–32. https://doi.org/10.1111/jcpp.12877Google Scholar

43 Jawaid A, Roszkowski M, Mansuy IM. Transgenerational epigenetics of traumatic stress. Prog Mol Biol Transl Sci. 2018;158:273–98.Google Scholar

44 Ben Maamar M, Beck D, Nilsson E, McCarrey JR, Skinner MK. Developmental origins of transgenerational sperm histone retention following ancestral exposures. Dev Biol. 2020;465(1):31–45. https://doi.org/10.1016/j.ydbio.2020.06.008Google Scholar

45 Ben Maamar M, King SE, Nilsson E, Beck D, Skinner MK. Epigenetic transgenerational inheritance of parent‐of‐origin allelic transmission of outcross pathology and sperm epimutations. Dev Biol. 2020;458(1):106–19. https://doi.org/10.1016/j.ydbio.2019.10.030Google Scholar

46 Ben Maamar M, Nilsson E, Thorson JLM, Beck D, Skinner MK. Transgenerational disease specific epigenetic sperm biomarkers after ancestral exposure to dioxin. Environ Res. 2021;192:110279. https://doi.org/10.1016/j.envres.2020.110279Google Scholar

47 Ben Maamar M, Sadler‐Riggleman I, Beck D, McBirney M, Nilsson E, Klukovich R, et al. Alterations in sperm DNA methylation, non‐coding RNA expression, and histone retention mediate vinclozolin‐induced epigenetic transgenerational inheritance of disease. Environ Epigen. 2018;4(2):dvy010. https://doi.org/10.1093/eep/dvy010Google Scholar

48 Chan JC, Nugent BM, Bale TL. Parental advisory: maternal and paternal stress can impact offspring neurodevelopment. Biol Psychiatry. 2018;83(10):886–94. https://doi.org/10.1016/j.biopsych.2017.10.005Google Scholar

49 Morgan CP, Chan JC, Bale TL. Driving the next generation: paternal lifetime experiences transmitted via extracellular vesicles and their small RNA Cargo. Biol Psychiatry. 2019;85(2):164–71. https://doi.org/10.1016/j.biopsych.2018.09.007Google Scholar

50 Rodgers AB, Bale TL. Germ cell origins of posttraumatic stress disorder risk: the transgenerational impact of parental stress experience. Biol Psychiatry. 2015;78(5):307–14. https://doi.org/10.1016/j.biopsych.2015.03.018Google Scholar

51 Rodgers AB, Morgan CP, Bronson SL, Revello S, Bale TL. Paternal stress exposure alters sperm microRNA content and reprograms offspring HPA stress axis regulation. J Neurosci. 2013;33(21):9003–12. https://doi.org/10.1523/jneurosci.0914‐13.2013Google Scholar

52 Rodgers AB, Morgan CP, Leu NA, Bale TL. Transgenerational epigenetic programming via sperm microRNA recapitulates effects of paternal stress. Proc Natl Acad Sci U S A. 2015;112(44):13699–704. https://doi.org/10.1073/pnas.1508347112Google Scholar

53 Skinner MK, Ben Maamar M, Sadler‐Riggleman I, Beck D, Nilsson E, McBirney M, et al. Alterations in sperm DNA methylation, non‐coding RNA and histone retention associate with DDT‐induced epigenetic transgenerational inheritance of disease. Epigenet Chromatin. 2018;11(1):8. https://doi.org/10.1186/s13072‐018‐0178‐0Google Scholar

54 Skinner MK, Nilsson E, Sadler‐Riggleman I, Beck D, Ben Maamar M, McCarrey JR. Transgenerational sperm DNA methylation epimutation developmental origins following ancestral vinclozolin exposure. Epigenetics. 2019;14(7):721–39. https://doi.org/10.1080/15592294.2019.1614417Google Scholar

55 Thorson JLM, Beck D, Ben Maamar M, Nilsson EE, McBirney M, Skinner MK. Epigenome‐wide association study for atrazine induced transgenerational DNA methylation and histone retention sperm epigenetic biomarkers for disease. PLoS One. 2020;15(12):e0239380. https://doi.org/10.1371/journal.pone.0239380Google Scholar

56 Nilsson EE, Sadler‐Riggleman I, Skinner MK. Environmentally induced epigenetic transgenerational inheritance of disease. Environ Epigenet. 2018;4(2):dvy016. https://doi.org/10.1093/eep/dvy016. eCollection 2018 AprGoogle Scholar

57 Beck D, Ben Maamar M, Skinner MK. Integration of sperm ncRNA‐directed DNA methylation and DNA methylation‐directed histone retention in epigenetic transgenerational inheritance. Epigenet Chromatin. 2021;14(1):6. https://doi.org/10.1186/s13072‐020‐00378‐0Google Scholar

58 Manikkam M, Haque MM, Guerrero‐Bosagna C, Nilsson EE, Skinner MK. Pesticide methoxychlor promotes the epigenetic transgenerational inheritance of adult‐onset disease through the female germline. PLoS One. 2014;9(7):e102091. https://doi.org/10.1371/journal.pone.0102091Google Scholar

59 Chan JC, Morgan CP, Adrian Leu N, Shetty A, Cisse YM, Nugent BM, et al. Reproductive tract extracellular vesicles are sufficient to transmit intergenerational stress and program neurodevelopment. Nat Commun. 2020;11(1):1499. https://doi.org/10.1038/s41467‐020‐15305‐wGoogle Scholar

60 Alshanbayeva A, Tanwar DK, Roszkowski M, Manuella F, Mansuy IM. Early life stress affects the miRNA cargo in epididymal extracellular vesicles in mouse. bioRxiv. 2021;105(3):593–602. https://doi.org/10.1093/biolre/ioab156Google Scholar

61 Rompala GR, Ferguson C, Homanics GE. Coincubation of sperm with epididymal extracellular vesicle preparations from chronic intermittent ethanol‐treated mice is sufficient to impart anxiety‐like and ethanol‐induced behaviors to adult progeny. Alcohol. 2020;87:111–20. https://doi.org/10.1016/j.alcohol.2020.05.001Google Scholar

62 Klengel T, Dias BG, Ressler KJ. Models of intergenerational and transgenerational transmission of risk for psychopathology in mice. Neuropsychopharmacology. 2016;41(1):219–31. https://doi.org/10.1038/npp.2015.249. Epub 2015 Aug 1018Google Scholar

63 McCreary JK, Erickson ZT, Metz GA. Environmental enrichment mitigates the impact of ancestral stress on motor skill and corticospinal tract plasticity. Neurosci Lett. 2016;632:181–6. https://doi.org/10.1016/j.neulet.2016.08.059Google Scholar

64 Dias BG, Ressler KJ. Parental olfactory experience influences behavior and neural structure in subsequent generations. Nat Neurosci. 2014;17(1):89–96. https://doi.org/10.1038/nn.3594Google Scholar

65 Gapp K, Jawaid A, Sarkies P, Bohacek J, Pelczar P, Prados J, et al. Implication of sperm RNAs in transgenerational inheritance of the effects of early trauma in mice. Nat Neurosci. 2014;17(5):667–9. https://doi.org/10.1038/nn.3695Google Scholar

66 Yao Y, Robinson AM, Zucchi FC, Robbins JC, Babenko O, Kovalchuk O, et al. Ancestral exposure to stress epigenetically programs preterm birth risk and adverse maternal and newborn outcomes. BMC Med. 2014;12(1):121. https://doi.org/10.1186/s12916‐014‐0121‐6Google Scholar

67 Chen Q, Yan M, Cao Z, Li X, Zhang Y, Shi J, et al. Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder. Science. 2016;351(6271):397–400. https://doi.org/10.1126/science.aad7977Google Scholar

68 de Castro Barbosa T, Ingerslev LR, Alm PS, Versteyhe S, Massart J, Rasmussen M, et al. High‐fat diet reprograms the epigenome of rat spermatozoa and transgenerationally affects metabolism of the offspring. Mol Metab. 2016;5(3):184–97. https://doi.org/10.1016/j.molmet.2015.12.002Google Scholar

69 Grandjean V, Fourré S, De Abreu DA, Derieppe MA, Remy JJ, Rassoulzadegan M. RNA‐mediated paternal heredity of diet‐induced obesity and metabolic disorders. Sci Rep. 2015;5(1):18193. https://doi.org/10.1038/srep18193Google Scholar

70 Ben Maamar M, Nilsson E, Sadler‐Riggleman I, Beck D, McCarrey JR, Skinner MK. Developmental origins of transgenerational sperm DNA methylation epimutations following ancestral DDT exposure. Dev Biol. 2019;445(2):280–93. https://doi.org/10.1016/j.ydbio.2018.11.016Google Scholar

71 Manikkam M, Tracey R, Guerrero‐Bosagna C, Skinner MK. Plastics derived endocrine disruptors (BPA, DEHP and DBP) induce epigenetic transgenerational inheritance of obesity, reproductive disease and sperm epimutations. PLoS One. 2013;8(1):e55387. https://doi.org/10.1371/journal.pone.0055387Google Scholar

72 Skinner MK, Manikkam M, Tracey R, Guerrero‐Bosagna C, Haque M, Nilsson EE. Ancestral dichlorodiphenyltrichloroethane (DDT) exposure promotes epigenetic transgenerational inheritance of obesity. BMC Med. 2013;11(1):228. https://doi.org/10.1186/1741‐7015‐11‐228Google Scholar

73 Anway MD, Cupp AS, Uzumcu M, Skinner MK. Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science. 2005;308(5727):1466–9. https://doi.org/10.1126/science.1108190Google Scholar

74 Thurston H, Fields BE, White J. Does increasing access to prenatal care reduce racial disparities in birth outcomes? J Pediatr Nurs. 2021;59:96–102. https://doi.org/10.1016/j.pedn.2021.01.012Google Scholar

75 Feng C, Osgood ND, Dyck RF. Low birth weight, cumulative obesity dose, and the risk of incident type 2 diabetes. J Diabetes Res. 2018;2018:8435762. https://doi.org/10.1155/2018/8435762Google Scholar

76 Jornayvaz FR, Vollenweider P, Bochud M, Mooser V, Waeber G, Marques‐Vidal P. Low birth weight leads to obesity, diabetes and increased leptin levels in adults: the CoLaus study. Cardiovasc Diabetol. 2016;15(1):73. https://doi.org/10.1186/s12933‐016‐0389‐2Google Scholar

77 Meyer DN, Crofts EJ, Akemann C, Gurdziel K, Farr R, Baker BB, et al. Developmental exposure to Pb(2+) induces transgenerational changes to zebrafish brain transcriptome. Chemosphere. 2020;244:125527. https://doi.org/10.1016/j.chemosphere.2019.125527Google Scholar

78 Jawaid A, Kunzi M, Mansoor M, et al. Distinct microRNA signature in human serum and germline after childhood trauma. medRxiv. under review.Google Scholar

79 Dickson DA, Paulus JK, Mensah V, Lem J, Saavedra‐Rodriguez L, Gentry A, et al. Reduced levels of miRNAs 449 and 34 in sperm of mice and men exposed to early life stress. Transl Psychiatry. 2018;8(1):101. https://doi.org/10.1038/s41398‐018‐0146‐2Google Scholar

80 Morgan CP, Shetty AC, Chan JC, Berger DS, Ament SA, Epperson CN, et al. Repeated sampling facilitates within‐ and between‐subject modeling of the human sperm transcriptome to identify dynamic and stress‐responsive sncRNAs. Sci Rep. 2020;10(1):17498. https://doi.org/10.1038/s41598‐020‐73867‐7Google Scholar

81 Aoued HS, Sannigrahi S, Doshi N, Morrison FG, Linsenbaum H, Hunter SC, et al. Reversing behavioral, neuroanatomical, and germline influences of intergenerational stress. Biol Psychiatry. 2019;85(3):248–56. https://doi.org/10.1016/j.biopsych.2018.07.028Google Scholar

82 Foa EB. Prolonged exposure therapy: past, present, and future. Depress Anxiety. 2011;28(12):1043–7. https://doi.org/10.1002/da.20907Google Scholar

83 Morland LA, Mackintosh MA, Greene CJ, Rosen CS, Chard KM, Resick P, et al. Cognitive processing therapy for posttraumatic stress disorder delivered to rural veterans via telemental health: a randomized noninferiority clinical trial. J Clin Psychiatry. 2014;75(5):470–6. https://doi.org/10.4088/JCP.13m08842Google Scholar

84 Cohen JA, Mannarino A. Trauma‐focused CBT for children and adolescents: treatment applications. New York: Guilford Press; 2012.Google Scholar

85 McPherson NO, Owens JA, Fullston T, Lane M. Preconception diet or exercise intervention in obese fathers normalizes sperm microRNA profile and metabolic syndrome in female offspring. Am J Physiol Endocrinol Metab. 2015;308(9):E805–821. https://doi.org/10.1152/ajpendo.00013.2015Google Scholar

86 Eleftheriou D, Benetou V, Trichopoulou A, La Vecchia C, Bamia C. Mediterranean diet and its components in relation to all‐cause mortality: meta‐analysis. Br J Nutr. 2018;120(10):1081–97. https://doi.org/10.1017/s0007114518002593Google Scholar

87 Li Y, Pan A, Wang DD, Liu X, Dhana K, Franco OH, et al. Impact of healthy lifestyle factors on life expectancies in the US population. Circulation. 2018;138(4):345–55. https://doi.org/10.1161/circulationaha.117.032047Google Scholar

88 Naghshi S, Sadeghi O, Willett WC, Esmaillzadeh A. Dietary intake of total, animal, and plant proteins and risk of all cause, cardiovascular, and cancer mortality: systematic review and dose‐response meta‐analysis of prospective cohort studies. BMJ. 2020;370:m2412. https://doi.org/10.1136/bmj.m2412Google Scholar

89 Alehagen U, Aaseth J, Alexander J, Johansson P. Still reduced cardiovascular mortality 12 years after supplementation with selenium and coenzyme Q10 for four years: a validation of previous 10‐year follow‐up results of a prospective randomized double‐blind placebo‐controlled trial in elderly. PLoS One. 2018;13(4):e0193120. https://doi.org/10.1371/journal.pone.0193120Google Scholar

90 Jenkins DJA, Kitts D, Giovannucci EL, Sahye‐Pudaruth S, Paquette M, Blanco Mejia S, et al. Selenium, antioxidants, cardiovascular disease, and all‐cause mortality: a systematic review and meta‐analysis of randomized controlled trials. Am J Clin Nutr. 2020;112(6):1642–52. https://doi.org/10.1093/ajcn/nqaa245Google Scholar

91 Zhang YB, Pan XF, Chen J, Cao A, Xia L, et al. Combined lifestyle factors, incident cancer, and cancer mortality: a systematic review and meta‐analysis of prospective cohort studies. Br J Cancer. 2020;122(7):1085–93. https://doi.org/10.1038/s41416‐020‐0741‐xGoogle Scholar

92 Jadhav RA, Hazari A, Monterio A, Kumar S, Maiya AG. Effect of physical activity intervention in prediabetes: a systematic review with meta‐analysis. J Phys Act Health. 2017;14(9):745–55. https://doi.org/10.1123/jpah.2016‐0632Google Scholar

93 Brooks K. Research shows food deserts more abundant in minority neighborhoods. Johns Hopkins Magazine; 2014. https://hub.jhu.edu/magazine/2014/spring/racial‐food‐deserts/Google Scholar

94 Cantor J, Beckman R, Collins RL, Dastidar MG, Richardson AS, Dubowitz T. SNAP participants improved food security and diet after a full‐service supermarket opened in an urban food desert. Health Aff (Millwood). 2020;39(8):1386–94. https://doi.org/10.1377/hlthaff.2019.01309Google Scholar

95 Freedman DA, Bell BA, Clark J, Ngendahimana D, Borawski E, Trapl E, et al. Small improvements in an urban food environment resulted in no changes in diet among residents. J Commun Health. 2021;46(1):1–12. https://doi.org/10.1007/s10900‐020‐00805‐zGoogle Scholar

96 Jilcott Pitts SB, Wu Q, Truesdale KP, Rafferty AP, Haynes‐Maslow L, Boys KA, et al. A four‐year observational study to examine the dietary impact of the North Carolina Healthy Food Small Retailer Program, 2017‐2020. Int J Behav Nutr Phys Act. 2021;18(1):44. https://doi.org/10.1186/s12966‐021‐01109‐8Google Scholar

97 Bustamante C, Henríquez R, Medina F, Reinoso C, Vargas R, Pascual R. Maternal exercise during pregnancy ameliorates the postnatal neuronal impairments induced by prenatal restraint stress in mice. Int J Dev Neurosci. 2013;31(4):267–73. https://doi.org/10.1016/j.ijdevneu.2013.02.007Google Scholar

98 Schander JA, Aisemberg J, Correa F, Wolfson ML, Juriol L, Cymeryng C, et al. The enrichment of maternal environment prevents pre‐term birth in a mice model. Reproduction. 2020;159(4):479–92. https://doi.org/10.1530/rep‐19‐0572Google Scholar

99 Gapp K, Bohacek J, Grossmann J, Brunner AM, Manuella F, Nanni P, et al. Potential of environmental enrichment to prevent transgenerational effects of paternal trauma. Neuropsychopharmacology. 2016;41(11):2749–58. https://doi.org/10.1038/npp.2016.87Google Scholar

100 Cao X, Huang S, Ruan D. Enriched environment restores impaired hippocampal long‐term potentiation and water maze performance induced by developmental lead exposure in rats. Dev Psychobiol. 2008;50(3):307–13. https://doi.org/10.1002/dev.20287Google Scholar

101 HCZ . Harlem Children’s Zone. https://hcz.org. Published 2021. Accessed 4 Jun 2021.Google Scholar

102 Slopen N, Williams DR. Resilience‐promoting policies and contexts for children of color in the United States: existing research and future priorities. Dev Psychopathol. 2021;33(2):614–24. https://doi.org/10.1017/s095457942000173xGoogle Scholar

103 Winkleby MA. The Stanford Medical Youth Science Program: 18 years of a biomedical program for low‐income high school students. Acad Med. 2007;82(2):139–45. https://doi.org/10.1097/acm.0b013e31802d8de6Google Scholar

104 Easterlin MC, Chung PJ, Leng M, Dudovitz R. Association of team sports participation with long‐term mental health outcomes among individuals exposed to adverse childhood experiences. JAMA Pediatr. 2019;173(7):681–8. https://doi.org/10.1001/jamapediatrics.2019.1212Google Scholar

105 Barton AW, Beach SRH, Bryant CM, Lavner JA, Brody GH. Stress spillover, African Americans' couple and health outcomes, and the stress‐buffering effect of family‐centered prevention. J Fam Psychol. 2018;32(2):186–96. https://doi.org/10.1037/fam0000376Google Scholar

106 Barton AW, Beach SRH, Wells AC, Ingels JB, Corso PS, Sperr MC, et al. The protecting strong African American families program: a randomized controlled trial with rural African American couples. Prev Sci. 2018;19(7):904–13. https://doi.org/10.1007/s11121‐018‐0895‐4Google Scholar

107 Brody GH, Chen YF, Kogan SM, Yu T, Molgaard VK, DiClemente RJ, et al. Family‐centered program deters substance use, conduct problems, and depressive symptoms in black adolescents. Pediatrics. 2012;129(1):108–15. Epub 2011 Dec 1512. https://doi.org/10.1542/peds.2011‐0623Google Scholar

108 Kogan SM, Yu T, Brody GH, Chen Yf, DiClemente RJ, Wingood GM, et al. Integrating condom skills into family‐centered prevention: efficacy of the Strong African American families‐teen program. J Adolesc Health. 2012;51(2):164–70. Epub 2012 Feb 1022. https://doi.org/10.1016/j.jadohealth.2011.11.022Google Scholar

109 Brody GH, Yu T, Chen E, Miller GE, Barton AW, Kogan SM. Family‐Centered prevention effects on the association between racial discrimination and mental health in black adolescents: secondary analysis of 2 randomized clinical trials. JAMA Netw Open. 2021;4(3):e211964. https://doi.org/10.1001/jamanetworkopen.2021.1964Google Scholar

110 Brody GH, Murry VM, Gerrard M, Gibbons FX, McNair L, Brown AC, et al. The strong African American families program: prevention of youths' high‐risk behavior and a test of a model of change. J Fam Psychol. 2006;20(1):1–11. https://doi.org/10.1037/0893‐3200.1020.1031.1031Google Scholar

111 Brody GH, Yu T, Chen E, Beach SR, Miller GE. Family‐centered prevention ameliorates the longitudinal association between risky family processes and epigenetic aging. J Child Psychol Psychiatry Allied Discip. 2015;57(5):12495–574. https://doi.org/10.1111/jcpp.12495Google Scholar

112 Brody GH. Strong African American families program. In: Kaufman J, editor. E‐mail communication; 2019.Google Scholar

113 Brody GH, Kogan SM, Chen YF, McBride Murry V. Long‐term effects of the strong African American families program on youths' conduct problems. J Adolesc Health. 2008;43(5):474–81. https://doi.org/10.1016/j.jadohealth.2008.1004.1016. Epub 2008 Jul 1031Google Scholar

114 Chen YF, Yu T, Brody GH. Parenting intervention at age 11 and cotinine levels at age 20 among African American youth. Pediatrics. 2017;140(1). Epub 2017 Jun 2014. https://doi.org/10.1542/peds.2016‐4162Google Scholar

115 Brody GH, Chen YF, Kogan SM, Murry VM, Brown AC. Long‐term effects of the strong African American families program on youths' alcohol use. J Consult Clin Psychol. 2010;78(2):281–5. https://doi.org/10.1037/a0018552Google Scholar

116 Brody GH, Yu T, Miller GE, Ehrlich KB, Chen E. Preventive parenting intervention during childhood and young black adults' unhealthful behaviors: a randomized controlled trial. J Child Psychol Psychiatry. 2019;60(1):63–71. https://doi.org/10.1111/jcpp.12968. Epub 12018 Sep 12911Google Scholar

117 Murry VM, Berkel C, Chen YF, Brody GH, Gibbons FX, Gerrard M. Intervention induced changes on parenting practices, youth self‐pride and sexual norms to reduce HIV‐related behaviors among rural African American youths. J Youth Adolesc. 2011;40(9):1147–63. Epub 12011 Mar 10965. https://doi.org/10.1007/s10964‐011‐9642‐xGoogle Scholar

118 Chen E, Miller GE, Yu T, Brody GH. Unsupportive parenting moderates the effects of family psychosocial intervention on metabolic syndrome in African American youth. Int J Obes (Lond). 2018;42(4):634–40. Epub 2017 Oct 1036. https://doi.org/10.1038/ijo.2017.246Google Scholar

119 Brody GH, Yu T, Chen E, Miller GE. Family‐centered prevention ameliorates the association between adverse childhood experiences and prediabetes status in young black adults. Prev Med. 2017;100:117–22. Epub 2017 Apr 1019. https://doi.org/10.1016/j.ypmed.2017.1004.1017Google Scholar

120 Brody GH, Yu T, Chen E, Miller GE. Prevention moderates associations between family risks and youth catecholamine levels. Health Psychol. 2014;33(11):1435–9. Epub 0002014 Mar 0000073. https://doi.org/10.1037/hea0000072Google Scholar

121 Miller GE, Brody GH, Yu T, Chen E. A family‐oriented psychosocial intervention reduces inflammation in low‐SES African American youth. Proc Natl Acad Sci U S A. 2014;111(31):11287–92. Epub 1406572014 Jul 1406578121. https://doi.org/10.1073/pnas.1406578111Google Scholar

122 Brody GH, Yu T, Chen E, Beach SR, Miller GE. Family‐centered prevention ameliorates the longitudinal association between risky family processes and epigenetic aging. J Child Psychol Psychiatry. 2016;57(5):566–74. https://doi.org/10.1111/jcpp.12495. Epub 12015 Dec 12417Google Scholar

123 Beach SRH, Lei MK, Brody GH, Philibert RA. Prevention of early substance use mediates, and variation at SLC6A4 moderates, SAAF intervention effects on OXTR methylation. Prev Sci. 2018;19(1):90–100. https://doi.org/10.1007/s11121‐016‐0709‐5Google Scholar

124 Brody GH, Gray JC, Yu T, Barton AW, Beach SRH, Galvan A, et al. Protective prevention effects on the association of poverty with brain development. JAMA Pediatr. 2017;171(1):46–52. https://doi.org/10.1001/jamapediatrics.2016.2988Google Scholar

125 Hall GC, Ibaraki AY, Huang ER, Marti CN, Stice E. A meta‐analysis of cultural adaptations of psychological interventions. Behav Ther. 2016;47(6):993–1014. https://doi.org/10.1016/j.beth.2016.09.005Google Scholar

126 Soto A, Smith TB, Griner D, Domenech Rodríguez M, Bernal G. Cultural adaptations and therapist multicultural competence: two meta‐analytic reviews. J Clin Psychol. 2018;74(11):1907–23. https://doi.org/10.1002/jclp.22679Google Scholar